Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Circ Res ; 132(11): e188-e205, 2023 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-37139790

RESUMO

BACKGROUND: Transverse tubules (t-tubules) form gradually in the developing heart, critically enabling maturation of cardiomyocyte Ca2+ homeostasis. The membrane bending and scaffolding protein BIN1 (bridging integrator 1) has been implicated in this process. However, it is unclear which of the various reported BIN1 isoforms are involved, and whether BIN1 function is regulated by its putative binding partners MTM1 (myotubularin), a phosphoinositide 3'-phosphatase, and DNM2 (dynamin-2), a GTPase believed to mediate membrane fission. METHODS: We investigated the roles of BIN1, MTM1, and DNM2 in t-tubule formation in developing mouse cardiomyocytes, and in gene-modified HL-1 and human-induced pluripotent stem cell-derived cardiomyocytes. T-tubules and proteins of interest were imaged by confocal and Airyscan microscopy, and expression patterns were examined by RT-qPCR and Western blotting. Ca2+ release was recorded using Fluo-4. RESULTS: We observed that in the postnatal mouse heart, BIN1 localizes along Z-lines from early developmental stages, consistent with roles in initial budding and scaffolding of t-tubules. T-tubule proliferation and organization were linked to a progressive and parallel increase in 4 detected BIN1 isoforms. All isoforms were observed to induce tubulation in cardiomyocytes but produced t-tubules with differing geometries. BIN1-induced tubulations contained the L-type Ca2+ channel, were colocalized with caveolin-3 and the ryanodine receptor, and effectively triggered Ca2+ release. BIN1 upregulation during development was paralleled by increasing expression of MTM1. Despite no direct binding between MTM1 and murine cardiac BIN1 isoforms, which lack exon 11, high MTM1 levels were necessary for BIN1-induced tubulation, indicating a central role of phosphoinositide homeostasis. In contrast, the developing heart exhibited declining levels of DNM2. Indeed, we observed that high levels of DNM2 are inhibitory for t-tubule formation, although this protein colocalizes with BIN1 along Z-lines, and binds all 4 isoforms. CONCLUSIONS: These findings indicate that BIN1, MTM1, and DNM2 have balanced and collaborative roles in controlling t-tubule growth in cardiomyocytes.


Assuntos
Dinamina II , Miócitos Cardíacos , Animais , Humanos , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dinamina II/genética , Dinamina II/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas Supressoras de Tumor/metabolismo
2.
Nat Rev Mol Cell Biol ; 12(1): 60-70, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21179061

RESUMO

Plasma membrane ATPases are primary active transporters of cations that maintain steep concentration gradients. The ion gradients and membrane potentials derived from them form the basis for a range of essential cellular processes, in particular Na(+)-dependent and proton-dependent secondary transport systems that are responsible for uptake and extrusion of metabolites and other ions. The ion gradients are also both directly and indirectly used to control pH homeostasis and to regulate cell volume. The plasma membrane H(+)-ATPase maintains a proton gradient in plants and fungi and the Na(+),K(+)-ATPase maintains a Na(+) and K(+) gradient in animal cells. Structural information provides insight into the function of these two distinct but related P-type pumps.


Assuntos
Membrana Celular/metabolismo , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Membrana Celular/química , Humanos
3.
J Biol Chem ; 295(6): 1454-1463, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31848226

RESUMO

Cellobiohydrolases effectively degrade cellulose and are of biotechnological interest because they can convert lignocellulosic biomass to fermentable sugars. Here, we implemented a fluorescence-based method for real-time measurements of complexation and decomplexation of the processive cellulase Cel7A and its insoluble substrate, cellulose. The method enabled detailed kinetic and thermodynamic analyses of ligand binding in a heterogeneous system. We studied WT Cel7A and several variants in which one or two of four highly conserved Trp residues in the binding tunnel had been replaced with Ala. WT Cel7A had on/off-rate constants of 1 × 105 m-1 s-1 and 5 × 10-3 s-1, respectively, reflecting the slow dynamics of a solid, polymeric ligand. Especially the off-rate constant was many orders of magnitude lower than typical values for small, soluble ligands. Binding rate and strength both were typically lower for the Trp variants, but effects of the substitutions were moderate and sometimes negligible. Hence, we propose that lowering the activation barrier for complexation is not a major driving force for the high conservation of the Trp residues. Using so-called Φ-factor analysis, we analyzed the kinetic and thermodynamic results for the variants. The results of this analysis suggested a transition state for complexation and decomplexation in which the reducing end of the ligand is close to the tunnel entrance (near Trp-40), whereas the rest of the binding tunnel is empty. We propose that this structure defines the highest free-energy barrier of the overall catalytic cycle and hence governs the turnover rate of this industrially important enzyme.


Assuntos
Celulase/metabolismo , Celulose/metabolismo , Proteínas Fúngicas/metabolismo , Trichoderma/metabolismo , Triptofano/metabolismo , Domínio Catalítico , Celulase/química , Ativação Enzimática , Proteínas Fúngicas/química , Cinética , Modelos Moleculares , Ligação Proteica , Especificidade por Substrato , Termodinâmica , Trichoderma/química , Triptofano/química
4.
J Biol Chem ; 291(27): 14340-14355, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27189939

RESUMO

Hepatocyte growth factor activator inhibitor-1 (HAI-1) is a type I transmembrane protein and inhibitor of several serine proteases, including hepatocyte growth factor activator and matriptase. The protein is essential for development as knock-out mice die in utero due to placental defects caused by misregulated extracellular proteolysis. HAI-1 contains two Kunitz-type inhibitor domains (Kunitz), which are generally thought of as a functionally self-contained protease inhibitor unit. This is not the case for HAI-1, where our results reveal how interdomain interactions have evolved to stimulate the inhibitory activity of an integrated Kunitz. Here we present an x-ray crystal structure of an HAI-1 fragment covering the internal domain and Kunitz-1. The structure reveals not only that the previously uncharacterized internal domain is a member of the polycystic kidney disease domain family but also how the two domains engage in interdomain interactions. Supported by solution small angle x-ray scattering and a combination of site-directed mutagenesis and functional assays, we show that interdomain interactions not only stabilize the fold of the internal domain but also stimulate the inhibitory activity of Kunitz-1. By completing our structural characterization of the previously unknown N-terminal region of HAI-1, we provide new insight into the interplay between tertiary structure and the inhibitory activity of a multidomain protease inhibitor. We propose a previously unseen mechanism by which the association of an auxiliary domain stimulates the inhibitory activity of a Kunitz-type inhibitor (i.e. the first structure of an intramolecular interaction between a Kunitz and another domain).


Assuntos
Glicoproteínas de Membrana/metabolismo , Doenças Renais Policísticas/metabolismo , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Glicoproteínas de Membrana/química , Camundongos , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Proteínas Secretadas Inibidoras de Proteinases , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
5.
J Biol Chem ; 291(38): 20096-112, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27466361

RESUMO

Intimin is an essential adhesin of attaching and effacing organisms such as entropathogenic Escherichia coli It is also the prototype of type Ve secretion or inverse autotransport, where the extracellular C-terminal region or passenger is exported with the help of an N-terminal transmembrane ß-barrel domain. We recently reported a stalled secretion intermediate of intimin, where the passenger is located in the periplasm but the ß-barrel is already inserted into the membrane. Stalling of this mutant is due to the insertion of an epitope tag at the very N terminus of the passenger. Here, we examined how this insertion disrupts autotransport and found that it causes misfolding of the N-terminal immunoglobulin (Ig)-like domain D00. We could also stall the secretion by making an internal deletion in D00, and introducing the epitope tag into the second Ig-like domain, D0, also resulted in reduced passenger secretion. In contrast to many classical autotransporters, where a proximal folding core in the passenger is required for secretion, the D00 domain is dispensable, as the passenger of an intimin mutant lacking D00 entirely is efficiently exported. Furthermore, the D00 domain is slightly less stable than the D0 and D1 domains, unfolding at ∼200 piconewtons (pN) compared with ∼250 pN for D0 and D1 domains as measured by atomic force microscopy. Our results support a model where the secretion of the passenger is driven by sequential folding of the extracellular Ig-like domains, leading to vectorial transport of the passenger domain across the outer membrane in an N to C direction.


Assuntos
Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Modelos Biológicos , Dobramento de Proteína , Adesinas Bacterianas/genética , Escherichia coli Enteropatogênica/genética , Proteínas de Escherichia coli/genética , Domínios Proteicos
6.
Nature ; 475(7354): 59-64, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21716286

RESUMO

Heavy-metal homeostasis and detoxification is crucial for cell viability. P-type ATPases of the class IB (PIB) are essential in these processes, actively extruding heavy metals from the cytoplasm of cells. Here we present the structure of a PIB-ATPase, a Legionella pneumophila CopA Cu(+)-ATPase, in a copper-free form, as determined by X-ray crystallography at 3.2 Å resolution. The structure indicates a three-stage copper transport pathway involving several conserved residues. A PIB-specific transmembrane helix kinks at a double-glycine motif displaying an amphipathic helix that lines a putative copper entry point at the intracellular interface. Comparisons to Ca(2+)-ATPase suggest an ATPase-coupled copper release mechanism from the binding sites in the membrane via an extracellular exit site. The structure also provides a framework to analyse missense mutations in the human ATP7A and ATP7B proteins associated with Menkes' and Wilson's diseases.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cobre/metabolismo , Legionella pneumophila/química , Adenosina Trifosfatases/genética , Sítios de Ligação , Transporte Biológico , Cálcio , Proteínas de Transporte de Cátions/genética , Membrana Celular/metabolismo , ATPases Transportadoras de Cobre , Cristalografia por Raios X , Citoplasma/metabolismo , Degeneração Hepatolenticular/genética , Humanos , Síndrome dos Cabelos Torcidos/genética , Modelos Moleculares , Mutação de Sentido Incorreto/genética , Estrutura Terciária de Proteína , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , Relação Estrutura-Atividade
7.
Nature ; 467(7311): 99-102, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20720542

RESUMO

The Na(+)/K(+)-ATPase pumps three sodium ions out of and two potassium ions into the cell for each ATP molecule that is split, thereby generating the chemical and electrical gradients across the plasma membrane that are essential in, for example, signalling, secondary transport and volume regulation in animal cells. Crystal structures of the potassium-bound form of the pump revealed an intimate docking of the alpha-subunit carboxy terminus at the transmembrane domain. Here we show that this element is a key regulator of a previously unrecognized ion pathway. Current models of P-type ATPases operate with a single ion conduit through the pump, but our data suggest an additional pathway in the Na(+)/K(+)-ATPase between the ion-binding sites and the cytoplasm. The C-terminal pathway allows a cytoplasmic proton to enter and stabilize site III when empty in the potassium-bound state, and when potassium is released the proton will also return to the cytoplasm, thus allowing an overall asymmetric stoichiometry of the transported ions. The C terminus controls the gate to the pathway. Its structure is crucial for pump function, as demonstrated by at least eight mutations in the region that cause severe neurological diseases. This novel model for ion transport by the Na(+)/K(+)-ATPase is established by electrophysiological studies of C-terminal mutations in familial hemiplegic migraine 2 (FHM2) and is further substantiated by molecular dynamics simulations. A similar ion regulation is likely to apply to the H(+)/K(+)-ATPase and the Ca(2+)-ATPase.


Assuntos
Transporte de Íons , Enxaqueca com Aura/metabolismo , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Cristalografia por Raios X , Humanos , Enxaqueca com Aura/genética , Modelos Moleculares , Simulação de Dinâmica Molecular , Oócitos/metabolismo , Potássio/metabolismo , Prótons , Squalus acanthias/metabolismo , Sus scrofa/metabolismo , Xenopus
8.
J Biol Chem ; 289(31): 21351-9, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24917679

RESUMO

The high resolution crystal structures of isatin hydrolase from Labrenzia aggregata in the apo and the product state are described. These are the first structures of a functionally characterized metal-dependent hydrolase of this fold. Isatin hydrolase converts isatin to isatinate and belongs to a novel family of metalloenzymes that include the bacterial kynurenine formamidase. The product state, mimicked by bound thioisatinate, reveals a water molecule that bridges the thioisatinate to a proton wire in an adjacent water channel and thus allows the proton released by the reaction to escape only when the product is formed. The functional proton wire present in isatin hydrolase isoform b represents a unique catalytic feature common to all hydrolases is here trapped and visualized for the first time. The local molecular environment required to coordinate thioisatinate allows stronger and more confident identification of orthologous genes encoding isatin hydrolases within the prokaryotic kingdom. The isatin hydrolase orthologues found in human gut bacteria raise the question as to whether the indole-3-acetic acid degradation pathway is present in human gut flora.


Assuntos
Aquaporinas/química , Hidrolases/química , Isatina/química , Sequência de Aminoácidos , Sequência de Bases , Domínio Catalítico , Dicroísmo Circular , Cristalografia por Raios X , Primers do DNA , Hidrolases/genética , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Prótons , Rhodobacteraceae/enzimologia , Homologia de Sequência de Aminoácidos
9.
J Biol Chem ; 288(15): 10759-65, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23400778

RESUMO

The sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) is a transmembrane ion transporter belonging to the P(II)-type ATPase family. It performs the vital task of re-sequestering cytoplasmic Ca(2+) to the sarco/endoplasmic reticulum store, thereby also terminating Ca(2+)-induced signaling such as in muscle contraction. This minireview focuses on the transport pathways of Ca(2+) and H(+) ions across the lipid bilayer through SERCA. The ion-binding sites of SERCA are accessible from either the cytoplasm or the sarco/endoplasmic reticulum lumen, and the Ca(2+) entry and exit channels are both formed mainly by rearrangements of four N-terminal transmembrane α-helices. Recent improvements in the resolution of the crystal structures of rabbit SERCA1a have revealed a hydrated pathway in the C-terminal transmembrane region leading from the ion-binding sites to the cytosol. A comparison of different SERCA conformations reveals that this C-terminal pathway is exclusive to Ca(2+)-free E2 states, suggesting that it may play a functional role in proton release from the ion-binding sites. This is in agreement with molecular dynamics simulations and mutational studies and is in striking analogy to a similar pathway recently described for the related sodium pump. We therefore suggest a model for the ion exchange mechanism in P(II)-ATPases including not one, but two cytoplasmic pathways working in concert.


Assuntos
Cálcio/metabolismo , Prótons , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Sítios de Ligação , Cálcio/química , Citosol/química , Citosol/metabolismo , Humanos , Transporte de Íons/fisiologia , Simulação de Dinâmica Molecular , Coelhos , Retículo Sarcoplasmático/química , Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética
10.
Nat Commun ; 15(1): 173, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38228619

RESUMO

Improved therapies are needed against snakebite envenoming, which kills and permanently disables thousands of people each year. Recently developed neutralizing monoclonal antibodies against several snake toxins have shown promise in preclinical rodent models. Here, we use phage display technology to discover a human monoclonal antibody and show that this antibody causes antibody-dependent enhancement of toxicity (ADET) of myotoxin II from the venomous pit viper, Bothrops asper, in a mouse model of envenoming that mimics a snakebite. While clinical ADET related to snake venom has not yet been reported in humans, this report of ADET of a toxin from the animal kingdom highlights the necessity of assessing even well-known antibody formats in representative preclinical models to evaluate their therapeutic utility against toxins or venoms. This is essential to avoid potential deleterious effects as exemplified in the present study.


Assuntos
Bothrops , Neurotoxinas , Camundongos , Animais , Humanos , Neurotoxinas/toxicidade , Bothrops asper , Anticorpos Facilitadores , Anticorpos Monoclonais/toxicidade
11.
Artigo em Inglês | MEDLINE | ID: mdl-23989163

RESUMO

High-quality crystals of Thermus thermophilus EF-Tu in the GTP-bound conformation at 1.7-2.7 Šresolution were used to test 18 small organic molecules, all brominated for confident identification in the anomalous difference maps. From this relatively small collection, it was possible to identify a small molecule bound in the functionally important tRNA CCA-end binding pocket. The antibiotic GE2270 A is known to interact with the same pocket in EF-Tu and to disrupt the association with tRNA. Bromide could be located from peaks in the anomalous map in data truncated to very low resolution without refining the structure. Considering the speed with which diffraction data can be collected today, it is proposed that it is worthwhile to collect the extra data from fragment screens while crystals are at hand to increase the knowledge of biological function and drug binding in an experimental structural context.


Assuntos
Proteínas de Bactérias/química , Guanosina Trifosfato/química , Fator Tu de Elongação de Peptídeos/química , RNA de Transferência/química , Bibliotecas de Moléculas Pequenas/química , Thermus thermophilus/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Sítios de Ligação , Cristalografia por Raios X , Escherichia coli/genética , Expressão Gênica , Halogenação , Ligantes , Espectrometria de Massas , Simulação de Acoplamento Molecular , Fator Tu de Elongação de Peptídeos/genética , Fator Tu de Elongação de Peptídeos/isolamento & purificação , Mapeamento de Peptídeos , Peptídeos Cíclicos/química , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Thermus thermophilus/genética , Thermus thermophilus/metabolismo , Tiazóis/química
12.
Nature ; 450(7172): 1111-4, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-18075595

RESUMO

A prerequisite for life is the ability to maintain electrochemical imbalances across biomembranes. In all eukaryotes the plasma membrane potential and secondary transport systems are energized by the activity of P-type ATPase membrane proteins: H+-ATPase (the proton pump) in plants and fungi, and Na+,K+-ATPase (the sodium-potassium pump) in animals. The name P-type derives from the fact that these proteins exploit a phosphorylated reaction cycle intermediate of ATP hydrolysis. The plasma membrane proton pumps belong to the type III P-type ATPase subfamily, whereas Na+,K+-ATPase and Ca2+-ATPase are type II. Electron microscopy has revealed the overall shape of proton pumps, however, an atomic structure has been lacking. Here we present the first structure of a P-type proton pump determined by X-ray crystallography. Ten transmembrane helices and three cytoplasmic domains define the functional unit of ATP-coupled proton transport across the plasma membrane, and the structure is locked in a functional state not previously observed in P-type ATPases. The transmembrane domain reveals a large cavity, which is likely to be filled with water, located near the middle of the membrane plane where it is lined by conserved hydrophilic and charged residues. Proton transport against a high membrane potential is readily explained by this structural arrangement.


Assuntos
Arabidopsis/enzimologia , Membrana Celular/enzimologia , Bombas de Próton/química , Sítios de Ligação , ATPases Transportadoras de Cálcio/química , ATPases Transportadoras de Cálcio/metabolismo , Cristalografia por Raios X , Transporte de Íons , Modelos Moleculares , Fosforilação , Bombas de Próton/metabolismo , Prótons , Eletricidade Estática , Homologia Estrutural de Proteína
13.
Nature ; 450(7172): 1036-42, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-18075584

RESUMO

The sarcoplasmic reticulum Ca2+-ATPase, a P-type ATPase, has a critical role in muscle function and metabolism. Here we present functional studies and three new crystal structures of the rabbit skeletal muscle Ca2+-ATPase, representing the phosphoenzyme intermediates associated with Ca2+ binding, Ca2+ translocation and dephosphorylation, that are based on complexes with a functional ATP analogue, beryllium fluoride and aluminium fluoride, respectively. The structures complete the cycle of nucleotide binding and cation transport of Ca2+-ATPase. Phosphorylation of the enzyme triggers the onset of a conformational change that leads to the opening of a luminal exit pathway defined by the transmembrane segments M1 through M6, which represent the canonical membrane domain of P-type pumps. Ca2+ release is promoted by translocation of the M4 helix, exposing Glu 309, Glu 771 and Asn 796 to the lumen. The mechanism explains how P-type ATPases are able to form the steep electrochemical gradients required for key functions in eukaryotic cells.


Assuntos
Cálcio/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Berílio , Cristalografia por Raios X , Fluoretos , Transporte de Íons , Espectrometria de Massas , Modelos Moleculares , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Fosforilação , Conformação Proteica , Coelhos , Relação Estrutura-Atividade , Tapsigargina
14.
Nature ; 450(7172): 1043-9, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-18075585

RESUMO

The Na+,K+-ATPase generates electrochemical gradients for sodium and potassium that are vital to animal cells, exchanging three sodium ions for two potassium ions across the plasma membrane during each cycle of ATP hydrolysis. Here we present the X-ray crystal structure at 3.5 A resolution of the pig renal Na+,K+-ATPase with two rubidium ions bound (as potassium congeners) in an occluded state in the transmembrane part of the alpha-subunit. Several of the residues forming the cavity for rubidium/potassium occlusion in the Na+,K+-ATPase are homologous to those binding calcium in the Ca2+-ATPase of sarco(endo)plasmic reticulum. The beta- and gamma-subunits specific to the Na+,K+-ATPase are associated with transmembrane helices alphaM7/alphaM10 and alphaM9, respectively. The gamma-subunit corresponds to a fragment of the V-type ATPase c subunit. The carboxy terminus of the alpha-subunit is contained within a pocket between transmembrane helices and seems to be a novel regulatory element controlling sodium affinity, possibly influenced by the membrane potential.


Assuntos
Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/metabolismo , Sódio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Cátions Monovalentes/metabolismo , Membrana Celular/metabolismo , Cristalização , Cristalografia por Raios X , Fluoretos , Rim/enzimologia , Compostos de Magnésio , Potenciais da Membrana , Modelos Moleculares , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Suínos
15.
Sci Rep ; 13(1): 14361, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37658186

RESUMO

The present study describes a detailed procedure for expressing and purifying the integral membrane protein RseP using the pSIP system and Lactiplantibacillus plantarum as an expression host. RseP is a membrane-bound site-2-protease and a known antibacterial target in multiple human pathogens. In the present study, we screened five RseP orthologs from Gram-positive bacteria and found RseP from Enterococcus faecium (EfmRseP) to yield the highest protein levels. The production conditions were optimized and EfmRseP was purified by immobilized metal ion affinity chromatography followed by size-exclusion chromatography. The purification resulted in an overall yield of approximately 1 mg of pure protein per 3 g of wet-weight cell pellet. The structural integrity of the purified protein was confirmed using circular dichroism. We further assessed the expression and purification of RseP from E. faecium in the Gram-negative Escherichia coli. Detection of soluble protein failed in two of the three E. coli strains tested. Purification of EfmRseP expressed in E. coli C43(DE3) resulted in a protein with lower purity compared to EfmRseP expressed in L. plantarum. To our knowledge, this is the first time L. plantarum and the pSIP expression system have been applied for the production of membrane proteins.


Assuntos
Proteínas de Escherichia coli , Proteínas de Membrana , Humanos , Proteínas de Membrana/genética , Benchmarking , Escherichia coli/genética , Antibacterianos , Cromatografia de Afinidade , Endopeptidases
16.
MAbs ; 15(1): 2268255, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37876265

RESUMO

The human immune system uses antibodies to neutralize foreign antigens. They are composed of heavy and light chains, both with constant and variable regions. The variable region has six hypervariable loops, also known as complementary-determining regions (CDRs) that determine antibody diversity and antigen specificity. Knowledge of their significance, and certain residues present in these areas, is vital for antibody therapeutics development. This study includes an analysis of more than 11,000 human antibody sequences from the International Immunogenetics information system (IMGT). The analysis included parameters such as length distribution, overall amino acid diversity, amino acid frequency per CDR and residue position within antibody chains. Overall, our findings confirm existing knowledge, such as CDRH3's high length diversity and amino acid variability, increased aromatic residue usage, particularly tyrosine, charged and polar residues like aspartic acid, serine, and the flexible residue glycine. Specific residue positions within each CDR influence these occurrences, implying a unique amino acid type distribution pattern. We compared amino acid type usage in CDRs and non-CDR regions, both in globular and transmembrane proteins, which revealed distinguishing features, such as increased frequency of tyrosine, serine, aspartic acid, and arginine. These findings should prove useful for future optimization, improvement of affinity, synthetic antibody library design, or the creation of antibodies de-novo in silico.


Assuntos
Anticorpos , Ácido Aspártico , Humanos , Sequência de Aminoácidos , Anticorpos/química , Regiões Determinantes de Complementaridade/química , Sistema Imunitário/metabolismo , Serina , Tirosina
17.
J Biol Chem ; 286(34): 29709-17, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21697084

RESUMO

The serpin plasminogen activator inhibitor-1 (PAI-1) is a crucial regulator in fibrinolysis and tissue remodeling. PAI-1 has been associated with several pathological conditions and is a validated prognostic marker in human cancers. However, structural information about the native inhibitory form of PAI-1 has been elusive because of its inherent conformational instability and rapid conversion to a latent, inactive structure. Here we report the crystal structure of PAI-1 W175F at 2.3 Å resolution as the first model of the metastable native molecule. Structural comparison with a quadruple mutant (14-1B) previously used as representative of the active state uncovered key differences. The most striking differences occur near the region that houses three of the four mutations in the 14-1B PAI-1 structure. Prominent changes are localized within a loop connecting ß-strand 3A with the F helix, in which a previously observed 3(10)-helix is absent in the new structure. Notably these structural changes are found near the binding site for the cofactor vitronectin. Because vitronectin is the only known physiological regulator of PAI-1 that slows down the latency conversion, the structure of this region is important. Furthermore, the previously identified chloride-binding site close to the F-helix is absent from the present structure and likely to be artifactual, because of its dependence on the 14-1B mutations. Instead we found a different chlorine-binding site that is likely to be present in wild type PAI-1 and that more satisfactorily accounts for the chlorine stabilizing effect on PAI-1.


Assuntos
Inibidor 1 de Ativador de Plasminogênio/química , Cristalografia por Raios X , Humanos , Mutação de Sentido Incorreto , Inibidor 1 de Ativador de Plasminogênio/genética , Estabilidade Proteica , Estrutura Secundária de Proteína , Termodinâmica
18.
Life Sci Alliance ; 5(8)2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35459736

RESUMO

Mitochondrial homeostasis is tightly controlled by ubiquitination. The mitochondrial integral membrane ubiquitin ligase MARCH5 is a crucial regulator of mitochondrial membrane fission, fusion, and disposal through mitophagy. In addition, the lipid composition of mitochondrial membranes can determine mitochondrial dynamics and organelle turnover. However, how lipids influence the ubiquitination processes that control mitochondrial homeostasis remains unknown. Here, we show that lipids common to the mitochondrial membranes interact with MARCH5 and affect its activity and stability depending on the lipid composition in vitro. As the only one of the tested lipids, cardiolipin binding to purified MARCH5 induces a significant decrease in thermal stability, whereas stabilisation increases the strongest in the presence of phosphatidic acid. Furthermore, we observe that the addition of lipids to purified MARCH5 alters the ubiquitination pattern. Specifically, cardiolipin enhances auto-ubiquitination of MARCH5. Our work shows that lipids can directly affect the activity of ubiquitin ligases and suggests that the lipid composition in mitochondrial membranes could control ubiquitination-dependent mechanisms that regulate the dynamics and turnover of mitochondria.


Assuntos
Membranas Mitocondriais , Proteínas Mitocondriais , Cardiolipinas/metabolismo , Proteínas de Membrana/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Fosfolipídeos/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
19.
J Struct Biol ; 174(2): 296-306, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21182963

RESUMO

The Na+,K+-ATPase belongs to the P-ATPase family, whose characteristic property is the formation of a phosphorylated intermediate. The enzyme is also a defined target for cardiotonic steroids which inhibit its functional activity and initiate intracellular signaling. Here we describe the 4.6 Å resolution crystal structure of the pig kidney Na+,K+-ATPase in its phosphorylated form stabilized by high affinity binding of the cardiotonic steroid ouabain. The steroid binds to a site formed at transmembrane segments αM1-αM6, plugging the ion pathway from the extracellular side. This structure differs from the previously reported low affinity complex with potassium. Most importantly, the A domain has rotated in response to phosphorylation and αM1-2 move towards the ouabain molecule, providing for high affinity interactions and closing the ion pathway from the extracellular side. The observed re-arrangements of the Na+,K+-ATPase stabilized by cardiotonic steroids may affect protein-protein interactions within the intracellular signal transduction networks.


Assuntos
Cardiotônicos/química , Ouabaína/química , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Sítios de Ligação , Cristalografia por Raios X , Magnésio/química , Modelos Moleculares , Fosforilação , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , ATPase Trocadora de Sódio-Potássio/química , Suínos
20.
J Struct Biol ; 171(3): 353-60, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20347991

RESUMO

Human serum albumin (HSA), the major protein component in blood plasma and in extravascular spaces, is known to participate in the binding and transport of a variety of endogenous and exogenous organic compounds with anionic or electronegative features. We here report on the 3.3A resolution crystal structure of HSA complexed with the cationic, and widely used, anesthetic lidocaine. We find that lidocaine and HSA co-crystallise as a dimer in the unusual space group I4(1). The dimer consists of one HSA molecule without ligand and one HSA molecule with a single, bound lidocaine. HSA is a heart-shaped protein composed of three homologous helical domains (I-III), which can be subdivided into two subdomains (A and B), and lidocaine binds to a unique site formed by residues from subdomain IB facing the central, interdomain crevice. In the crystal, binding seems to introduce only local conformational changes in the protein. According to intrinsic fluorescence experiments with aqueous HSA binding results in widespread conformational changes involving Trp214 in subdomain IIA. Results obtained with equilibrium dialysis and isothermal titration calorimetry show that lidocaine binding is of a low affinity and occurs at one discrete binding site in accordance with the X-ray data. Another crystal form of ligand-free HSA obtained in the presence of ammonium sulphate was determined at 2.3A resolution revealing a sulphate ion accepting cavity at the surface of subdomain IIIA. The present results contribute to a further characterisation of the exceptional binding properties of HSA.


Assuntos
Sítios de Ligação/fisiologia , Cristalografia por Raios X/métodos , Lidocaína/metabolismo , Albumina Sérica/química , Albumina Sérica/metabolismo , Sítios de Ligação/genética , Humanos , Lidocaína/química , Ligação Proteica , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa