Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(3)2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38338831

RESUMO

Many proteins lack stable 3D structures. These intrinsically disordered proteins (IDPs) or hybrid proteins containing ordered domains with intrinsically disordered protein regions (IDPRs) often carry out regulatory functions related to molecular recognition and signal transduction. IDPs/IDPRs constitute a substantial portion of the human proteome and are termed "the unfoldome". Herein, we probe the human breast cancer unfoldome and investigate relations between IDPs and key disease genes and pathways. We utilized bottom-up proteomics, MudPIT (Multidimensional Protein Identification Technology), to profile differentially expressed IDPs in human normal (MCF-10A) and breast cancer (BT-549) cell lines. Overall, we identified 2271 protein groups in the unfoldome of normal and cancer proteomes, with 148 IDPs found to be significantly differentially expressed in cancer cells. Further analysis produced annotations of 140 IDPs, which were then classified to GO (Gene Ontology) categories and pathways. In total, 65% (91 of 140) IDPs were related to various diseases, and 20% (28 of 140) mapped to cancer terms. A substantial portion of the differentially expressed IDPs contained disordered regions, confirmed by in silico characterization. Overall, our analyses suggest high levels of interactivity in the human cancer unfoldome and a prevalence of moderately and highly disordered proteins in the network.


Assuntos
Neoplasias da Mama , Proteínas Intrinsicamente Desordenadas , Humanos , Feminino , Dobramento de Proteína , Conformação Proteica , Proteômica , Proteínas Intrinsicamente Desordenadas/química , Proteoma/metabolismo , Neoplasias da Mama/genética
2.
Int J Biol Macromol ; 252: 126526, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37633550

RESUMO

Proteins play a crucial role in many biological processes, where their interaction with other proteins are integral. Abnormal protein-protein interactions (PPIs) have been linked to various diseases including cancer, and thus targeting PPIs holds promise for drug development. However, experimental confirmation of the peculiarities of PPIs is challenging due to their dynamic and transient nature. As a complement to experimental technologies, multiple computational molecular docking (MD) methods have been developed to predict the structures of protein-protein complexes and their dynamics, still requiring further improvements in several issues. Here, we report an improved MD method, namely three-software docking (3SD), by employing three popular protein-peptide docking software (CABS-dock, HPEPDOCK, and HADDOCK) in combination to ensure constant quality for most targets. We validated our 3SD performance in known protein-peptide interactions (PpIs). We also enhanced MD performance in proteins having intrinsically disordered regions (IDRs) by applying the modified 3SD strategy, the three-software docking after removing random coiled IDR (3SD-RR), to the comparable crystal PpI structures. At the end, we applied 3SD-RR to the AlphaFold2-predicted receptors, yielding an efficient prediction of PpI pose with high relevance to the experimental data regardless of the presence of IDRs or the availability of receptor structures. Our study provides an improved solution to the challenges in studying PPIs through computational docking and has the potential to contribute to PPIs-targeted drug discovery. SIGNIFICANCE STATEMENT: Protein-protein interactions (PPIs) are integral to life, and abnormal PPIs are associated with diseases such as cancer. Studying protein-peptide interactions (PpIs) is challenging due to their dynamic and transient nature. Here we developed improved docking methods (3SD and 3SD-RR) to predict the PpI poses, ensuring constant quality in most targets and also addressing issues like intrinsically disordered regions (IDRs) and artificial intelligence-predicted structures. Our study provides an improved solution to the challenges in studying PpIs through computational docking and has the potential to contribute to PPIs-targeted drug discovery.


Assuntos
Inteligência Artificial , Neoplasias , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Proteínas/química , Software , Peptídeos/química
3.
Synth Syst Biotechnol ; 6(4): 429-436, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34901481

RESUMO

Tuberculosis drug resistance continues to threaten global health but the underline molecular mechanisms are not clear. Ethambutol (EMB), one of the well-known first - line drugs in tuberculosis treatment is, unfortunately, not free from drug resistance problems. Genomic studies have shown that some genetic mutations in Mycobacterium tuberculosis (Mtb) EmbR, and EmbC/A/B genes cause EMB resistance. EmbR-PknH pair controls embC/A/B operon, which encodes EmbC/A/B genes, and EMB interacts with EmbA/B proteins. However, the EmbR binding site on PknH was unknown. We conducted molecular simulation on the EmbR- peptides binding structures and discovered phosphorylated PknH 273-280 (N'-HEALSPDPD-C') makes ß strand with the EmbR FHA domain, as ß-MoRF (MoRF; molecular recognition feature) does at its binding site. Hydrogen bond number analysis also supported the peptides' ß-MoRF forming activity at the EmbR FHA domain. Also, we discovered that previously known phosphorylation residues might have their chronological order according to the phosphorylation status. The discovery validated that Mtb PknH 273-280 (N'-HEALSDPD-C') has reliable EmbR binding affinity. This approach is revolutionary in the computer-aided drug discovery field, because it is the first trial to discover the protein-protein interaction site, and find binding partner in nature from this site.

4.
FEBS Lett ; 595(24): 2981-2994, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34704263

RESUMO

The Asp233-Asp246 pair is highly conserved in Class A ß-lactamases, which hydrolyze ß-lactam antibiotics. Here, we characterize its function using CTX-M-14 ß-lactamase. The D233N mutant displayed decreased activity that is substrate-dependent, with reductions in kcat /Km ranging from 20% for nitrocefin to 6-fold for cefotaxime. In comparison, the mutation reduced the binding of a known reversible inhibitor by 10-fold. The mutant structures showed movement of the 213-219 loop and the loss of the Thr216-Thr235 hydrogen bond, which was restored by inhibitor binding. Mutagenesis of Thr216 further highlighted its contribution to CTX-M activity. These results demonstrate the importance of the aspartate pair to CTX-M hydrolysis of substrates with bulky side chains, while suggesting increased protein flexibility as a means to evolve drug resistance.


Assuntos
Ácido Aspártico/genética , Sequência Conservada , Mutação/genética , Inibidores de beta-Lactamases/farmacologia , beta-Lactamases/química , beta-Lactamases/genética , Cristalografia por Raios X , Ligantes , Proteínas Mutantes/química , Especificidade por Substrato , Tetrazóis/química , Tetrazóis/farmacologia , Inibidores de beta-Lactamases/química , beta-Lactamases/metabolismo
5.
Int J Mol Sci ; 21(15)2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32722024

RESUMO

Intrinsically disordered proteins exist as highly dynamic conformational ensembles of diverse forms. However, the majority of virtual screening only focuses on proteins with defined structures. This means that computer-aided drug discovery is restricted. As a breakthrough, understanding the structural characteristics of intrinsically disordered proteins and its application can open the gate for unrestricted drug discovery. First, we segmented the target disorder-to-order transition region into a series of overlapping 20-amino-acid-long peptides. Folding prediction generated diverse conformations of these peptides. Next, we applied molecular docking, new evaluation score function, and statistical analysis. This approach successfully distinguished known compounds and their corresponding binding regions. Especially, Myc proto-oncogene protein (MYC) inhibitor 10058F4 was well distinguished from others of the chemical compound library. We also studied differences between the two Methyl-CpG-binding domain protein 2 (MBD2) inhibitors (ABA (2-amino-N-[[(3S)-2,3-dihydro-1,4-benzodioxin-3-yl]methyl]-acetamide) and APC ((R)-(3-(2-Amino-acetylamino)-pyrrolidine-1-carboxylic acid tert-butyl ester))). Both compounds bind MBD2 through electrostatic interaction behind its p66α-binding site. ABA is also able to bind p66α through electrostatic interaction behind its MBD2-binding site while APC-p66α binding was nonspecific. Therefore, structural heterogeneity mimicking of the disorder-to-order transition region at the peptide level and utilization of the new docking score function represent a useful approach that can efficiently discriminate compounds for expanded virtual screening toward intrinsically disordered proteins.


Assuntos
Proteínas de Ligação a DNA/química , Descoberta de Drogas , Proteínas Intrinsicamente Desordenadas/química , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Humanos , Ligação Proteica , Proto-Oncogene Mas , Eletricidade Estática
6.
Cells ; 9(5)2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32403252

RESUMO

Chromatin remodeling, including histone modification, chromatin (un)folding, and nucleosome remodeling, is a significant transcriptional regulation mechanism. By these epigenetic modifications, transcription factors and their regulators are recruited to the promoters of target genes, and thus gene expression is controlled through either transcriptional activation or repression. The Mat1-mediated transcriptional repressor (MMTR)/DNA methyltransferase 1 (DNMT1)-associated protein (Dmap1) is a transcription corepressor involved in chromatin remodeling, cell cycle regulation, DNA double-strand break repair, and tumor suppression. The Tip60-p400 complex proteins, including MMTR/Dmap1, interact with the oncogene Myc in embryonic stem cells (ESCs). These proteins interplay with the stem cell-related proteome networks and regulate gene expressions. However, the detailed mechanisms of their functions are unknown. Here, we show that MMTR/Dmap1, along with other Tip60-p400 complex proteins, bind the promoters of differentiation commitment genes in mouse ESCs. Hence, MMTR/Dmap1 controls gene expression alterations during differentiation. Furthermore, we propose a novel mechanism of MMTR/Dmap1 function in early stage lineage commitment of mouse ESCs by crosstalk with the polycomb group (PcG) proteins. The complex controls histone mark bivalency and transcriptional poising of commitment genes. Taken together, our comprehensive findings will help better understand the MMTR/Dmap1-mediated transcriptional regulation in ESCs and other cell types.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diferenciação Celular , Montagem e Desmontagem da Cromatina , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Lisina/metabolismo , Lisina Acetiltransferase 5/metabolismo , Metilação , Camundongos , Camundongos SCID , Modelos Biológicos , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/química , Transativadores/metabolismo
7.
Sci Adv ; 5(11): eaav9810, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31799386

RESUMO

Although intrinsically disordered protein regions (IDPRs) are commonly engaged in promiscuous protein-protein interactions (PPIs), using them as drug targets is challenging due to their extreme structural flexibility. We report a rational discovery of inhibitors targeting an IDPR of MBD2 that undergoes disorder-to-order transition upon PPI and is critical for the regulation of the Mi-2/NuRD chromatin remodeling complex (CRC). Computational biology was essential for identifying target site, searching for promising leads, and assessing their binding feasibility and off-target probability. Molecular action of selected leads inhibiting the targeted PPI of MBD2 was validated in vitro and in cell, followed by confirming their inhibitory effects on the epithelial-mesenchymal transition of various cancer cells. Identified lead compounds appeared to potently inhibit cancer metastasis in a murine xenograft tumor model. These results constitute a pioneering example of rationally discovered IDPR-targeting agents and suggest Mi-2/NuRD CRC and/or MBD2 as a promising target for treating cancer metastasis.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas Intrinsicamente Desordenadas/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Domínios Proteicos/efeitos dos fármacos , Animais , Biologia Computacional , Descoberta de Drogas/métodos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/antagonistas & inibidores , Camundongos , Modelos Moleculares , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Genet Metab ; 128(3): 178-189, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31345668

RESUMO

5-Aminolevulinate (ALA) synthase (ALAS), a homodimeric pyridoxal-5'-phosphate (PLP)-dependent enzyme, catalyzes the first step of heme biosynthesis in metazoa, fungi and α-proteobacteria. In this review, we focus on the advances made in unraveling the mechanism of the ALAS-catalyzed reaction during the past decade. The interplay between the PLP cofactor and the protein moiety determines and modulates the multi-intermediate reaction cycle of ALAS, which involves the decarboxylative condensation of two substrates, glycine and succinyl-CoA. Substrate binding and catalysis are rapid, and product (ALA) release dominates the overall ALAS kinetic mechanism. Interconversion between a catalytically incompetent, open conformation and a catalytically competent, closed conformation is linked to ALAS catalysis. Reversion to the open conformation, coincident with ALA dissociation, defines the slowest step of the reaction cycle. These findings were further substantiated by introducing seven mutations in the16-amino acid loop that gates the active site, yielding an ALAS variant with a greatly increased rate of catalytic turnover and heightened specificity constants for both substrates. Recently, molecular dynamics (MD) simulation analysis of various dimeric ALAS forms revealed that the seven active site loop mutations caused the proteins to adopt different conformations. In particular, the emergence of a ß-strand in the mutated loop, which interacted with two preexisting ß-strands to form an anti-parallel three-stranded ß-sheet, conferred the murine heptavariant with a more stable open conformation and prompted faster product release than wild-type mALAS2. Moreover, the dynamics of the mALAS2 active site loop anti-correlated with that of the 35 amino acid C-terminal sequence. This led us to propose that this C-terminal extension, which is absent in prokaryotic ALASs, finely tunes mammalian ALAS activity. Based on the above results, we extend our previous proposal to include that discovery of a ligand inducing the mammalian C-terminal extension to fold offers a good prospect for the development of a new drug for X-linked protoporphyria and/or other porphyrias associated with enhanced ALAS activity and/or porphyrin accumulation.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Vias Biossintéticas , Heme/biossíntese , Fosfato de Piridoxal/metabolismo , Catálise , Humanos , Cinética , Simulação de Dinâmica Molecular , Conformação Proteica , Especificidade por Substrato
9.
Int J Mol Sci ; 19(7)2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29958424

RESUMO

Biosynthesis of heme represents a complex process that involves multiple stages controlled by different enzymes. The first of these proteins is a pyridoxal 5'-phosphate (PLP)-dependent homodimeric enzyme, 5-aminolevulinate synthase (ALAS), that catalyzes the rate-limiting step in heme biosynthesis, the condensation of glycine with succinyl-CoA. Genetic mutations in human erythroid-specific ALAS (ALAS2) are associated with two inherited blood disorders, X-linked sideroblastic anemia (XLSA) and X-linked protoporphyria (XLPP). XLSA is caused by diminished ALAS2 activity leading to decreased ALA and heme syntheses and ultimately ineffective erythropoiesis, whereas XLPP results from "gain-of-function" ALAS2 mutations and consequent overproduction of protoporphyrin IX and increase in Zn2+-protoporphyrin levels. All XLPP-linked mutations affect the intrinsically disordered C-terminal tail of ALAS2. Our earlier molecular dynamics (MD) simulation-based analysis showed that the activity of ALAS2 could be regulated by the conformational flexibility of the active site loop whose structural features and dynamics could be changed due to mutations. We also revealed that the dynamic behavior of the two protomers of the ALAS2 dimer differed. However, how the structural dynamics of ALAS2 active site loop and C-terminal tail dynamics are related to each other and contribute to the homodimer asymmetry remained unanswered questions. In this study, we used bioinformatics and computational biology tools to evaluate the role(s) of the C-terminal tail dynamics in the structure and conformational dynamics of the murine ALAS2 homodimer active site loop. To assess the structural correlation between these two regions, we analyzed their structural displacements and determined their degree of correlation. Here, we report that the dynamics of ALAS2 active site loop is anti-correlated with the dynamics of the C-terminal tail and that this anti-correlation can represent a molecular basis for the functional and dynamic asymmetry of the ALAS2 homodimer.


Assuntos
5-Aminolevulinato Sintetase/química , Anemia Sideroblástica/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Heme/química , 5-Aminolevulinato Sintetase/genética , Anemia Sideroblástica/patologia , Animais , Domínio Catalítico , Biologia Computacional , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Heme/biossíntese , Heme/genética , Humanos , Camundongos , Simulação de Dinâmica Molecular , Mutação/genética , Multimerização Proteica/genética
10.
J Biomol Struct Dyn ; 36(1): 152-165, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-27928941

RESUMO

Heme biosynthesis, a complex, multistage, and tightly controlled process, starts with 5-aminolevulinate (ALA) production, which, in metazoa and certain bacteria, is a reaction catalyzed by 5-aminolevulinate synthase (ALAS), a pyridoxal 5'-phosphate (PLP)-dependent enzyme. Functional aberrations in ALAS are associated with several human diseases. ALAS can adopt open and closed conformations, with segmental rearrangements of a C-terminal, 16-amino acid loop and an α-helix regulating accessibility to the ALAS active site. Of the murine erythroid ALAS (mALAS2) forms previously engineered to assess the role of the flexible C-terminal loop versus mALAS2 function one stood out due to its impressive gain in catalytic power. To elucidate how the simultaneously introduced seven mutations of this activity-enhanced variant affected structural and dynamic properties of mALAS2, we conducted extensive molecular dynamics simulation analysis of the dimeric forms of wild-type mALAS2, hepta-variant and Rhodobacter capsulatus ALAS (aka R. capsulatus HemA). This analysis revealed that the seven simultaneous mutations in the C-terminal loop, which extends over the active site of the enzyme, caused the bacterial and murine proteins to adopt different conformations. Specifically, a new ß-strand in the mutated 'loop' led to interaction with two preexisting ß-strands and formation of an anti-parallel three-stranded ß-sheet, which likely endowed the murine hepta-variant a more 'stable' open conformation than that of wild-type mALAS2, consistent with a kinetic mechanism involving a faster closed-to-open conformation transition and product release for the mutated than wild-type enzyme. Further, the dynamic behavior of the mALAS2 protomers was strikingly different in the two dimeric forms.


Assuntos
5-Aminolevulinato Sintetase/química , Domínio Catalítico , Simulação de Dinâmica Molecular , Conformação Proteica , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , Sequência de Aminoácidos , Animais , Biocatálise , Humanos , Cinética , Camundongos , Mutação , Rhodobacter capsulatus/enzimologia , Rhodobacter capsulatus/genética , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
11.
Curr Protein Pept Sci ; 18(5): 453-481, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27804860

RESUMO

Prostate cancer is one of the major threats to the man's health. There are several mechanisms of the prostate cancer development characterized by the involvement of various androgen-related and androgen-unrelated factors in prostate cancer pathogenesis and in the metastatic carcinogenesis of prostate. In all these processes, proteins play various important roles, and the KEGG database has information on 88 human proteins experimentally shown to be involved in prostate cancer. It is known that many proteins associated with different human maladies are intrinsically disordered (i.e., they do not have stable secondary and/or tertiary structure in their unbound states). The goal of this review is to consider several highly disordered proteins known to be associated with the prostate cancer pathogenesis in order to better understand the roles of disordered proteins in this disease. We also hope that consideration of the pathology-related proteins from the perspective of intrinsic disorder can potentially lead to future experimental studies of these proteins to find novel pathways associated with prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Intrinsicamente Desordenadas/química , Proteínas de Neoplasias/química , Neoplasias da Próstata/genética , Processamento de Proteína Pós-Traducional , Proteoma/química , Sequência de Aminoácidos , Redes Reguladoras de Genes , Humanos , Proteínas Intrinsicamente Desordenadas/genética , Proteínas Intrinsicamente Desordenadas/metabolismo , Masculino , Modelos Moleculares , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Conformação Proteica , Mapeamento de Interação de Proteínas , Proteoma/genética , Proteoma/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
12.
Asian J Androl ; 18(5): 662-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27453073

RESUMO

Prostatic diseases such as prostate cancer and benign prostatic hyperplasia are highly prevalent among men. The number of studies focused on the abundance and roles of intrinsically disordered proteins in prostate cancer is rather limited. The goal of this study is to analyze the prevalence and degree of disorder in proteins that were previously associated with the prostate cancer pathogenesis and to compare these proteins to the entire human proteome. The analysis of these datasets provides means for drawing conclusions on the roles of disordered proteins in this common male disease. We also hope that the results of our analysis can potentially lead to future experimental studies of these proteins to find novel pathways associated with this disease.


Assuntos
Proteínas Intrinsicamente Desordenadas/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Bases de Dados de Proteínas , Humanos , Masculino , Próstata/patologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Dobramento de Proteína , Proteoma
13.
Mol Biosyst ; 12(9): 2798-817, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27377881

RESUMO

Recent analyses indicated that autophagy can be regulated via some nuclear transcriptional networks and many important players in the autophagy and other forms of programmed cell death are known to be intrinsically disordered. To this end, we analyzed similarities and differences in the intrinsic disorder distribution of nuclear and non-nuclear proteins related to autophagy. We also looked at the peculiarities of the distribution of the intrinsically disordered autophagy-related proteins in various intra-nuclear organelles, such as the nucleolus, chromatin, Cajal bodies, nuclear speckles, promyelocytic leukemia (PML) nuclear bodies, nuclear lamina, nuclear pores, and perinucleolar compartment. This analysis revealed that the autophagy-related proteins constitute about 2.5% of the non-nuclear proteins and 3.3% of the nuclear proteins, which corresponds to a substantial enrichment by about 32% in the nucleus. Curiously, although, in general, the autophagy-related proteins share similar characteristics of disorder with a generic set of all non-nuclear proteins, chromatin and nuclear speckles are enriched in the intrinsically disordered autophagy proteins (29 and 37% of these proteins are disordered, respectively) and have high disorder content at 0.24 and 0.27, respectively. Therefore, our data suggest that some of the nuclear disordered proteins may play important roles in autophagy.


Assuntos
Autofagia , Núcleo Celular/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Animais , Autofagia/genética , Núcleo Celular/genética , Cromatina/metabolismo , Bases de Dados de Proteínas , Proteínas Fúngicas/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/genética , Camundongos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Biol Chem ; 397(8): 731-51, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27074551

RESUMO

Cardiac troponin is a dynamic complex of troponin C, troponin I, and troponin T (TnC, TnI, and TnT, respectively) found in the myocyte thin filament where it plays an essential role in cardiac muscle contraction. Mutations in troponin subunits are found in inherited cardiomyopathies, such as hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). The highly dynamic nature of human cardiac troponin and presence of numerous flexible linkers in its subunits suggest that understanding of structural and functional properties of this important complex can benefit from the consideration of the protein intrinsic disorder phenomenon. We show here that mutations causing decrease in the disorder score in TnI and TnT are significantly more abundant in HCM and DCM than mutations leading to the increase in the disorder score. Identification and annotation of intrinsically disordered regions in each of the troponin subunits conducted in this study can help in better understanding of the roles of intrinsic disorder in regulation of interactomes and posttranslational modifications of these proteins. These observations suggest that disease-causing mutations leading to a decrease in the local flexibility of troponins can trigger a whole plethora of functional changes in the heart.


Assuntos
Cardiomiopatias/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Troponina C/metabolismo , Troponina I/metabolismo , Troponina T/metabolismo , Cardiomiopatias/genética , Humanos , Modelos Moleculares , Mutação , Troponina C/análise , Troponina C/genética , Troponina I/análise , Troponina I/genética , Troponina T/análise , Troponina T/genética
15.
Int J Mol Sci ; 17(1)2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26712748

RESUMO

The cell nucleus contains a number of membrane-less organelles or intra-nuclear compartments. These compartments are dynamic structures representing liquid-droplet phases which are only slightly denser than the bulk intra-nuclear fluid. They possess different functions, have diverse morphologies, and are typically composed of RNA (or, in some cases, DNA) and proteins. We analyzed 3005 mouse proteins localized in specific intra-nuclear organelles, such as nucleolus, chromatin, Cajal bodies, nuclear speckles, promyelocytic leukemia (PML) nuclear bodies, nuclear lamina, nuclear pores, and perinuclear compartment and compared them with ~29,863 non-nuclear proteins from mouse proteome. Our analysis revealed that intrinsic disorder is enriched in the majority of intra-nuclear compartments, except for the nuclear pore and lamina. These compartments are depleted in proteins that lack disordered domains and enriched in proteins that have multiple disordered domains. Moonlighting proteins found in multiple intra-nuclear compartments are more likely to have multiple disordered domains. Protein-protein interaction networks in the intra-nuclear compartments are denser and include more hubs compared to the non-nuclear proteins. Hubs in the intra-nuclear compartments (except for the nuclear pore) are enriched in disorder compared with non-nuclear hubs and non-nuclear proteins. Therefore, our work provides support to the idea of the functional importance of intrinsic disorder in the cell nucleus and shows that many proteins associated with sub-nuclear organelles in nuclei of mouse cells are enriched in disorder. This high level of disorder in the mouse nuclear proteins defines their ability to serve as very promiscuous binders, possessing both large quantities of potential disorder-based interaction sites and the ability of a single such site to be involved in a large number of interactions.


Assuntos
Núcleo Celular/metabolismo , Proteínas Intrinsicamente Desordenadas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Ligação a DNA/metabolismo , Proteínas Intrinsicamente Desordenadas/química , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Proteoma/química , Proteoma/metabolismo , Proteínas de Ligação a RNA/metabolismo
16.
Mol Biosyst ; 10(4): 925-40, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24549315

RESUMO

Sup35 protein (Sup35p), or eukaryotic peptide chain release factor GTP binding subunit (eRF3), is a well-known yeast prion responsible for the characteristic [PSI(+)] trait. N- and M-domains of this protein have been the foci of intensive research due to their importance for the prion formation. Sup35p C-terminal domain (Sup35pC) is essential for translation termination and cell viability. Deletion of Sup35pC was shown to lead to malformation of cells during mitosis. In this study we confirm that Sup35pC domain possesses high sequence and structural similarity to the eukaryotic translation elongation factor 1-α (eEF1A) from yeast and show that its sequence is conserved across different species including human. Because cell malformation during mitosis could be due to the deregulation of cytoskeleton formation, and since a Sup35 paralog eEF1A is known to act as an actin modulating protein, we focused on establishing of the relationships between the Sup35pC and modulation of the cytoskeleton formation. We found 104 co-partners between Sup35pC and EF1A of S. cerevisiae, and 18 partners of human ERF3A. Based on the analysis of known and modeled structures of some effectors and partners we found possible protein-protein interactions. Based on our study, we propose that Sup35pC may serve as actin modulator during mitosis.


Assuntos
Citoesqueleto/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Fatores de Terminação de Peptídeos/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Adenosina Trifosfatases/genética , Sequência de Aminoácidos , Classe Ia de Fosfatidilinositol 3-Quinase , Proteínas de Choque Térmico HSP70/genética , Humanos , Mitose , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/genética , Príons/genética , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência , Análise de Sequência de Proteína
17.
PLoS One ; 8(8): e73476, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24014171

RESUMO

The abundance and potential functional roles of intrinsically disordered regions in aquaporin-4, Kir4.1, a dystrophin isoforms Dp71, α-1 syntrophin, and α-dystrobrevin; i.e., proteins constituting the functional core of the astrocytic dystrophin-associated protein complex (DAPC), are analyzed by a wealth of computational tools. The correlation between protein intrinsic disorder, single nucleotide polymorphisms (SNPs) and protein function is also studied together with the peculiarities of structural and functional conservation of these proteins. Our study revealed that the DAPC members are typical hybrid proteins that contain both ordered and intrinsically disordered regions. Both ordered and disordered regions are important for the stabilization of this complex. Many disordered binding regions of these five proteins are highly conserved among vertebrates. Conserved eukaryotic linear motifs and molecular recognition features found in the disordered regions of five protein constituting DAPC likely enhance protein-protein interactions that are required for the cellular functions of this complex. Curiously, the disorder-based binding regions are rarely affected by SNPs suggesting that these regions are crucial for the biological functions of their corresponding proteins.


Assuntos
Galinhas/genética , Complexo de Proteínas Associadas Distrofina/genética , Lagartos/genética , Distrofias Musculares/genética , Polimorfismo de Nucleotídeo Único , Peixe-Zebra/genética , Animais , Galinhas/metabolismo , Complexo de Proteínas Associadas Distrofina/química , Complexo de Proteínas Associadas Distrofina/metabolismo , Humanos , Lagartos/metabolismo , Camundongos , Distrofias Musculares/metabolismo , Distrofia Muscular Animal , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Xenopus laevis , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...