Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(6): e0274723, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37352301

RESUMO

Class II Fructose-1,6-bisphosphatases (FBPaseII) (EC: 3.1.3.11) are highly conserved essential enzymes in the gluconeogenic pathway of microorganisms. Previous crystallographic studies of FBPasesII provided insights into various inactivated states of the enzyme in different species. Presented here is the first crystal structure of FBPaseII in an active state, solved for the enzyme from Francisella tularensis (FtFBPaseII), containing native metal cofactor Mn2+ and complexed with catalytic product fructose-6-phosphate (F6P). Another crystal structure of the same enzyme complex is presented in the inactivated state due to the structural changes introduced by crystal packing. Analysis of the interatomic distances among the substrate, product, and divalent metal cations in the catalytic centers of the enzyme led to a revision of the catalytic mechanism suggested previously for class II FBPases. We propose that phosphate-1 is cleaved from the substrate fructose-1,6-bisphosphate (F1,6BP) by T89 in a proximal α-helix backbone (G88-T89-T90-I91-T92-S93-K94) in which the substrate transition state is stabilized by the positive dipole of the 〈-helix backbone. Once cleaved a water molecule found in the active site liberates the inorganic phosphate from T89 completing the catalytic mechanism. Additionally, a crystal structure of Mycobacterium tuberculosis FBPaseII (MtFBPaseII) containing a bound F1,6BP is presented to further support the substrate binding and novel catalytic mechanism suggested for this class of enzymes.


Assuntos
Francisella tularensis , Frutose-Bifosfatase , Frutose-Bifosfatase/metabolismo , Francisella tularensis/metabolismo , Catálise , Domínio Catalítico , Frutose/metabolismo , Cristalografia por Raios X
2.
Front Pharmacol ; 13: 970234, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36160429

RESUMO

The expanding roles of macrophages in physiological and pathophysiological mechanisms now include normal tissue homeostasis, tissue repair and regeneration, including neuronal tissue; initiation, progression, and resolution of the inflammatory response and a diverse array of anti-microbial activities. Two hallmarks of macrophage activity which appear to be fundamental to their diverse cellular functionalities are cellular plasticity and phenotypic heterogeneity. Macrophage plasticity allows these cells to take on a broad spectrum of differing cellular phenotypes in response to local and possibly previous encountered environmental signals. Cellular plasticity also contributes to tissue- and stimulus-dependent macrophage heterogeneity, which manifests itself as different macrophage phenotypes being found at different tissue locations and/or after different cell stimuli. Together, plasticity and heterogeneity align macrophage phenotypes to their required local cellular functions and prevent inappropriate activation of the cell, which could lead to pathology. To execute the appropriate function, which must be regulated at the qualitative, quantitative, spatial and temporal levels, macrophages constantly monitor intracellular and extracellular parameters to initiate and control the appropriate cell signaling cascades. The sensors and signaling mechanisms which control macrophages are the focus of a considerable amount of research. Ion channels regulate the flow of ions between cellular membranes and are critical to cell signaling mechanisms in a variety of cellular functions. It is therefore surprising that the role of ion channels in the macrophage biology has been relatively overlooked. In this review we provide a summary of ion channel research in macrophages. We begin by giving a narrative-based explanation of the membrane potential and its importance in cell biology. We then report on research implicating different ion channel families in macrophage functions. Finally, we highlight some areas of ion channel research in macrophages which need to be addressed, future possible developments in this field and therapeutic potential.

3.
Front Immunol ; 13: 840069, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757775

RESUMO

The traditional view of the nuclear envelope (NE) was that it represented a relatively inert physical barrier within the cell, whose main purpose was to separate the nucleoplasm from the cytoplasm. However, recent research suggests that this is far from the case, with new and important cellular functions being attributed to this organelle. In this review we describe research suggesting an important contribution of the NE and its constituents in regulating the functions of cells of the innate and adaptive immune system. One of the standout properties of immune cells is their ability to migrate around the body, allowing them to carry out their physiological/pathophysiology cellular role at the appropriate location. This together with the physiological role of the tissue, changes in tissue matrix composition due to disease and aging, and the activation status of the immune cell, all result in immune cells being subjected to different mechanical forces. We report research which suggests that the NE may be an important sensor/transducer of these mechanical signals and propose that the NE is an integrator of both mechanical and chemical signals, allowing the cells of the innate immune system to precisely regulate gene transcription and functionality. By presenting this overview we hope to stimulate the interests of researchers into this often-overlooked organelle and propose it should join the ranks of mitochondria and phagosome, which are important organelles contributing to immune cell function.


Assuntos
Núcleo Celular , Membrana Nuclear , Núcleo Celular/genética , Citoplasma
4.
Pharmacol Rep ; 73(3): 881-890, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33713314

RESUMO

BACKGROUND: Macrophages are important cells of the innate immune system and contribute to a variety of physiological and pathophysiological responses. Monovalent and divalent ion channels have been studied in macrophage function, and while much research is still required, a role for these channels is beginning to emerge in macrophages. In addition to the plasma membrane, ion channels are also found in intracellular membranes including mitochondrial, lysosomal and nuclear membranes. While studying the function of plasma membrane located large conductance voltage- and calcium-activated potassium channels (BK channels) in a macrophage cell line RAW264.7, we became aware of the expression of these ion channels in other cellular locations. METHODS: Immunofluorescence and Western blot analysis were used to identify the expression of BK channels. To demonstrate a functional role for the nuclear located channel, we investigated the effect of the lipid soluble BK channel inhibitor paxilline on CREB phosphorylation. RESULTS: Treatment of resting macrophages with paxilline resulted in increased CREB phosphorylation. To confirm a role for nuclear BK channels, these experiments were repeated in isolated nuclei and similar results were found. Ca2+ and calmodulin-dependent kinases (CaMK) have been demonstrated to regulate CREB phosphorylation. Inhibition of CaMKII and CaMKIV resulted in the reversal of paxilline-induced CREB phosphorylation. CONCLUSIONS: These results suggest that nuclear BK channels regulate CREB phosphorylation in macrophages. Nuclear located ion channels may therefore be part of novel signalling pathways in macrophages and should be taken into account when studying the role of ion channels in these and other cells.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Macrófagos/metabolismo , Fosforilação/fisiologia , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Indóis/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Membrana Nuclear/metabolismo , Fosforilação/efeitos dos fármacos , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
5.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 11): 524-535, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-33135671

RESUMO

The crystal structure of the class II fructose-1,6-bisphosphatase (FBPaseII) from the important pathogen Francisella tularensis is presented at 2.4 Šresolution. Its structural and functional relationships to the closely related phosphatases from Mycobacterium tuberculosis (MtFBPaseII) and Escherichia coli (EcFBPaseII) and to the dual phosphatase from Synechocystis strain 6803 are discussed. FBPaseII from F. tularensis (FtFBPaseII) was crystallized in a monoclinic crystal form (space group P21, unit-cell parameters a = 76.30, b = 100.17, c = 92.02 Å, ß = 90.003°) with four chains in the asymmetric unit. Chain A had two coordinated Mg2+ ions in its active center, which is distinct from previous findings, and is presumably deactivated by their presence. The structure revealed an approximate 222 (D2) symmetry homotetramer analogous to that previously described for MtFBPaseII, which is formed by a crystallographic dyad and which differs from the exact tetramer found in EcFBPaseII at a 222 symmetry site in the crystal. Instead, the approximate homotetramer is very similar to that found in the dual phosphatase from Synechocystis, even though no allosteric effector was found in FtFBPase. The amino-acid sequence and folding of the active site of FtFBPaseII result in structural characteristics that are more similar to those of the previously published EcFBPaseII than to those of MtFBPaseII. The kinetic parameters of native FtFBPaseII were found to be in agreement with published studies. Kinetic analyses of the Thr89Ser and Thr89Ala mutations in the active site of the enzyme are consistent with the previously proposed mechanism for other class II bisphosphatases. The Thr89Ala variant enzyme was inactive but the Thr89Ser variant was partially active, with an approximately fourfold lower Km and Vmax than the native enzyme. The structural and functional insights derived from the structure of FtFBPaseII will provide valuable information for the design of specific inhibitors.


Assuntos
Francisella tularensis/enzimologia , Frutose-Bifosfatase/química , Frutose-Bifosfatase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/enzimologia , Frutose-Bifosfatase/genética , Frutose-Bifosfatase/isolamento & purificação , Modelos Moleculares , Mycobacterium tuberculosis/enzimologia , Conformação Proteica , Estrutura Quaternária de Proteína , Synechocystis/enzimologia
6.
J Mol Biol ; 425(18): 3301-10, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-23806658

RESUMO

IRP1 [iron regulatory protein (IRP) 1] is a bifunctional protein with mutually exclusive end-states. In one mode of operation, IRP1 binds iron-responsive element (IRE) stem-loops in messenger RNAs encoding proteins of iron metabolism to control their rate of translation. In its other mode, IRP1 serves as cytoplasmic aconitase to correlate iron availability with the energy and oxidative stress status of the cell. IRP1/IRE binding occurs through two separate interfaces, which together contribute about two-dozen hydrogen bonds. Five amino acids make base-specific contacts and are expected to contribute significantly to binding affinity and specificity of this protein:RNA interaction. In this mutagenesis study, each of the five base-specific amino acids was changed to alter binding at each site. Analysis of IRE binding affinity and translational repression activity of the resulting IRP1 mutants showed that four of the five contact points contribute uniquely to the overall binding affinity of the IRP1:IRE interaction, while one site was found to be unimportant. The stronger-than-expected effect on binding affinity of mutations at Lys379 and Ser681, residues that make contact with the conserved nucleotides G16 and C8, respectively, identified them as particularly critical for providing specificity and stability to IRP1:IRE complex formation. We also show that even though the base-specific RNA-binding residues are not part of the aconitase active site, their substitutions can affect the aconitase activity of holo-IRP1, positively or negatively.


Assuntos
Proteína 1 Reguladora do Ferro/metabolismo , Ferro/farmacologia , Nucleotídeos/metabolismo , Elementos de Resposta/genética , Animais , Sítios de Ligação/genética , Domínio Catalítico/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína 1 Reguladora do Ferro/química , Proteína 1 Reguladora do Ferro/genética , Proteína 1 Reguladora do Ferro/fisiologia , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação de Ácido Nucleico , Nucleotídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Saccharomyces cerevisiae , Especificidade por Substrato
7.
FEBS Lett ; 586(1): 32-5, 2012 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-22119729

RESUMO

Iron responsive elements (IREs) are short stem-loop structures found in several mRNAs encoding proteins involved in cellular iron metabolism. Iron regulatory proteins (IRPs) control iron homeostasis through differential binding to the IREs, accommodating any sequence or structural variations that the IREs may present. Here we report the structure of IRP1 in complex with transferrin receptor 1 B (TfR B) IRE, and compare it to the complex with ferritin H (Ftn H) IRE. The two IREs are bound to IRP1 through nearly identical protein-RNA contacts, although their stem conformations are significantly different. These results support the view that binding of different IREs with IRP1 depends both on protein and RNA conformational plasticity, adapting to RNA variation while retaining conserved protein-RNA contacts.


Assuntos
Proteína 1 Reguladora do Ferro/química , Proteína 1 Reguladora do Ferro/metabolismo , Ferro , Receptores da Transferrina/química , Receptores da Transferrina/metabolismo , Elementos de Resposta , Animais , Cristalografia por Raios X , Conformação de Ácido Nucleico , Conformação Proteica , Coelhos
8.
J Mol Biol ; 400(2): 145-54, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20460131

RESUMO

The nuclear protein cyclophilin 33 (Cyp33) is a peptidyl-prolyl cis-trans isomerase that catalyzes cis-trans isomerization of the peptide bond preceding a proline and promotes folding and conformational changes in folded and unfolded proteins. The N-terminal RNA-recognition motif (RRM) domain of Cyp33 has been found to associate with the third plant homeodomain (PHD3) finger of the mixed lineage leukemia (MLL) proto-oncoprotein and a poly(A) RNA sequence. Here, we report a 1.9 A resolution crystal structure of the RRM domain of Cyp33 and describe the molecular mechanism of PHD3 and RNA recognition. The Cyp33 RRM domain folds into a five-stranded antiparallel beta-sheet and two alpha-helices. The RRM domain, but not the catalytic module of Cyp33, binds strongly to PHD3, exhibiting a 2 muM affinity as measured by isothermal titration calorimetry. NMR chemical shift perturbation (CSP) analysis and dynamics data reveal that the beta strands and the beta2-beta3 loop of the RRM domain are involved in the interaction with PHD3. Mutations in the PHD3-binding site or deletions in the beta2-beta3 loop lead to a significantly reduced affinity or abrogation of the interaction. The RNA-binding pocket of the Cyp33 RRM domain, mapped on the basis of NMR CSP and mutagenesis, partially overlaps with the PHD3-binding site, and RNA association is abolished in the presence of MLL PHD3. Full-length Cyp33 acts as a negative regulator of MLL-induced transcription and reduces the expression levels of MLL target genes MEIS1 and HOXA9. Together, these in vitro and in vivo data provide insight into the multiple functions of Cyp33 RRM and suggest a Cyp33-dependent mechanism for regulating the transcriptional activity of MLL.


Assuntos
Ciclofilinas/química , Proteínas de Drosophila/química , Proteína de Leucina Linfoide-Mieloide/química , Proteína de Leucina Linfoide-Mieloide/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Ciclofilinas/genética , Ciclofilinas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteína de Leucina Linfoide-Mieloide/genética , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , RNA/química , RNA/metabolismo , Alinhamento de Sequência , Transcrição Gênica
9.
Science ; 314(5807): 1903-8, 2006 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-17185597

RESUMO

Iron regulatory protein 1 (IRP1) binds iron-responsive elements (IREs) in messenger RNAs (mRNAs), to repress translation or degradation, or binds an iron-sulfur cluster, to become a cytosolic aconitase enzyme. The 2.8 angstrom resolution crystal structure of the IRP1:ferritin H IRE complex shows an open protein conformation compared with that of cytosolic aconitase. The extended, L-shaped IRP1 molecule embraces the IRE stem-loop through interactions at two sites separated by approximately 30 angstroms, each involving about a dozen protein:RNA bonds. Extensive conformational changes related to binding the IRE or an iron-sulfur cluster explain the alternate functions of IRP1 as an mRNA regulator or enzyme.


Assuntos
Apoferritinas/genética , Proteína 1 Reguladora do Ferro/química , Proteína 1 Reguladora do Ferro/metabolismo , Sequências Reguladoras de Ácido Ribonucleico , Elementos de Resposta , Regiões não Traduzidas/química , Regiões não Traduzidas/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Ligação de Hidrogênio , Ferro/metabolismo , Modelos Moleculares , Conformação de Ácido Nucleico , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Enxofre/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-16511314

RESUMO

Iron regulatory protein 1 (IRP1) is a bifunctional protein with activity as an RNA-binding protein or as a cytoplasmic aconitase. Interconversion of IRP1 between these mutually exclusive states is central to cellular iron regulation and is accomplished through iron-responsive assembly and disassembly of a [4Fe-4S] cluster. When in its apo form, IRP1 binds to iron responsive elements (IREs) found in mRNAs encoding proteins of iron storage and transport and either prevents translation or degradation of the bound mRNA. Excess cellular iron stimulates the assembly of a [4Fe-4S] cluster in IRP1, inhibiting its IRE-binding ability and converting it to an aconitase. The three-dimensional structure of IRP1 in its different active forms will provide details of the interconversion process and clarify the selective recognition of mRNA, Fe-S sites and catalytic activity. To this end, the apo form of IRP1 bound to a ferritin IRE was crystallized. Crystals belong to the monoclinic space group P2(1), with unit-cell parameters a = 109.6, b = 80.9, c = 142.9 A, beta = 92.0 degrees. Native data sets have been collected from several crystals with resolution extending to 2.8 A and the structure has been solved by molecular replacement.


Assuntos
Ferritinas/química , Proteína 1 Reguladora do Ferro/química , Proteínas Reguladoras de Ferro/química , Animais , Apoproteínas/química , Cristalização/métodos , Cristalografia por Raios X , Ferritinas/genética , Proteínas Reguladoras de Ferro/genética , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...