Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Biol Chem ; 298(2): 101514, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929165

RESUMO

Recognition of human autophagy-related 8 (hATG8) proteins by autophagy receptors represents a critical step within this cellular quality control system. Autophagy impairment is known to be a pathogenic mechanism in the motor neuron disorder amyotrophic lateral sclerosis (ALS). Overlapping but specific roles of hATG8 proteins belonging to the LC3 and GABARAP subfamilies are incompletely understood, and binding selectivity is typically overlooked. We previously showed that an ALS-associated variant of the SQSTM1/p62 (p62) autophagy receptor bearing an L341V mutation within its ATG8-interacting motif (AIM) impairs recognition of LC3B in vitro, yielding an autophagy-deficient phenotype. Improvements in understanding of hATG8 recognition by AIMs now distinguish LC3-interaction and GABARAP-interaction motifs and predict the effects of L341V substitution may extend beyond loss of function to biasing AIM binding preference. Through biophysical analyses, we confirm impaired binding of the L341V-AIM mutant to LC3A, LC3B, GABARAP, and GABARAPL1. In contrast, p62 AIM interactions with LC3C and GABARAPL2 are unaffected by this mutation. Isothermal titration calorimetry and NMR investigations provided insights into the entropy-driven GABARAPL2/p62 interaction and how the L341V mutation may be tolerated. Competition binding demonstrated reduced association of the L341V-AIM with one hATG8 manifests as a relative increase in association with alternate hATG8s, indicating effective reprogramming of hATG8 selectivity. These data highlight how a single AIM peptide might compete for binding with different hATG8s and suggest that the L341V-AIM mutation may be neomorphic, representative of a disease mechanism that likely extends into other human disorders.


Assuntos
Esclerose Lateral Amiotrófica , Família da Proteína 8 Relacionada à Autofagia , Proteína Sequestossoma-1 , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo
2.
Nucleic Acids Res ; 48(8): 4179-4194, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32182342

RESUMO

Abnormal DUX4 expression in skeletal muscles plays a key role in facioscapulohumeral muscular dystrophy (FSHD) pathogenesis, although the molecular mechanisms regulating DUX4 expression are not fully defined. Using bioinformatic analysis of the genomic DUX4 locus, we have identified a number of putative G-quadruplexes (GQs) forming sequences. Their presence was confirmed in synthetic oligonucleotiode sequences derived from the enhancer, promoter and transcript of DUX4 through circular dichroism and nuclear magnetic resonance analysis. We further examined the binding affinity of a naturally occurring GQ stabilizing compound, berberine, to these non-canonical genetic structures using UV-Vis and fluorescence spectroscopy. Subsequent in vitro study in FSHD patient myoblasts indicated that berberine treatment reduced DUX4 expression and also expression of genes normally switched on by DUX4. Further investigation in a mouse model overexpressing exogenous DUX4 confirmed the therapeutic effects of berberine in downregulating DUX4 protein expression, inhibiting muscle fibrosis, and consequently rescuing muscle function. Our data demonstrate for the first time that GQs are present in the DUX4 locus and that the GQ interactive ligand reduces DUX4 expression suggesting potential role of GQs in FSHD pathogenesis. Our work provides the basis of a novel therapeutic strategy for the treatment of FSHD.


Assuntos
Quadruplex G , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Animais , Berberina/química , Berberina/farmacologia , Fusão Celular , Linhagem Celular Tumoral , Células Clonais , Regulação para Baixo , Elementos Facilitadores Genéticos , Fibrose , Proteínas de Homeodomínio/metabolismo , Ligantes , Masculino , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiologia , Distrofia Muscular Facioescapuloumeral/metabolismo , Mioblastos/fisiologia , Motivos de Nucleotídeos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo
3.
Blood Adv ; 3(9): 1450-1459, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31053572

RESUMO

Cell-surface receptor interactions between leukocyte integrin macrophage-1 antigen (Mac-1, also known as CR3, αMß2, CD11b/CD18) and platelet glycoprotein Ibα (GPIbα) are critical to vascular inflammation. To define the key residues at the binding interface, we used nuclear magnetic resonance (NMR) to assign the spectra of the mouse Mac-1 I-domain and mapped the residues contacting the mouse GPIbα N-terminal domain (GPIbαN) to the locality of the integrin metal ion-dependant adhesion site (MIDAS) surface. We next determined the crystal structures of the mouse GPIbαN and Mac-1 I-domain to 2 Å and 2.5 Å resolution, respectively. The mouse Mac-1 I-domain crystal structure reveals an active conformation that is stabilized by a crystal contact from the α7-helix with a glutamate side chain completing the octahedral coordination sphere of the MIDAS Mg2+ ion. The amino acid sequence of the α7-helix and disposition of the glutamic acid matches the C-terminal capping region α-helix of GPIbα effectively acting as a ligand mimetic. Using these crystal structures in combination with NMR measurements and docking analysis, we developed a model whereby an acidic residue from the GPIbα leucine-rich repeat (LRR) capping α-helix coordinates directly to the Mac-1 MIDAS Mg2+ ion. The Mac-1:GPIbαN complex involves additional interactions consolidated by an elongated pocket flanking the GPIbαN LRR capping α-helix. The GPIbαN α-helix has an HxxxE motif, which is equivalent by homology to RxxxD from the human GPIbαN. Subsequent mutagenesis of residues at this interface, coupled with surface plasmon resonance studies, confirmed the importance of GPIbαN residues H218, E222, and the Mac-1 MIDAS residue T209 to formation of the complex.


Assuntos
Antígeno de Macrófago 1/química , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Motivos de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Leucócitos/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/metabolismo , Magnésio/química , Camundongos , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Estrutura Terciária de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação
4.
Nucleic Acids Res ; 47(4): 2101-2112, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30534966

RESUMO

The homotetrameric DnaD protein is essential in low G+C content gram positive bacteria and is involved in replication initiation at oriC and re-start of collapsed replication forks. It interacts with the ubiquitously conserved bacterial master replication initiation protein DnaA at the oriC but structural and functional details of this interaction are lacking, thus contributing to our incomplete understanding of the molecular details that underpin replication initiation in bacteria. DnaD comprises N-terminal (DDBH1) and C-terminal (DDBH2) domains, with contradicting bacterial two-hybrid and yeast two-hybrid studies suggesting that either the former or the latter interact with DnaA, respectively. Using Nuclear Magnetic Resonance (NMR) we showed that both DDBH1 and DDBH2 interact with the N-terminal domain I of DnaA and studied the DDBH2 interaction in structural detail. We revealed two families of conformations for the DDBH2-DnaA domain I complex and showed that the DnaA-interaction patch of DnaD is distinct from the DNA-interaction patch, suggesting that DnaD can bind simultaneously DNA and DnaA. Using sensitive single-molecule FRET techniques we revealed that DnaD remodels DnaA-DNA filaments consistent with stretching and/or untwisting. Furthermore, the DNA binding activity of DnaD is redundant for this filament remodelling. This in turn suggests that DnaA and DnaD are working collaboratively in the oriC to locally melt the DNA duplex during replication initiation.


Assuntos
Proteínas de Bactérias/genética , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Origem de Replicação/genética , Bacillus subtilis/genética , Proteínas de Bactérias/química , Proteínas de Ligação a DNA/química , DnaB Helicases/química , DnaB Helicases/genética , Espectroscopia de Ressonância Magnética , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexo de Reconhecimento de Origem/genética , Ligação Proteica/genética , Domínios Proteicos/genética , Relação Estrutura-Atividade
5.
Methods Mol Biol ; 1449: 203-13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27613037

RESUMO

Unanchored polyubiquitin chains are endogenous non-substrate linked ubiquitin polymers which have emerging roles in the control of cellular physiology. We describe an affinity purification method based on an isolated ubiquitin-binding domain, the ZnF_UBP domain of the deubiquitinating enzyme USP5, which permits the selective purification of mixtures of endogenous unanchored polyubiquitin chains that are amenable to downstream molecular analyses. Further, we present methods for detection of unanchored polyubiquitin chains in purified fractions.


Assuntos
Poliubiquitina/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Poliubiquitina/isolamento & purificação , Ligação Proteica , Proteômica/métodos , Ubiquitina/isolamento & purificação , Proteases Específicas de Ubiquitina/isolamento & purificação , Proteases Específicas de Ubiquitina/metabolismo , Proteínas Ubiquitinadas/isolamento & purificação , Proteínas Ubiquitinadas/metabolismo , Ubiquitinação
6.
Mol Cell Neurosci ; 76: 52-58, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27554286

RESUMO

The transcription factor Nrf2 and its repressor protein Keap1 play key roles in the regulation of antioxidant stress responses and both Keap1-Nrf2 signalling and oxidative stress have been implicated in the pathogenesis of the ALS-FTLD spectrum of neurodegenerative disorders. The Keap1-binding partner and autophagy receptor SQSTM1/p62 has also recently been linked genetically to ALS-FTLD, with some missense mutations identified in patients mapping within or close to its Keap1-interacting region (KIR, residues 347-352). Here we report the effects on protein function of four different disease associated mutations of SQSTM1/p62 which affect the KIR region. Only mutations mapping precisely to the KIR (P348L and G351A) were associated with a loss of Keap1 binding in co-immunoprecipitations comparable to wild-type SQSTM1/p62. These selective effects on Keap1 recognition were entirely rational based on protein structural models. Consistent with impaired Keap1 binding, the P348L and G351A KIR mutants showed reduced ability to activate Nrf2 signalling compared to wild-type SQSTM1/p62 in antioxidant response element (ARE)-luciferase reporter assays. The results suggest that SQSTM1 mutations within the KIR of SQSTM1/p62 contribute to aetiology of some cases of ALS-FTLD through a mechanism involving aberrant expression or regulation of oxidative response genes.


Assuntos
Esclerose Lateral Amiotrófica/genética , Degeneração Lobar Frontotemporal/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Mutação de Sentido Incorreto , Fator 2 Relacionado a NF-E2/metabolismo , Proteína Sequestossoma-1/genética , Transdução de Sinais , Sítios de Ligação , Células HEK293 , Humanos , Ligação Proteica , Elementos de Resposta , Proteína Sequestossoma-1/química , Proteína Sequestossoma-1/metabolismo
7.
Autophagy ; 12(7): 1094-104, 2016 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-27158844

RESUMO

Growing evidence implicates impairment of autophagy as a candidate pathogenic mechanism in the spectrum of neurodegenerative disorders which includes amyotrophic lateral sclerosis and frontotemporal lobar degeneration (ALS-FTLD). SQSTM1, which encodes the autophagy receptor SQSTM1/p62, is genetically associated with ALS-FTLD, although to date autophagy-relevant functional defects in disease-associated variants have not been described. A key protein-protein interaction in autophagy is the recognition of a lipid-anchored form of LC3 (LC3-II) within the phagophore membrane by SQSTM1, mediated through its LC3-interacting region (LIR), and notably some ALS-FTLD mutations map to this region. Here we show that although representing a conservative substitution and predicted to be benign, the ALS-associated L341V mutation of SQSTM1 is defective in recognition of LC3B. We place our observations on a firm quantitative footing by showing the L341V-mutant LIR is associated with a ∼3-fold reduction in LC3B binding affinity and using protein NMR we rationalize the structural basis for the effect. This functional deficit is realized in motor neuron-like cells, with the L341V mutant EGFP-mCherry-SQSTM1 less readily incorporated into acidic autophagic vesicles than the wild type. Our data supports a model in which the L341V mutation limits the critical step of SQSTM1 recruitment to the phagophore. The oligomeric nature of SQSTM1, which presents multiple LIRs to template growth of the phagophore, potentially gives rise to avidity effects which amplify the relatively modest impact of any single mutation on LC3B binding. Over the lifetime of a neuron, impaired autophagy could expose a vulnerability, which ultimately tips the balance from cell survival toward cell death.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Autofagia/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Proteína Sequestossoma-1/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Autofagia/fisiologia , Degeneração Lobar Frontotemporal , Humanos , Proteínas Associadas aos Microtúbulos/genética , Neurônios Motores/metabolismo , Neurônios Motores/patologia
8.
Mol Microbiol ; 101(5): 731-42, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27085056

RESUMO

Ag(+) resistance was initially found on the Salmonella enetrica serovar Typhimurium multi-resistance plasmid pMG101 from burns patients in 1975. The putative model of Ag(+) resistance, encoded by the sil operon from pMG101, involves export of Ag(+) via an ATPase (SilP), an effluxer complex (SilCFBA) and a periplasmic chaperon of Ag(+) (SilE). SilE is predicted to be intrinsically disordered. We tested this hypothesis using structural and biophysical studies and show that SilE is an intrinsically disordered protein in its free apo-form but folds to a compact structure upon optimal binding to six Ag(+) ions in its holo-form. Sequence analyses and site-directed mutagenesis established the importance of histidine and methionine containing motifs for Ag(+) -binding, and identified a nucleation core that initiates Ag(+) -mediated folding of SilE. We conclude that SilE is a molecular sponge for absorbing metal ions.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/metabolismo , Prata/farmacologia , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Farmacorresistência Bacteriana , Escherichia coli/genética , Genes Bacterianos , Mutagênese Sítio-Dirigida , Óperon , Periplasma/metabolismo , Plasmídeos/efeitos dos fármacos , Plasmídeos/metabolismo , Infecções por Salmonella/tratamento farmacológico , Infecções por Salmonella/microbiologia , Homologia de Sequência de Aminoácidos
9.
Proteomics ; 16(14): 1961-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27037516

RESUMO

Unanchored polyubiquitin chains are emerging as important regulators of cellular physiology with diverse roles paralleling those of substrate-conjugated polyubiquitin. However tools able to discriminate unanchored polyubiquitin chains of different isopeptide linkages have not been reported. We describe the design of a linker-optimized ubiquitin-binding domain hybrid (t-UBD) containing two UBDs, a ZnF-UBP domain in tandem with a linkage-selective UBA domain, which exploits avidity effects to afford selective recognition of unanchored Lys48-linked polyubiquitin chains. Utilizing native MS to quantitatively probe binding affinities we confirm cooperative binding of the UBDs within the synthetic protein, and desired binding specificity for Lys48-linked ubiquitin dimers. Furthermore, MS/MS analyses indicate that the t-UBD, when applied as an affinity enrichment reagent, can be used to favor the purification of endogenous unanchored Lys48-linked polyubiquitin chains from mammalian cell extracts. Our study indicates that strategies for the rational design and engineering of polyubiquitin chain-selective binding in nonbiological polymers are possible, paving the way for the generation of reagents to probe unanchored polyubiquitin chains of different linkages and more broadly the 'ubiquitome'. All MS data have been deposited in the ProteomeXchange with identifier PXD004059 (http://proteomecentral.proteomexchange.org/dataset/PXD004059).


Assuntos
Bioensaio/normas , Lisina/metabolismo , Poliubiquitina/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Sítios de Ligação , Misturas Complexas/química , Expressão Gênica , Células HEK293 , Humanos , Cinética , Lisina/química , Modelos Moleculares , Poliubiquitina/química , Ligação Proteica , Domínios Proteicos , Engenharia de Proteínas , Multimerização Proteica , Proteínas Recombinantes de Fusão/genética , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem , Ubiquitinação
10.
Metallomics ; 8(2): 260, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26847676

RESUMO

Correction for 'Enantiopure titanocene complexes--direct evidence for paraptosis in cancer cells' by Melchior Cini et al., Metallomics, 2016, DOI: 10.1039/c5mt00297d.

11.
Metallomics ; 8(3): 286-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26806372

RESUMO

Tolerated by normal tissues, anti-cancer therapies based on titanium compounds are limited by low efficacy/selectivity and lack of understanding of their mode(s) of action. In vitro antitumour activity and mode of cell death incurred by enantiopure TiCl2{η-C5H4CHEt(2-MeOPh)}2 (abbreviated Cp(R)2TiCl2) has been investigated. The in vitro anti-tumour activity of Cp(R)2TiCl2 is selective for cancer cells; in clonogenic assays, (S,S)-Cp(R)2TiCl2 was twice as effective at inhibiting colony formation than other stereoisomers after 24 h exposure. HPLC, MS and NMR techniques determined hydrolysis of Cp(R)2TiCl2; data strongly correlate with soluble [Cp(R)2Ti(OH)(OH2)](+) being the biological trigger. Treatment of cells with Cp(R)2TiCl2 provoked extensive cytoplasmic vacuolization, endoplasmic reticulum (ER) swelling and activation of MAPKinase signal transduction, consistent with ligand-induced paraptosis, type III cell death, which is morphologically distinct from, and independent of apoptosis. Indeed, distinct from cisplatin, Cp(R)2TiCl2 failed to perturb cell cycle dynamics, induce γH2AX foci or evoke apoptosis in MDA-MB-468 and HCT-116 cells.


Assuntos
Apoptose/efeitos dos fármacos , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Citometria de Fluxo , Histonas/metabolismo , Humanos , Espectroscopia de Prótons por Ressonância Magnética , Estereoisomerismo
12.
Proteomics ; 15(5-6): 844-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25327553

RESUMO

Ubiquitin-binding domains (UBDs) are modular units found within ubiquitin-binding proteins that mediate the non-covalent recognition of (poly)ubiquitin modifications. A variety of mechanisms are employed in vivo to achieve polyubiquitin linkage and chain length selectivity by UBDs, the structural basis of which have in some instances been determined. Here, we review current knowledge related to ubiquitin recognition mechanisms at the molecular level and explore how such information has been exploited in the design and application of UBDs in isolation or artificially arranged in tandem as tools to investigate ubiquitin-modified proteomes. Specifically, we focus on the use of UBDs to directly purify or detect (poly)ubiquitin-modified proteins and more broadly for the targeted manipulation of ubiquitin-mediated processes, highlighting insights into ubiquitin signalling that have been provided.


Assuntos
Sítios de Ligação , Proteômica , Biologia Sintética , Ubiquitina , Animais , Linhagem Celular , Humanos , Camundongos , Estrutura Terciária de Proteína
13.
Chem Commun (Camb) ; 50(96): 15202-5, 2014 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-25338751

RESUMO

Biophysical studies of ligand interactions with three human telomeric repeat sequences (d(AGGG(TTAGGG)n, n = 3, 7 and 11)) show that an oxazole-based 'click' ligand, which induces parallel folded quadruplexes, preferentially stabilises longer telomeric repeats providing evidence for selectivity in binding at the interface between tandem quadruplex motifs.


Assuntos
Quadruplex G , Ligantes , Telômero/química , Sequência de Bases , Sítios de Ligação , Dicroísmo Circular , Química Click , Humanos , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico , Oxazóis/química
14.
Cell Rep ; 9(1): 193-205, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25263560

RESUMO

The use of oligonucleotides to activate the splicing of selected exons is limited by a poor understanding of the mechanisms affected. A targeted bifunctional oligonucleotide enhancer of splicing (TOES) anneals to SMN2 exon 7 and carries an exonic splicing enhancer (ESE) sequence. We show that it stimulates splicing specifically of intron 6 in the presence of repressing sequences in intron 7. Complementarity to the 5' end of exon 7 increases U2AF65 binding, but the ESE sequence is required for efficient recruitment of U2 snRNP. The ESE forms at least three coexisting discrete states: a quadruplex, a complex containing only hnRNP F/H, and a complex enriched in the activator SRSF1. Neither hnRNP H nor quadruplex formation contributes to ESE activity. The results suggest that splicing limited by weak signals can be rescued by rapid exchange of TOES oligonucleotides in various complexes and raise the possibility that SR proteins associate transiently with ESEs.


Assuntos
Éxons , Oligonucleotídeos/genética , Spliceossomos/genética , Sequência de Bases , Humanos , Íntrons , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oligonucleotídeos/metabolismo , Sítios de Splice de RNA , Splicing de RNA , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Spliceossomos/metabolismo , Fator de Processamento U2AF , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo
15.
Nucleic Acids Res ; 42(11): 6811-25, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24782516

RESUMO

CsrA/RsmA homologs are an extensive family of ribonucleic acid (RNA)-binding proteins that function as global post-transcriptional regulators controlling important cellular processes such as secondary metabolism, motility, biofilm formation and the production and secretion of virulence factors in diverse bacterial species. While direct messenger RNA binding by CsrA/RsmA has been studied in detail for some genes, it is anticipated that there are numerous additional, as yet undiscovered, direct targets that mediate its global regulation. To assist in the discovery of these targets, we propose a sequence-based approach to predict genes directly regulated by these regulators. In this work, we develop a computer code (CSRA_TARGET) implementing this approach, which leads to predictions for several novel targets in Escherichia coli and Pseudomonas aeruginosa. The predicted targets in other bacteria, specifically Salmonella enterica serovar Typhimurium, Pectobacterium carotovorum and Legionella pneumophila, also include global regulators that control virulence in these pathogens, unraveling intricate indirect regulatory roles for CsrA/RsmA. We have experimentally validated four predicted RsmA targets in P. aeruginosa. The sequence-based approach developed in this work can thus lead to several testable predictions for direct targets of CsrA homologs, thereby complementing and accelerating efforts to unravel global regulation by this important family of proteins.


Assuntos
Algoritmos , Pseudomonas aeruginosa/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Análise de Sequência de RNA/métodos , Sítios de Ligação , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Pseudomonas aeruginosa/metabolismo , RNA Mensageiro/química
16.
Biochemistry ; 53(14): 2371-9, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24665925

RESUMO

Evolution has produced proteins with exquisite ligand binding specificity, and manipulating this effect has been the basis for much of modern rational drug design. However, there are general classes of proteins with broader ligand selectivity linked to function, the origin of which is poorly understood. The odorant binding proteins (OBPs) sequester volatile molecules for transportation to the olfactory receptors. Rat OBP3, which we characterize by X-ray crystallography and NMR, binds a homologous series of aliphatic γ-lactones within its aromatic-rich hydrophobic pocket with remarkably little variation in affinity but extensive enthalpy/entropy compensation effects. We show that the binding energetics are modulated by two desolvation processes with quite different thermodynamic signatures. Ligand desolvation follows the classical hydrophobic effect; however, cavity desolvation is consistent with the liberation of "high energy" water molecules back into bulk solvent with a strong, but compensated, enthalpic contribution, which together underpin the origins of broad ligand binding selectivity.


Assuntos
Receptores Odorantes/metabolismo , Solventes/química , Termodinâmica , Animais , Cristalografia por Raios X , Lactonas/metabolismo , Ligantes , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Ratos , Receptores Odorantes/química
17.
Biochim Biophys Acta ; 1842(7): 992-1000, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24642144

RESUMO

SQSTM1 mutations are common in patients with Paget disease of bone (PDB), with most affecting the C-terminal ubiquitin-associated (UBA) domain of the SQSTM1 protein. We performed structural and functional analyses of two UBA domain mutations, an I424S mutation relatively common in UK PDB patients, and an A427D mutation associated with a severe phenotype in Southern Italian patients. Both impaired SQSTM1's ubiquitin-binding function in pull-down assays and resulted in activation of basal NF-κB signalling, compared to wild-type, in reporter assays. We found evidence for a relationship between the ability of different UBA domain mutants to activate NF-κB signalling in vitro and number of affected sites in vivo in 1152 PDB patients from the UK and Italy, with A427D-SQSTM1 producing the greatest level of activation (relative to wild-type) of all PDB mutants tested to date. NMR and isothermal titration calorimetry studies were able to demonstrate that I424S is associated with global structural changes in the UBA domain, resulting in 10-fold weaker UBA dimer stability than wild-type and reduced ubiquitin-binding affinity of the UBA monomer. Our observations provide insights into the role of SQSTM1-mediated NF-κB signalling in PDB aetiology, and demonstrate that different mutations in close proximity within loop 2/helix 3 of the SQSTM1 UBA domain exert distinct effects on protein structure and stability, including indirect effects at the UBA/ubiquitin-binding interface.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Osteíte Deformante/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Linhagem Celular , Predisposição Genética para Doença , Células HEK293 , Humanos , Modelos Moleculares , Mutação , NF-kappa B/genética , NF-kappa B/metabolismo , Osteíte Deformante/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Sequestossoma-1 , Transdução de Sinais , Ubiquitina/genética , Ubiquitina/metabolismo
18.
Exp Cell Res ; 325(1): 27-37, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24486447

RESUMO

Paget disease of bone (PDB) is a skeletal disorder common in Western Europe but extremely rare in the Indian subcontinent and Far East. The condition has a strong genetic element with mutations affecting the SQSTM1 gene, encoding the p62 protein, frequently identified. Recently SQSTM1 mutations have also been reported in a small number of patients with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), neurodegenerative disorders in which significant coexistence with PDB has not been previously recognized. Although several SQSTM1 mutations are common to both ALS/FTLD and PDB, many are ALS/FTLD-specific. The p62 protein regulates various cellular processes including NF-κB signaling and autophagy pathways. Here we consider how knowledge of the impact of PDB-associated SQSTM1 mutations (several of which are now known to be relevant for ALS/FTLD) on these pathways, as well as the locations of the mutations within the p62 primary sequence, may provide new insights into ALS/FTLD disease mechanisms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Esclerose Lateral Amiotrófica/genética , Degeneração Lobar Frontotemporal/genética , Osteíte Deformante/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Autofagia , Predisposição Genética para Doença , Humanos , NF-kappa B/metabolismo , Proteína Sequestossoma-1 , Transdução de Sinais
19.
J Exp Clin Cancer Res ; 32: 68, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24330541

RESUMO

Quadruplexes DNA are present in telomeric DNA as well as in several cancer-related gene promoters and hence affect gene expression and subsequent biological processes. The conformations of G4 provide selective recognition sites for small molecules and thus these structures have become important drug-design targets for cancer treatment. The DNA G-quadruplex binding pentacyclic acridinium salt RHPS4 (1) has many pharmacological attributes of an ideal telomere-targeting agent but has undesirable off-target liabilities. Notably a cardiovascular effect was evident in a guinea pig model, manifested by a marked and sustained increase in QTcB interval. In accordance with this, significant interaction with the human recombinant ß2 adrenergic receptor, and M1, M2 and M3 muscarinic receptors was observed, together with a high inhibition of the hERG tail current tested in a patch clamp assay. Two related pentacyclic structures, the acetylamines (2) and (3), both show a modest interaction with ß2 adrenergic receptor, and do not significatively inhibit the hERG tail current while demonstrating potent telomere on-target properties comparing closely with 1. Of the two isomers, the 2-acetyl-aminopentacycle (2) more closely mimics the overall biological profile of 1 and this information will be used to guide further synthetic efforts to identify novel variants of this chemotype, to maximize on-target and minimize off-target activities. Consequently, the improvement of toxicological profile of these compounds could therefore lead to the obtainment of suitable molecules for clinical development offering new pharmacological strategies in cancer treatment.


Assuntos
Acridinas/química , Acridinas/farmacologia , Quadruplex G , Telômero/metabolismo , Acridinas/síntese química , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cobaias , Humanos , Ligantes , Telomerase/antagonistas & inibidores
20.
Structure ; 21(9): 1659-71, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23954502

RESUMO

In bacteria, the highly conserved RsmA/CsrA family of RNA-binding proteins functions as global posttranscriptional regulators acting on mRNA translation and stability. Through phenotypic complementation of an rsmA mutant in Pseudomonas aeruginosa, we discovered a family member, termed RsmN. Elucidation of the RsmN crystal structure and that of the complex with a hairpin from the sRNA, RsmZ, reveals a uniquely inserted α helix, which redirects the polypeptide chain to form a distinctly different protein fold to the domain-swapped dimeric structure of RsmA homologs. The overall ß sheet structure required for RNA recognition is, however, preserved with compensatory sequence and structure differences, allowing the RsmN dimer to target binding motifs in both structured hairpin loops and flexible disordered RNAs. Phylogenetic analysis indicates that, although RsmN appears unique to P. aeruginosa, homologous proteins with the inserted α helix are more widespread and arose as a consequence of a gene duplication event.


Assuntos
Proteínas de Bactérias/química , Pseudomonas aeruginosa , Proteínas de Ligação a RNA/química , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Cristalografia por Raios X , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Sequências Repetidas Invertidas , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Bacteriano/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA