Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 2024 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-38769033

RESUMO

The emergence and ongoing evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the need for rapid vaccine development platforms that can be updated to counteract emerging variants of currently circulating and future emerging coronaviruses. Here we report the development of a "train model" subunit vaccine platform that contains a SARS-CoV-2 Wuhan S1 protein (the "engine") linked to a series of flexible receptor binding domains (RBDs; the "cars") derived from SARS-CoV-2 variants of concern (VOCs). We demonstrate that these linked subunit vaccines when combined with Sepivac SWE™, a squalene in water emulsion (SWE) adjuvant, are immunogenic in Syrian hamsters and subsequently provide protection from infection with SARS-CoV-2 VOCs Omicron (BA.1), Delta, and Beta. Importantly, the bivalent and trivalent vaccine candidates offered protection against some heterologous SARS-CoV-2 VOCs that were not included in the vaccine design, demonstrating the potential for broad protection against a range of different VOCs. Furthermore, these formulated vaccine candidates were stable at 2-8 °C for up to 13 months post-formulation, highlighting their utility in low-resource settings. Indeed, our vaccine platform will enable the development of safe and broadly protective vaccines against emerging betacoronaviruses that pose a significant health risk for humans and agricultural animals.

2.
Elife ; 132024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38640016

RESUMO

Mediator of ERBB2-driven cell motility 1 (MEMO1) is an evolutionary conserved protein implicated in many biological processes; however, its primary molecular function remains unknown. Importantly, MEMO1 is overexpressed in many types of cancer and was shown to modulate breast cancer metastasis through altered cell motility. To better understand the function of MEMO1 in cancer cells, we analyzed genetic interactions of MEMO1 using gene essentiality data from 1028 cancer cell lines and found multiple iron-related genes exhibiting genetic relationships with MEMO1. We experimentally confirmed several interactions between MEMO1 and iron-related proteins in living cells, most notably, transferrin receptor 2 (TFR2), mitoferrin-2 (SLC25A28), and the global iron response regulator IRP1 (ACO1). These interactions indicate that cells with high-MEMO1 expression levels are hypersensitive to the disruptions in iron distribution. Our data also indicate that MEMO1 is involved in ferroptosis and is linked to iron supply to mitochondria. We have found that purified MEMO1 binds iron with high affinity under redox conditions mimicking intracellular environment and solved MEMO1 structures in complex with iron and copper. Our work reveals that the iron coordination mode in MEMO1 is very similar to that of iron-containing extradiol dioxygenases, which also display a similar structural fold. We conclude that MEMO1 is an iron-binding protein that modulates iron homeostasis in cancer cells.


Assuntos
Homeostase , Ferro , Neoplasias , Humanos , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Ferroptose , Ferro/metabolismo , Proteína 1 Reguladora do Ferro , Neoplasias/metabolismo , Neoplasias/genética , Ligação Proteica , Receptores da Transferrina/metabolismo , Receptores da Transferrina/genética
3.
PLoS Pathog ; 19(8): e1011572, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37607182

RESUMO

Pathogen life history theory assumes a positive relationship between pathogen load in host tissues and pathogen transmission. Empirical evidence for this relationship is surprisingly rare due to the difficulty of measuring transmission for many pathogens. The comparative method, where a common host is experimentally infected with a set of pathogen strains, is a powerful approach for investigating the relationships between pathogen load and transmission. The validity of such experimental estimates of strain-specific transmission is greatly enhanced if they can predict the pathogen population strain structure in nature. Borrelia burgdorferi is a multi-strain, tick-borne spirochete that causes Lyme disease in North America. This study used 11 field-collected strains of B. burgdorferi, a rodent host (Mus musculus, C3H/HeJ) and its tick vector (Ixodes scapularis) to determine the relationship between pathogen load in host tissues and lifetime host-to-tick transmission (HTT). Mice were experimentally infected via tick bite with 1 of 11 strains. Lifetime HTT was measured by infesting mice with I. scapularis larval ticks on 3 separate occasions. The prevalence and abundance of the strains in the mouse tissues and the ticks were determined by qPCR. We used published databases to obtain estimates of the frequencies of these strains in wild I. scapularis tick populations. Spirochete loads in ticks and lifetime HTT varied significantly among the 11 strains of B. burgdorferi. Strains with higher spirochete loads in the host tissues were more likely to infect feeding larval ticks, which molted into nymphal ticks that had a higher probability of B. burgdorferi infection (i.e., higher HTT). Our laboratory-based estimates of lifetime HTT were predictive of the frequencies of these strains in wild I. scapularis populations. For B. burgdorferi, the strains that establish high abundance in host tissues and that have high lifetime transmission are the strains that are most common in nature.


Assuntos
Borrelia burgdorferi , Ixodes , Doença de Lyme , Animais , Camundongos , Camundongos Endogâmicos C3H , Larva
4.
Biophys J ; 120(20): 4600-4607, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34461106

RESUMO

ATP7A and ATP7B are structurally similar but functionally distinct active copper transporters that regulate copper levels in the human cells and deliver copper to the biosynthetic pathways. Both proteins have a chain of six cytosolic metal-binding domains (MBDs) believed to be involved in the copper-dependent regulation of the activity and intracellular localization of these enzymes. Although all the MBDs are quite similar in structure, their spacing differs markedly between ATP7A and ATP7B. We show by NMR that the long polypeptide between MBD1 and MBD2 of ATP7A forms an additional seventh metastable domain, which we called HMA1A (heavy metal associated domain 1A). The structure of HMA1A resembles the MBDs but contains no copper-binding site. The HMA1A domain, which is unique to ATP7A, may modulate regulatory interactions between MBD1-3, contributing to the distinct functional properties of ATP7A and ATP7B.


Assuntos
ATPases Transportadoras de Cobre , Cobre , Sítios de Ligação , Cobre/metabolismo , ATPases Transportadoras de Cobre/genética , Humanos , Domínios Proteicos
5.
Metallomics ; 12(12): 1941-1950, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33094790

RESUMO

Nanobodies are genetically engineered single domain antibodies derived from the unusual heavy-chain only antibodies found in llamas and camels. The small size of the nanobodies and flexible selection schemes make them uniquely versatile tools for protein biochemistry and cell biology. We have developed a panel of nanobodies against the metal binding domains of the human copper transporter ATP7B, a multidomain membrane protein with a complex regulation of enzymatic activity and intracellular localization. To enable the use of the nanobodies as tools to investigate copper transport in the cell, we characterized their binding sites and affinity by isothermal titration calorimetry and NMR. We have identified nanobodies against each of the first four metal binding domains of ATP7B, with a wide affinity range, as evidenced by dissociation constants from below 10-9 to 10-6 M. We found both the inhibitory and activating nanobodies among those tested. The diverse properties of the nanobodies make the panel useful for the structural studies of ATP7B, immunoaffinity purification of the protein, modulation of its activity in the cell, protein dynamics studies, and as mimics of copper chaperone ATOX1, the natural interaction partner of ATP7B.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Anticorpos de Domínio Único/farmacologia , Sítios de Ligação/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , ATPases Transportadoras de Cobre/química , Humanos , Simulação de Acoplamento Molecular , Domínios Proteicos/efeitos dos fármacos
6.
J Mol Biol ; 431(15): 2777-2789, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31158365

RESUMO

Proteins that perform active transport must alternate the access of a binding site, first to one side of a membrane and then to the other, resulting in the transport of bound substrates across the membrane. To better understand this process, we sought to identify mutants of the small multidrug resistance transporter EmrE with reduced rates of alternating access. We performed extensive scanning mutagenesis by changing every amino acid residue to Val, Ala, or Gly, and then screening the drug resistance phenotypes of the resulting mutants. We identified EmrE mutants that had impaired transport activity but retained the ability to bind substrate and further tested their alternating access rates using NMR. Ultimately, we were able to identify a single mutation, S64V, which significantly reduced the rate of alternating access but did not impair substrate binding. Six other transport-impaired mutants did not have reduced alternating access rates, highlighting the importance of other aspects of the transport cycle to achieve drug resistance activity in vivo. To better understand the transport cycle of EmrE, efforts are now underway to determine a high-resolution structure using the S64V mutant identified here.


Assuntos
Substituição de Aminoácidos , Antiporters/genética , Antiporters/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Sítios de Ligação , Transporte Biológico , Resistência a Múltiplos Medicamentos , Escherichia coli/genética , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica
7.
Sci Rep ; 8(1): 11361, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-30054535

RESUMO

ATP synthase is powered by the flow of protons through the molecular turbine composed of two α-helical integral membrane proteins, subunit a, which makes a stator, and a cylindrical rotor assembly made of multiple copies of subunit c. Transient protonation of a universally conserved carboxylate on subunit c (D61 in E. coli) gated by the electrostatic interaction with arginine on subunit a (R210 in E. coli) is believed to be a crucial step in proton transfer across the membrane. We used a fusion protein consisting of subunit a and the adjacent helices of subunit c to test by NMR spectroscopy if cD61 and aR210 are involved in an electrostatic interaction with each other, and found no evidence of such interaction. We have also determined that R140 does not form a salt bridge with either D44 or D124 as was suggested previously by mutation analysis. Our results demonstrate the potential of using arginines as NMR reporter groups for structural and functional studies of challenging membrane proteins.


Assuntos
Modelos Moleculares , Engenharia de Proteínas , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo , Sais/química , Arginina/química , Ácido Aspártico/química , Escherichia coli/enzimologia , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
8.
Mol Biol Cell ; 27(20): 3052-3064, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27559134

RESUMO

The spindle midzone harbors both microtubules and proteins necessary for furrow formation and the completion of cytokinesis. However, the mechanisms that mediate the temporal and spatial recruitment of cell division factors to the spindle midzone and midbody remain unclear. Here we describe a mechanism governed by the conserved RNA-binding protein ATX-2/Ataxin-2, which targets and maintains ZEN-4 at the spindle midzone. ATX-2 does this by regulating the amount of PAR-5 at mitotic structures, particularly the spindle, centrosomes, and midbody. Preventing ATX-2 function leads to elevated levels of PAR-5, enhanced chromatin and centrosome localization of PAR-5-GFP, and ultimately a reduction of ZEN-4-GFP at the spindle midzone. Codepletion of ATX-2 and PAR-5 rescued the localization of ZEN-4 at the spindle midzone, indicating that ATX-2 mediates the localization of ZEN-4 upstream of PAR-5. We provide the first direct evidence that ATX-2 is necessary for cytokinesis and suggest a model in which ATX-2 facilitates the targeting of ZEN-4 to the spindle midzone by mediating the posttranscriptional regulation of PAR-5.


Assuntos
Ataxina-2/metabolismo , Ataxina-2/fisiologia , Citocinese/fisiologia , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Centrossomo/metabolismo , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitose , RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fuso Acromático/metabolismo
9.
Protein Sci ; 21(2): 279-88, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22162071

RESUMO

NMR structure determination of large membrane proteins is hampered by broad spectral lines, overlap, and ambiguity of signal assignment. Chemical shift and NOE assignment can be facilitated by amino acid selective isotope labeling in cell-free protein synthesis system. However, many biological detergents are incompatible with the cell-free synthesis, and membrane proteins often have to be synthesized in an insoluble form. We report cell-free synthesis of subunits a and c of the proton channel of Escherichia coli ATP synthase in a soluble form in a mixture of phosphatidylcholine derivatives. In comparison, subunit a was purified from the cell-free system and from the bacterial cell membranes. NMR spectra of both preparations were similar, indicating that our procedure for cell-free synthesis produces protein structurally similar to that prepared from the cell membranes.


Assuntos
Membrana Celular/metabolismo , Proteínas de Membrana/biossíntese , Dobramento de Proteína , ATPases Translocadoras de Prótons/biossíntese , ATPases Bacterianas Próton-Translocadoras/química , ATPases Bacterianas Próton-Translocadoras/metabolismo , Membrana Celular/química , Sistema Livre de Células/enzimologia , Sistema Livre de Células/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana/química , Micelas , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Fosfolipídeos/química , Fosfolipídeos/farmacologia , Multimerização Proteica/fisiologia , Estrutura Quaternária de Proteína , Subunidades Proteicas/biossíntese , Subunidades Proteicas/química , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/metabolismo
10.
J Biol Chem ; 286(44): 38583-38591, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21900248

RESUMO

Subunit a is the main part of the membrane stator of the ATP synthase molecular turbine. Subunit c is the building block of the membrane rotor. We have generated two molecular fusions of a and c subunits with different orientations of the helical hairpin of subunit c. The a/c fusion protein with correct orientation of transmembrane helices was inserted into the membrane, and co-incorporated into the F(0) complex of ATP synthase with wild type subunit c. The fused c subunit was incorporated into the c-ring tethering the ATP synthase rotor to the stator. The a/c fusion with incorrect orientation of the c-helices required wild type subunit c for insertion into the membrane. In this case, the fused c subunit remained on the periphery of the c-ring and did not interfere with rotor movement. Wild type subunit a inserted into the membrane equally well with wild type subunit c and c-ring assembly mutants that remained monomeric in the membrane. These results show that interaction with monomeric subunit c triggers insertion of subunit a into the membrane, and initiates formation of the a-c complex, the ion-translocating module of the ATP synthase. Correct assembly of the ATP synthase incorporating topologically correct fusion of subunits a and c validates using this model protein for high resolution structural studies of the ATP synthase proton channel.


Assuntos
Adenosina Trifosfatases/química , ATPases Bacterianas Próton-Translocadoras/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , ATPases Translocadoras de Prótons/química , Transporte Biológico , Catálise , Membrana Celular/metabolismo , Conformação Molecular , Plasmídeos/metabolismo , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Prótons , Proteínas Recombinantes de Fusão/química
11.
J Biol Chem ; 286(18): 16355-62, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21398519

RESUMO

Wilson disease (WD) is a disorder of copper metabolism caused by mutations in the Cu-transporting ATPase ATP7B. WD is characterized by significant phenotypic variability, the molecular basis of which is poorly understood. The E1064A mutation in the N-domain of ATP7B was previously shown to disrupt ATP binding. We have now determined, by NMR, the structure of the N-domain containing this mutation and compared properties of E1064A and H1069Q, another mutant with impaired ATP binding. The E1064A mutation does not change the overall fold of the N-domain. However, the position of the α1,α2-helical hairpin (α-HH) that houses Glu(1064) and His(1069) is altered. The α-HH movement produces a more open structure compared with the wild-type ATP-bound form and misaligns ATP coordinating residues, thus explaining complete loss of ATP binding. In the cell, neither the stability nor targeting of ATP7B-E1064A to the trans-Golgi network differs significantly from the wild type. This is in a contrast to the H1069Q mutation within the same α-HH, which greatly destabilizes protein both in vitro and in cells. The difference between two mutants can be linked to a lower stability of the α-HH in the H1069Q variant at the physiological temperature. We conclude that the structural stability of the N-domain rather than the loss of ATP binding plays a defining role in the ability of ATP7B to reach the trans-Golgi network, thus contributing to phenotypic variability in WD.


Assuntos
Adenosina Trifosfatases/química , Trifosfato de Adenosina/química , Proteínas de Transporte de Cátions/química , Mutação de Sentido Incorreto , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Substituição de Aminoácidos , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , ATPases Transportadoras de Cobre , Células HEK293 , Degeneração Hepatolenticular/enzimologia , Degeneração Hepatolenticular/genética , Humanos , Ligação Proteica , Estabilidade Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Rede trans-Golgi/enzimologia , Rede trans-Golgi/genética
12.
EMBO J ; 26(1): 232-41, 2007 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-17159901

RESUMO

To date, the biological role of prokaryotic K(+) channels remains unknown. Helicobacter pylori contains a gene encoding a putative K(+) channel (HpKchA) of the two-transmembrane RCK (regulation of K(+) conductance) domain family, but lacks known bacterial K(+) uptake systems. A H. pylori DeltahpKchA mutant presented a strong growth defect at low K(+) concentration, which was compensated by KCl addition. The role of the separate RCK domain was investigated in H. pylori by mutagenesis of its internal start codon, which led to a K(+)-dependent intermediate growth phenotype, consistent with RCK activating channel function. Tagging HpKchA C-terminally, we detected a 1:1 stoichiometry of the full-length HpKchA and the separate RCK domain. We constructed single amino-acid exchanges within the unusual selectivity filter of HpKchA (ATGFGA) in H. pylori and observed complete loss (G74A), a slight defect (G76A or F75G) or wild-type (A77D) channel function. HpKchA was essential for colonization of the murine stomach. These data show, for the first time, a biological function for a prokaryotic K(+) channel, as a K(+) uptake system, essential for the persistence of H. pylori in the gastric environment.


Assuntos
Mucosa Gástrica/microbiologia , Helicobacter pylori/metabolismo , Canais de Potássio/química , Potássio/farmacocinética , Sequência de Aminoácidos , Animais , Códon de Iniciação , Citoplasma/metabolismo , Concentração de Íons de Hidrogênio , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Potássio/química , Canais de Potássio/fisiologia , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
13.
J Bacteriol ; 184(11): 3053-60, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12003947

RESUMO

In the presence of urea the neutrophilic human pathogen Helicobacter pylori survives for several hours at pH 1 with concomitant cytoplasmic pH homeostasis. To study this effect in detail, the transmembrane proton motive force and cytoplasmic urease activity of H. pylori were determined at various pH values. In the absence of urea, the organism maintained a close-to-neutral cytoplasm and an internally negative membrane potential at external pH values greater than 4 to 5. In the presence of urea, H. pylori accomplished cytoplasmic pH homeostasis down to an external pH of 1.2. At this external pH, the cytoplasmic pH was 4.9 and the membrane potential was slightly negative inside. The latter finding is in contrast to the situation in acidophiles, which develop inside-positive membrane potentials under similar conditions. Measurements of the time course of the membrane potential confirmed that addition of urea to the cells led to hyperpolarization. Most likely, this effect was due to electrogenic export of ammonium cations from the cytoplasm. The urease activity of intact cells increased nearly exponentially with decreasing external pH. This activation was not due to enhanced gene expression at low external pH values. In cell extracts the pH optimum of urease activity was dependent on the buffer system and was about pH 5 in sodium citrate buffer. Since this is the cytoplasmic pH of the cells at pH 1 to 2, we propose that cytoplasmic pH is a factor in the in vivo activation of the urease at low external pH values. The mechanism by which urease activity leads to cytoplasmic pH homeostasis in H. pylori is discussed.


Assuntos
Helicobacter pylori/fisiologia , Urease/metabolismo , Citratos , Meios de Cultura , Citoplasma/química , Citoplasma/metabolismo , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/metabolismo , Homeostase , Concentração de Íons de Hidrogênio , Potenciais da Membrana , Força Próton-Motriz , Citrato de Sódio , Ureia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...