Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
J Chromatogr A ; 1730: 465064, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-38865749

RESUMO

Small ubiquitin-like modifier (SUMO) modification regulates various eukaryotic cellular processes and plays a pivotal role in interferon (IFN)-mediated antiviral defense. While immunoprecipitation enrichment method is widely used for proteome-wide analysis of endogenous SUMOylation, the inability to target all SUMO forms and high cost of antibodies limited its further application. Herein, we proposed an antibody-free enrichment method based on SUMO-specific protease and strong anion exchange chromatography (SPAX) to globally profile the endogenous SUMOylation. The SUMO1/2/3-modified peptides could be simultaneously enriched by SAX chromatography by utilizing its electrostatic interaction with SUMO1/2/3 remnants, which contained multiple aspartic acids (D) and glutamic acids (E). To remove the co-enriched D/E-containing peptides which might interfere with the detection of low-abundance SUMOylated peptides, SUMO-specific protease was used to cleave the SUMO1/2/3 remnants from enriched SUMOylated peptides. As the deSUMOylated peptides lost SUMO remnants, their interaction with SAX materials became weaker, and the D/E-containing peptides could thus be depleted through the second SAX separation. The SPAX method identified over twice the SUMOylated sites than using SAX method only, greatly improving the identification coverage of endogenous SUMOylated sites. Our strategy was then applied to the site-specific identification and quantification of endogenous SUMOylation in A549 cells stimulated by IFN-γ for the first time. A total of 226 SUMOylated sites on 146 proteins were confidently identified, among which multiple up-regulated sites were involved in IFN-mediated antiviral defense, demonstrating the great promise of SPAX to globally profile and discover endogenous SUMOylation with significant biological functions.


Assuntos
Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina , Sumoilação , Humanos , Cromatografia por Troca Iônica/métodos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Proteína SUMO-1/metabolismo , Proteína SUMO-1/química , Peptídeos/química , Peptídeos/análise , Peptídeos/metabolismo
2.
World J Microbiol Biotechnol ; 40(6): 183, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722449

RESUMO

Heterologous production of proteins in Escherichia coli has raised several challenges including soluble production of target proteins, high levels of expression and purification. Fusion tags can serve as the important tools to overcome these challenges. SUMO (small ubiquitin-related modifier) is one of these tags whose fusion to native protein sequence can enhance its solubility and stability. In current research, a simple, efficient and cost-effective method is being discussed for the construction of pET28a-SUMO vector. In order to improve the stability and activity of lysophospholipase from Pyrococcus abyssi (Pa-LPL), a 6xHis-SUMO tag was fused to N-terminal of Pa-LPL by using pET28a-SUMO vector. Recombinant SUMO-fused enzyme (6 H-S-PaLPL) works optimally at 35 °C and pH 6.5 with remarkable thermostability at 35-95 °C. Thermo-inactivation kinetics of 6 H-S-PaLPL were also studied at 35-95 °C with first order rate constant (kIN) of 5.58 × 10- 2 h-1 and half-life of 12 ± 0 h at 95 °C. Km and Vmax for the hydrolysis of 4-nitrophenyl butyrate were calculated to be 2 ± 0.015 mM and 3882 ± 22.368 U/mg, respectively. 2.4-fold increase in Vmax of Pa-LPL was observed after fusion of 6xHis-SUMO tag to its N-terminal. It is the first report on the utilization of SUMO fusion tag to enhance the overall stability and activity of Pa-LPL. Fusion of 6xHis-SUMO tag not only aided in the purification process but also played a crucial role in increasing the thermostability and activity of the enzyme. SUMO-fused enzyme, thus generated, can serve as an important candidate for degumming of vegetable oils at industrial scale.


Assuntos
Estabilidade Enzimática , Escherichia coli , Pyrococcus abyssi , Proteínas Recombinantes de Fusão , Temperatura , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/química , Escherichia coli/genética , Escherichia coli/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Pyrococcus abyssi/genética , Pyrococcus abyssi/enzimologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Vetores Genéticos/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Proteína SUMO-1/química , Clonagem Molecular , Solubilidade
3.
J Phys Chem Lett ; 14(40): 9060-9068, 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37782899

RESUMO

Liquid-liquid phase separation (LLPS) plays a crucial role in cellular organization, primarily driven by intrinsically disordered proteins (IDPs) leading to the formation of biomolecular condensates. A folded protein SUMO that post-translationally modifies cellular proteins has recently emerged as a regulator of LLPS. Given its compact structure and limited flexibility, the precise role of SUMO in condensate formation remains to be investigated. Here, we show the rapid phase separation of SUMO1 into micrometer-sized liquid-like condensates in inert crowders under physiological conditions. Subsequent time-dependent conformational changes and aggregation are probed by label-free methods (tryptophan fluorescence and Raman spectroscopy). Remarkably, experiments on a SUMO1 variant lacking the N-terminal disordered region further corroborate the role of its structured part in phase transitions. Our findings highlight the potential of folded proteins to engage in LLPS and emphasize further investigation into the influence of the SUMO tag on IDPs associated with membrane-less assemblies in cells.


Assuntos
Proteínas Intrinsicamente Desordenadas , Proteína SUMO-1 , Proteínas Intrinsicamente Desordenadas/química , Triptofano , Ubiquitinas , Proteína SUMO-1/química
4.
Bioorg Med Chem Lett ; 94: 129460, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37640164

RESUMO

The transiently-activated SUMO probes are conducive to understand the dynamic control of SENPs activity. Here, we developed a photocaged glycine-assisted strategy for the construction of on demand-activated SUMO-ABPs. The light-sensitive groups installed at G92 and G64 backbone of SUMO-2 can temporarily block probes activity and hamper aspartimide formation, respectively, which enabled the efficient synthesis of inert SUMO-2 propargylamide (PA). The probe could be activated to capture SENPs upon photo-irradiation not only in vitro but also in intact cells, providing opportunities to further perform intracellular time-resolved proteome-wide profiling of SUMO-related enzymes.


Assuntos
Sondas Moleculares , Proteína SUMO-1 , Glicina/química , Piruvatos , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Fotoquímica/métodos
5.
Plant Cell Rep ; 42(8): 1279-1290, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37178216

RESUMO

KEY MESSAGE: SMC5/6 complex subunit OsMMS21 is involved in cell cycle and hormone signaling and required for stem cell proliferation during shoot and root development in rice. The structural maintenance of chromosome (SMC)5/6 complex is required for nucleolar integrity and DNA metabolism. Moreover, METHYL METHANESULFONATE SENSITIVITY GENE 21 (MMS21), a SUMO E3 ligase that is part of the SMC5/6 complex, is essential for the root stem cell niche and cell cycle transition in Arabidopsis. However, its specific role in rice remains unclear. Here, OsSMC5 and OsSMC6 single heterozygous mutants were generated using CRISPR/Cas9 technology to elucidate the function of SMC5/6 subunits, including OsSMC5, OsSMC6, and OsMMS21, in cell proliferation in rice. ossmc5/ + and ossmc6/ + heterozygous single mutants did not yield homozygous mutants in their progeny, indicating that OsSMC5 and OsSMC6 both play necessary roles during embryo formation. Loss of OsMMS21 caused severe defects in both the shoot and roots in rice. Transcriptome analysis showed a significant decrease in the expression of genes involved in auxin signaling in the roots of osmms21 mutants. Moreover, the expression levels of the cycB2-1 and MCM genes, which are involved the cell cycle, were significantly lower in the shoots of the mutants, indicating that OsMMS21 was involved in both hormone signaling pathways and the cell cycle. Overall, these findings indicate that the SUMO E3 ligase OsMMS21 is required for both shoot and root stem cell niches, improving the understanding of the function of the SMC5/6 complex in rice.


Assuntos
Oryza , Proteínas de Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Oryza/genética , Oryza/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Proteína SUMO-1/química , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Ubiquitina-Proteína Ligases/genética , Divisão Celular , Hormônios
6.
J Biomol Struct Dyn ; 41(21): 12372-12386, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36656084

RESUMO

SUMOylation regulates various cellular process and SENP1 (SUMO-specific protease 1) serves as a SUMO (small ubiquitin-related modifier) specific protease that participates in the SUMO cycle. Given its extensive influences on metabolic activities, SENP1 has gained more and more attentions in clinical treatments. However, there remains a question on why does the SENP1 prefer to process SUMO1 rather than SUMO2. Here, we performed molecular dynamics simulations of SENP1-SUMO1, SENP1-SUMO2, and apo SENP1 systems and observed distinct conformational dynamics in the upper half of the clamp and the three loops in the catalytic center of the SENP1. Principal component analysis revealed that the most prominent canonical variable represented the spatial distribution of the upper half of the clamp, while the openness of the cleft was closely related to the catalytic ability of SENP1. Further analysis of the SENP1-SUMO interactions revealed that the extensive and strong interactions between the SENP1 and SUMO1 were both in the interface of the upper half region and the catalytic center. Dynamic cross-correlation matrix analysis demonstrated that the inter-residue correlations in the SUMO1 system was much stronger, especially in the two essential regions belonging to the upper and lower half of cleft. Based on these observations, we proposed an allosteric propagation model and further testified it using the community analysis. These results revealed the propagation pathway of allosteric communication that contributed to the substrate discrimination of SENP1 upon SUMO1 and SUMO2.Communicated by Ramaswamy H. Sarma.


Assuntos
Cisteína Endopeptidases , Proteína SUMO-1 , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina , Simulação de Dinâmica Molecular , Ubiquitina , Cisteína Endopeptidases/química , Proteína SUMO-1/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química
7.
Protein Expr Purif ; 191: 106025, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34826607

RESUMO

The detection of antibody to non-structural protein (NSP) of Foot-and-mouth disease virus (FMDV) is the reliable diagnostic method for differentiating infected from vaccinated animals (DIVA). For this purpose, the detection of antibodies to non-structural 3ABC protein is suitable for identification of virus activity in the animals exposed to FMDV infection. However, large-scale production of recombinant 3ABC protein is challenging due to the formation of inclusion bodies in Escherichia coli and low yield due to protein aggregation during in vitro refolding. In this study, 3ABC gene was fused with SUMO (small ubiquitin-like modifiers) fusion system which significantly enhanced expression of recombinant 3ABC protein in E. coli. The solubility of the recombinant 6xHis-SUMO 3ABC fusion protein was improved by mild detergent treatment and purified through Ni-NTA chromatography under non-denaturing conditions which yielded 9 mg protein obtained from 1-L bacterial fermentation culture. The diagnostic potential of recombinant 3ABC protein was also tested by ELISA that provided reliable diagnostic performance (DSn = 92%, DSp = 94%) upon comparison with commercially available kit. The thermal stability of fusion protein was also tested which presented reliable performance at different temperatures. In conclusion, we presented SUMO fusion for the enhanced expression in E. coli and purification of active recombinant 3ABC protein using non-denaturing conditions without refolding steps. This protein can be used as a suitable diagnostic antigen to detect antibodies following FMDV infection.


Assuntos
Vírus da Febre Aftosa/genética , Expressão Gênica , Proteínas Recombinantes de Fusão , Proteína SUMO-1 , Proteínas não Estruturais Virais , Vírus da Febre Aftosa/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteína SUMO-1/biossíntese , Proteína SUMO-1/química , Proteína SUMO-1/genética , Proteína SUMO-1/isolamento & purificação , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/isolamento & purificação
8.
J Biol Chem ; 297(2): 100970, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34274315

RESUMO

Protein structural bioinformatic analyses suggest preferential associations between methionine and aromatic amino acid residues in proteins. Ab initio energy calculations highlight a conformation-dependent stabilizing interaction between the interacting sulfur-aromatic molecular pair. However, the relevance of buried methionine-aromatic motifs to protein folding and function is relatively unexplored. The Small Ubiquitin-Like Modifier (SUMO) is a ß-grasp fold protein and a common posttranslational modifier that affects diverse cellular processes, including transcriptional regulation, chromatin remodeling, metabolic regulation, mitosis, and meiosis. SUMO is a member of the Ubiquitin-Like (UBL) protein family. Herein, we report that a highly conserved and buried methionine-phenylalanine motif is a unique signature of SUMO proteins but absent in other homologous UBL proteins. We also detect that a specific "up" conformation between the methionine-phenylalanine pair of interacting residues in SUMO is critical to its ß-grasp fold. The noncovalent interactions of SUMO with its ligands are dependent on the methionine-phenylalanine pair. MD simulations, NMR, and biophysical and biochemical studies suggest that perturbation of the methionine-aromatic motif disrupts native contacts, modulates noncovalent interactions, and attenuates SUMOylation activity. Our results highlight the importance of conserved orientations of Met-aromatic structural motifs inside a protein core for its structure and function.


Assuntos
Metionina/química , Simulação de Dinâmica Molecular , Fenilalanina/química , Domínios e Motivos de Interação entre Proteínas , Proteína SUMO-1/química , Sumoilação , Motivos de Aminoácidos , Sequência de Aminoácidos , Humanos , Dobramento de Proteína , Estabilidade Proteica , Proteína SUMO-1/metabolismo , Relação Estrutura-Atividade
9.
Elife ; 102021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34110283

RESUMO

The nuclear pore complex (NPC) is the sole and selective gateway for nuclear transport, and its dysfunction has been associated with many diseases. The metazoan NPC subcomplex RanBP2, which consists of RanBP2 (Nup358), RanGAP1-SUMO1, and Ubc9, regulates the assembly and function of the NPC. The roles of immune signaling in regulation of NPC remain poorly understood. Here, we show that in human and murine T cells, following T-cell receptor (TCR) stimulation, protein kinase C-θ (PKC-θ) directly phosphorylates RanGAP1 to facilitate RanBP2 subcomplex assembly and nuclear import and, thus, the nuclear translocation of AP-1 transcription factor. Mechanistically, TCR stimulation induces the translocation of activated PKC-θ to the NPC, where it interacts with and phosphorylates RanGAP1 on Ser504 and Ser506. RanGAP1 phosphorylation increases its binding affinity for Ubc9, thereby promoting sumoylation of RanGAP1 and, finally, assembly of the RanBP2 subcomplex. Our findings reveal an unexpected role of PKC-θ as a direct regulator of nuclear import and uncover a phosphorylation-dependent sumoylation of RanGAP1, delineating a novel link between TCR signaling and assembly of the RanBP2 NPC subcomplex.


Assuntos
Proteínas Ativadoras de GTPase , Chaperonas Moleculares , Complexo de Proteínas Formadoras de Poros Nucleares , Receptores de Antígenos de Linfócitos T/metabolismo , Proteína SUMO-1 , Enzimas de Conjugação de Ubiquitina , Animais , Proteínas Ativadoras de GTPase/química , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Camundongos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Poro Nuclear/química , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Fosforilação , Proteína Quinase C-theta/química , Proteína Quinase C-theta/metabolismo , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Transdução de Sinais/fisiologia , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo
10.
PLoS One ; 16(2): e0247132, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33600463

RESUMO

Protein sumoylation, especially when catalyzed by the Mms21 SUMO E3 ligase, plays a major role in suppressing duplication-mediated gross chromosomal rearrangements (dGCRs). How Mms21 targets its substrates in the cell is insufficiently understood. Here, we demonstrate that Esc2, a protein with SUMO-like domains (SLDs), recruits the Ubc9 SUMO conjugating enzyme to specifically facilitate Mms21-dependent sumoylation and suppress dGCRs. The D430R mutation in Esc2 impairs its binding to Ubc9 and causes a synergistic growth defect and accumulation of dGCRs with mutations that delete the Siz1 and Siz2 E3 ligases. By contrast, esc2-D430R does not appreciably affect sensitivity to DNA damage or the dGCRs caused by the catalytically inactive mms21-CH. Moreover, proteome-wide analysis of intracellular sumoylation demonstrates that esc2-D430R specifically down-regulates sumoylation levels of Mms21-preferred targets, including the nucleolar proteins, components of the SMC complexes and the MCM complex that acts as the catalytic core of the replicative DNA helicase. These effects closely resemble those caused by mms21-CH, and are relatively unaffected by deleting Siz1 and Siz2. Thus, by recruiting Ubc9, Esc2 facilitates Mms21-dependent sumoylation to suppress the accumulation of dGCRs independent of Siz1 and Siz2.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteína SUMO-1/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Dano ao DNA , Replicação do DNA , Regulação para Baixo , Mutagênese , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , Proteômica , Proteína SUMO-1/química , Proteína SUMO-1/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alinhamento de Sequência , Sumoilação , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
11.
Cell Chem Biol ; 28(2): 180-190.e6, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33444530

RESUMO

The accumulation of α-synuclein amyloid fibrils in the brain is linked to Parkinson's disease and other synucleinopathies. The intermediate species in the early aggregation phase of α-synuclein are involved in the emergence of amyloid toxicity and considered to be the most neurotoxic. The N-terminal region flanking the non-amyloid-ß component domain of α-synuclein has been implicated in modulating its aggregation. Herein, we report the development of a SUMO1-derived peptide inhibitor (SUMO1(15-55)), which targets two SUMO-interacting motifs (SIMs) within this aggregation-regulating region and suppresses α-synuclein aggregation. Molecular modeling, site-directed mutagenesis, and binding studies are used to elucidate the mode of interaction, namely, via the binding of either of the two SIM sequences on α-synuclein to a putative hydrophobic binding groove on SUMO1(15-55). Subsequent studies show that SUMO1(15-55) also reduces α-synuclein-induced cytotoxicity in cell-based and Drosophila disease models.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Proteína SUMO-1/química , Proteína SUMO-1/farmacologia , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Drosophila , Descoberta de Drogas , Humanos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Peptídeos/metabolismo , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/metabolismo , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteína SUMO-1/metabolismo
12.
Structure ; 28(11): 1225-1230.e3, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-32726574

RESUMO

Human ubiquitin ligase HERC2, a component of the DNA repair machinery, has been linked to neurological diseases and cancer. Here, we show that the ZZ domain of HERC2 (HERC2ZZ) binds to histone H3 tail and tolerates posttranslational modifications commonly present in H3. The crystal structure of the HERC2ZZ:H3 complex provides the molecular basis for this interaction and highlights a critical role of the negatively charged site of HERC2ZZ in capturing of A1 of H3. NMR, mutagenesis, and fluorescence data reveal that HERC2ZZ binds to H3 and the N-terminal tail of SUMO1, a previously reported ligand of HERC2ZZ, with comparable affinities. Like H3, the N-terminal tail of SUMO1 occupies the same negatively charged site of HERC2ZZ in the crystal structure of the complex, although in contrast to H3 it adopts an α-helical conformation. Our data suggest that HERC2ZZ may play a role in mediating the association of HERC2 with chromatin.


Assuntos
Cromatina/química , Histonas/química , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/química , Ubiquitina-Proteína Ligases/química , Sítios de Ligação , Cromatina/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Cinética , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Eletricidade Estática , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
13.
Biochem Biophys Res Commun ; 524(3): 677-682, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32033752

RESUMO

Proteins can stabilize upon binding a ligand. Due to allosteric effects, the changes in stability can occur at regions far from the protein:ligand interface. Efficient methods to measure the changes in local stability upon ligand binding will be useful to understand allostery and may be helpful in protein engineering. In this work, we suggest the measurement of backbone amide temperature coefficients to probe the effect of ligand binding on the local stability of ß-sheet rich proteins. The method was applied for two protein:ligand complexes with different binding affinities. The protein includes a beta-sheet network connected by hydrogen bonds. The measured temperature coefficient data captured the stabilizing effect of ligand binding, which propagated across the beta-sheet network of the protein. Intriguingly, the impact on the local and global stability of the protein was proportional to the strength of protein:ligand interaction.


Assuntos
Amidas/química , Temperatura , Regulação Alostérica , Motivos de Aminoácidos , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Ligação Proteica , Estabilidade Proteica , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo
14.
Int J Biol Macromol ; 151: 204-211, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32061854

RESUMO

Metal ions like Cu2+ and Zn2+ have been shown to impact protein misfolding pathways in neurodegenerative proteinopathies like Alzheimer's and Parkinson's. Also, due to their strong interaction with Ubiquitin, they interfere in degradation of misfolded proteins by impairing the ubiquitin-proteasome system (UPS). In this work, we have studied the interaction of these metal ions with a small Ubiquitin like post-translation modifier SUMO1, which is known to work co-operatively with Ubiquitin to regulate UPS system. Between Cu2+ and Zn2+, the former binds more strongly with SUMO1 as determined using fluorescence spectroscopy. SUMO1 aggregates, forming trimer and higher oligomers in presence of Cu2+ ions which were characterized using gel electrophoresis, Bradford assay, and transmission electron microscopy. Chemical shift analysis using 15N/1H based NMR spectroscopy revealed that SUMO1 retains its structural fold in its trimeric state. Cu2+ induced paramagnetic quenching and Zn2+ induced chemical shift perturbation of 15N-1H cross-peaks were used to identify their respective binding sites in SUMO1. Binding sites so obtained were further validated with molecular dynamics studies. Our findings provide structural insights into the SUMO1-Cu2+/Zn2+ interaction, and its impact on aggregation of SUMO1 which might affect its ability to modify functions of target proteins.


Assuntos
Sítios de Ligação , Cobre/química , Íons , Proteína SUMO-1/química , Zinco/química , Sequência de Aminoácidos , Simulação de Dinâmica Molecular , Ligação Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes , Proteína SUMO-1/metabolismo , Análise Espectral , Relação Estrutura-Atividade
15.
ACS Synth Biol ; 9(2): 427-436, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-31977192

RESUMO

Protein-protein interactions control a wide variety of natural biological processes. α-Helical coiled coils frequently mediate such protein-protein interactions. Due to the relative simplicity of their sequences and structures and the ease with which properties such as strength and specificity of interaction can be controlled, coiled coils can be designed de novo to deliver a variety of non-natural protein-protein interaction domains. Herein, several de novo designed coiled coils are tested for their ability to mediate protein-protein interactions in Escherichia coli cells. The set includes a parallel homodimer, a parallel homotetramer, an antiparallel homotetramer, and a newly designed heterotetramer, all of which have been characterized in vitro by biophysical and structural methods. Using a transcription repression assay based on reconstituting the Lac repressor, we find that the modules behave as designed in the cellular environment. Each design imparts a different property to the resulting Lac repressor-coiled coil complexes, resulting in the benefit of being able to reconfigure the system in multiple ways. Modification of the system also allows the interactions to be controlled: assembly can be tuned by controlling the expression of the constituent components, and complexes can be disrupted through helix sequestration. The small and straightforward de novo designed components that we deliver are highly versatile and have considerable potential as protein-protein interaction domains in synthetic biology where proteins must be assembled in highly specific ways. The relative simplicity of the designs makes them amenable to future modifications to introduce finer control over their assembly and to adapt them for different contexts.


Assuntos
Proteínas/metabolismo , Sequência de Aminoácidos , Dicroísmo Circular , Escherichia coli/metabolismo , Óperon Lac/genética , Plasmídeos/genética , Plasmídeos/metabolismo , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas/química , Proteínas/genética , Proteína SUMO-1/química , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Transcrição Gênica
16.
Chemistry ; 26(22): 4952-4957, 2020 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-31960982

RESUMO

Despite their biological importance, post-translationally modified proteins are notoriously difficult to produce in a homogeneous fashion by using conventional expression systems. Chemical protein synthesis or semisynthesis offers a solution to this problem; however, traditional strategies often rely on sulfur-based chemistry that is incompatible with the presence of any cysteine residues in the target protein. To overcome these limitations, we present the design and synthesis of γ-selenolysine, a selenol-containing form of the commonly modified proteinogenic amino acid, lysine. The utility of γ-selenolysine is demonstrated with the traceless ligation of the small ubiquitin-like modifier protein, SUMO-1, to a peptide segment of human glucokinase. The resulting polypeptide is poised for native chemical ligation and chemoselective deselenization in the presence of unprotected cysteine residues. Selenolysine's straightforward synthesis and incorporation into synthetic peptides marks it as a universal handle for conjugating any ubiquitin-like modifying protein to its target.


Assuntos
Cisteína/química , Lisina/química , Peptídeos/química , Proteína SUMO-1/química , Compostos de Selênio/química , Aminoácidos , Humanos , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/metabolismo , Enxofre/química
17.
Structure ; 28(2): 157-168.e5, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31879127

RESUMO

The interactions between SUMO proteins and SUMO-interacting motif (SIM) in nuclear bodies formed by the promyelocytic leukemia (PML) protein (PML-NBs) have been shown to be modulated by either phosphorylation of the SIMs or acetylation of SUMO proteins. However, little is known about how this occurs at the atomic level. In this work, we examined the role that acetylation of SUMO1 plays on its binding to the phosphorylated SIMs (phosphoSIMs) of PML and Daxx. Our results demonstrate that SUMO1 binding to the phosphoSIM of either PML or Daxx is dramatically reduced by acetylation at either K39 or K46. However, acetylation at K37 only impacts binding to Daxx. Structures of acetylated SUMO1 variants bound to the phosphoSIMs of PML and Daxx demonstrate that there is structural plasticity in SUMO-SIM interactions. The plasticity observed in these structures provides a robust mechanism for regulating SUMO-SIM interactions in PML-NBs using signaling generated post-translational modifications.


Assuntos
Proteínas Correpressoras/química , Proteínas Correpressoras/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteína da Leucemia Promielocítica/química , Proteína da Leucemia Promielocítica/metabolismo , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Acetilação , Sítios de Ligação , Cristalografia por Raios X , Células HEK293 , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutação , Fosforilação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Dobramento de Proteína , Proteína SUMO-1/genética
18.
Proteins ; 88(3): 449-461, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31587348

RESUMO

Small ubiquitin-related modifiers (SUMO1 and SUMO2) are ubiquitin family proteins, structurally similar to ubiquitin, differing in terms of their amino acid sequence and functions. Therefore, they provide a great platform for investigating sequence-structure-stability-function relationship. Here, we used chemical denaturation in comparing the folding-unfolding pathways of the SUMO proteins with their structural homologue ubiquitin (UF45W-pseudo wild-type [WT] tryptophan variant) with structurally analogous tryptophan mutations (SUMO1 [S1F66W], SUMO2 [S2F62W]). Equilibrium denaturation studies report that ubiquitin is the most stable protein among the three. The observed denaturant-dependent folding rates of SUMOs are much lower than ubiquitin and primarily exhibit a two-state folding pathway unlike ubiquitin, which has a kinetic folding intermediate. We hypothesize that, as SUMO proteins start off as slow folders, they avoid stabilizing their folding intermediates and the presence of which might further slow-down their folding rates. The denaturant-dependent unfolding of ubiquitin is the fastest, followed by SUMO2, and slowest for SUMO1. However, the spontaneous unfolding rate constant is the lowest for ubiquitin (~40 times), and similar for SUMOs. This correlation between thermodynamic stability and kinetic stability is achieved by having different unfolding transition state positions with respect to the solvent-accessible surface area, as quantified by the Tanford ß u values: ubiquitin (0.42) > SUMO2 (0.20) > SUMO1 (0.16). The results presented here highlight the unique energy landscape features which help in optimizing the folding-unfolding rates within a structurally homologous protein family.


Assuntos
Proteína SUMO-1/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Ubiquitina/química , Sequência de Aminoácidos , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Guanidina/química , Humanos , Cinética , Modelos Moleculares , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Desnaturação Proteica , Dobramento de Proteína , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Termodinâmica , Ubiquitina/genética , Ubiquitina/metabolismo
19.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597768

RESUMO

Some viruses take advantage of conjugation of ubiquitin or ubiquitin-like proteins to enhance their own replication. One example is Ebola virus, which has evolved strategies to utilize these modification pathways to regulate the viral proteins VP40 and VP35 and to counteract the host defenses. Here, we show a novel mechanism by which Ebola virus exploits the ubiquitin and SUMO pathways. Our data reveal that minor matrix protein VP24 of Ebola virus is a bona fide SUMO target. Analysis of a SUMOylation-defective VP24 mutant revealed a reduced ability to block the type I interferon (IFN) pathway and to inhibit IFN-mediated STAT1 nuclear translocation, exhibiting a weaker interaction with karyopherin 5 and significantly diminished stability. Using glutathione S-transferase (GST) pulldown assay, we found that VP24 also interacts with SUMO in a noncovalent manner through a SIM domain. Mutation of the SIM domain in VP24 resulted in a complete inability of the protein to downmodulate the IFN pathway and in the monoubiquitination of the protein. We identified SUMO deubiquitinating enzyme ubiquitin-specific-processing protease 7 (USP7) as an interactor and a negative modulator of VP24 ubiquitination. Finally, we show that mutation of one ubiquitination site in VP24 potentiates the IFN modulatory activity of the viral protein and its ability to block IFN-mediated STAT1 nuclear translocation, pointing to the ubiquitination of VP24 as a negative modulator of the VP24 activity. Altogether, these results indicate that SUMO interacts with VP24 and promotes its USP7-mediated deubiquitination, playing a key role in the interference with the innate immune response mediated by the viral protein.IMPORTANCE The Ebola virus VP24 protein plays a critical role in escape of the virus from the host innate immune response. Therefore, deciphering the molecular mechanisms modulating VP24 activity may be useful to identify potential targets amenable to therapeutics. Here, we identify the cellular proteins USP7, SUMO, and ubiquitin as novel interactors and regulators of VP24. These interactions may represent novel potential targets to design new antivirals with the ability to modulate Ebola virus replication.


Assuntos
Ebolavirus/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Proteína SUMO-1/química , Peptidase 7 Específica de Ubiquitina/genética , Proteínas Virais/química , Animais , Sítios de Ligação , Chlorocebus aethiops , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Transporte Proteico , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Proteína SUMO-1/genética , Proteína SUMO-1/imunologia , Transdução de Sinais , Sumoilação , Peptidase 7 Específica de Ubiquitina/imunologia , Células Vero , Proteínas Virais/genética , Proteínas Virais/imunologia , alfa Carioferinas/genética , alfa Carioferinas/imunologia
20.
J Biol Chem ; 294(40): 14546-14561, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31371453

RESUMO

Many viral factors manipulate the host post-translational modification (PTM) machinery for efficient viral replication. In particular, phosphorylation and SUMOylation can distinctly regulate the activity of the human cytomegalovirus (HCMV) transactivator immediate early 2 (IE2). However, the molecular mechanism of this process is unknown. Using various structural, biochemical, and cell-based approaches, here we uncovered that IE2 exploits a cross-talk between phosphorylation and SUMOylation. A scan for small ubiquitin-like modifier (SUMO)-interacting motifs (SIMs) revealed two SIMs in IE2, and a real-time SUMOylation assay indicated that the N-terminal SIM (IE2-SIM1) enhances IE2 SUMOylation up to 4-fold. Kinetic analysis and structural studies disclosed that IE2 is a SUMO cis-E3 ligase. We also found that two putative casein kinase 2 (CK2) sites adjacent to IE2-SIM1 are phosphorylated in vitro and in cells. The phosphorylation drastically increased IE2-SUMO affinity, IE2 SUMOylation, and cis-E3 activity of IE2. Additional salt bridges between the phosphoserines and SUMO accounted for the increased IE2-SUMO affinity. Phosphorylation also enhanced the SUMO-dependent transactivation activity and auto-repression activity of IE2. Together, our findings highlight a novel mechanism whereby SUMOylation and phosphorylation of the viral cis-E3 ligase and transactivator protein IE2 work in tandem to enable transcriptional regulation of viral gene.


Assuntos
Caseína Quinase II/genética , Proteínas Imediatamente Precoces/genética , Fosforilação/genética , Proteína SUMO-1/genética , Sumoilação/genética , Transativadores/genética , Sítios de Ligação , Caseína Quinase II/química , Citomegalovirus/enzimologia , Citomegalovirus/genética , Regulação Viral da Expressão Gênica/genética , Humanos , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Cinética , Domínios e Motivos de Interação entre Proteínas/genética , Processamento de Proteína Pós-Traducional , Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Transativadores/química , Transativadores/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA