Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Infect Dis ; 6(4): 725-737, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32092260

RESUMEN

The ability to respire and generate adenosine triphosphate (ATP) is essential for the physiology, persistence, and pathogenicity of Mycobacterium tuberculosis, which causes tuberculosis. By employing a lead repurposing strategy, the malarial cytochrome bc1 inhibitor SCR0911 was tested against mycobacteria. Docking studies were carried out to reveal potential binding and to understand the binding interactions with the target, cytochrome bcc. Whole-cell-based and in vitro assays demonstrated the potency of SCR0911 by inhibiting cell growth and ATP synthesis in both the fast- and slow-growing M. smegmatis and M. bovis bacillus Calmette-Guérin, respectively. The variety of biochemical assays and the use of a cytochrome bcc deficient mutant strain validated the cytochrome bcc oxidase as the direct target of the drug. The data demonstrate the broad-spectrum activity of SCR0911 and open the door for structure-activity relationship studies to improve the potency of new mycobacteria specific SCR0911 analogues.


Asunto(s)
Antimaláricos/farmacología , Antituberculosos/farmacología , Reposicionamiento de Medicamentos , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Mycobacterium/efectos de los fármacos , Adenosina Trifosfato/biosíntesis , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana Múltiple , Simulación del Acoplamiento Molecular
2.
ACS Infect Dis ; 5(12): 2055-2060, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31599569

RESUMEN

Mycobacterium abscessus (M. abscessus) is a rapidly growing nontuberculous mycobacteria that is quickly emerging as a global health concern. M. abscessus pulmonary infections are frequently intractable due to the high intrinsic resistance to most antibiotics. Therefore, there is an urgent need to discover effective pharmacological options for M. abscessus infections. In this study, the potency of the antituberculosis drug Telacebec (Q203) was evaluated against M. abscessus. Q203 is a clinical-stage drug candidate targeting the subunit QcrB of the cytochrome bc1:aa3 terminal oxidase. We demonstrated that the presence of four naturally-occurring polymorphisms in the M. abscessus QcrB is responsible for the high resistance of the bacterium to Q203. Genetics reversion of the four polymorphisms sensitized M. abscessus to Q203. While this study highlights the limitation of a direct drug repurposing approach of Q203 and related drugs for M. abscessus infections, it reveals that the M. abscessus cytochrome bc1:aa3 respiratory branch is sensitive to chemical inhibition.


Asunto(s)
Citocromos/genética , Farmacorresistencia Bacteriana , Imidazoles/farmacología , Mycobacterium abscessus/crecimiento & desarrollo , Piperidinas/farmacología , Polimorfismo de Nucleótido Simple , Piridinas/farmacología , Citocromos/antagonistas & inhibidores , Citocromos/química , Reposicionamiento de Medicamentos , Imidazoles/química , Modelos Moleculares , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Mycobacterium abscessus/efectos de los fármacos , Mycobacterium abscessus/genética , Operón , Piperidinas/química , Unión Proteica , Conformación Proteica , Piridinas/química
3.
J Gen Virol ; 99(2): 194-208, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29300159

RESUMEN

Direct-acting antivirals (DAAs) targeting the non-structural 5A (NS5A) protein of the hepatitis C virus (HCV) are crucial drugs that have shown exceptional clinical success in patients. However, their mode of action (MoA) remains unclear, and drug-resistant HCV strains are rapidly emerging. It is critical to characterize the behaviour of the NS5A protein in solution, which can facilitate the development of new classes of inhibitors or improve the efficacy of the currently available DAAs. Using biophysical methods, including dynamic light scattering, size exclusion chromatography and chemical cross-linking experiments, we showed that the NS5A domain 1 from genotypes 1b and 1a of the HCV intrinsically self-associated and existed as a heterogeneous mixture in solution. Interestingly, the NS5A domain 1 from genotypes 1b and 1a exhibited different dynamic equilibria of monomers to higher-order structures. Using small-angle X-ray scattering, we studied the structural dynamics of the various states of the NS5A domain 1 in solution. We also tested the effect of daclatasvir (DCV), the most prominent DAA, on self-association of the wild and DCV-resistant mutant (Y93H) NS5A domain 1 proteins, and demonstrated that DCV induced the formation of large and irreversible protein aggregates that eventually precipitated out. This study highlights the conformational variability of the NS5A domain 1 of HCV, which may be an intrinsic structural behaviour of the HCV NS5A domain 1 in solution.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Imidazoles/farmacología , Conformación Molecular , Proteínas no Estructurales Virales/química , Carbamatos , Cromatografía en Gel , Farmacorresistencia Viral , Dispersión Dinámica de Luz , Genotipo , Hepacivirus/genética , Dominios Proteicos , Pirrolidinas , Dispersión del Ángulo Pequeño , Valina/análogos & derivados , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética
4.
ACS Infect Dis ; 3(11): 807-819, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28991455

RESUMEN

Previously, we showed that a major in vitro and in vivo mechanism of resistance to pyrazinoic acid (POA), the bioactive component of the critical tuberculosis (TB) prodrug pyrazinamide (PZA), involves missense mutations in the aspartate decarboxylase PanD, an enzyme required for coenzyme A biosynthesis. What is the mechanism of action of POA? Upon demonstrating that treatment of M. bovis BCG with POA resulted in a depletion of intracellular coenzyme A and confirming that this POA-mediated depletion is prevented by either missense mutations in PanD or exogenous supplementation of pantothenate, we hypothesized that POA binds to PanD and that this binding blocks the biosynthetic pathway. Here, we confirm both hypotheses. First, metabolomic analyses showed that POA treatment resulted in a reduction of the concentrations of all coenzyme A precursors downstream of the PanD-mediated catalytic step. Second, using isothermal titration calorimetry, we established that POA, but not its prodrug PZA, binds to PanD. Binding was abolished for mutant PanD proteins. Taken together, these findings support a mechanism of action of POA in which the bioactive component of PZA inhibits coenzyme A biosynthesis via binding to aspartate decarboxylase PanD. Together with previous works, these results establish PanD as a genetically, metabolically, and biophysically validated target of PZA.


Asunto(s)
Antituberculosos/farmacología , Carboxiliasas/antagonistas & inhibidores , Mycobacterium tuberculosis/enzimología , Pirazinamida/análogos & derivados , Sitios de Unión , Carbono/metabolismo , Coenzima A , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Modelos Moleculares , Mycobacterium bovis/efectos de los fármacos , NAD/biosíntesis , Unión Proteica , Conformación Proteica , Pirazinamida/farmacología
5.
Biochim Biophys Acta Gen Subj ; 1861(12): 3201-3214, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28935609

RESUMEN

The ability of the vancomycin-resistant Enterococcus faecalis (V583) to restore redox homeostasis via antioxidant defense mechanism is of importance, and knowledge into this defense is essential to understand its antibiotic-resistance and survival in hosts. The flavoprotein disulfide reductase AhpR, composed of the subunits AhpC and AhpF, represents one such vital part. Circular permutation was found to be a feature of the AhpF protein family. E. faecalis (V583) AhpF (EfAhpF) appears to be a representative of a minor subclass of this family, the typically N-terminal two-fold thioredoxin-like domain (NTD_N/C) is located at the C-terminus, whereas the pyridine nucleotide-disulfide oxidoreductase domain is encoded in the N-terminal part of its sequence. In EfAhpF, these two domains are connected via an unusually long linker region providing optimal communication between both domains. EfAhpF forms a dimer in solution similar to Escherichia coli AhpF. The crystallographic 2.3Å resolution structure of the NTD_N/C domain reveals a unique loop-helix stretch (409ILKDTEPAKELLYGIEKM426) not present in homologue domains of other prokaryotic AhpFs. Deletion of the unique 415PAKELLY421-helix or of 415PAKELL420 affects protein stability or attenuates peroxidase activity. Furthermore, mutation of Y421 is described to be essential for E. faecalis AhpF's optimal NADH-oxidative activity.


Asunto(s)
Enterococcus faecalis/enzimología , Peroxirredoxinas/química , Enterococos Resistentes a la Vancomicina/enzimología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Modelos Moleculares , Dominios Proteicos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína , Dispersión del Ángulo Pequeño
6.
FEBS Lett ; 591(15): 2323-2337, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28672070

RESUMEN

Modulation of intracellular guanosine 3',5'-bispyrophosphate ((p)ppGpp) level, the effector of the stringent response, is crucial for survival as well as optimal growth of prokaryotes and, thus, for bacterial pathogenesis and dormancy. In Mycobacterium tuberculosis (Mtb), (p)ppGpp synthesis and degradation are carried out by the bifunctional enzyme MtRel, which consists of 738 residues, including an N-terminal hydrolase- and synthetase-domain (N-terminal domain or NTD) and a C-terminus with a ribosome-binding site. Here, we present the first crystallographic structure of the enzymatically active MtRel NTD determined at 3.7 Å resolution. The structure provides insights into the residues of MtRel NTD responsible for nucleotide binding. Small-angle X-ray scattering experiments were performed to investigate the dimeric state of the MtRel NTD and possible substrate-dependent structural alterations.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/química , Pirofosfatasas/química , Pirofosfatasas/metabolismo , Proteínas Bacterianas/genética , Cromatografía Líquida de Alta Presión , Cristalografía por Rayos X , Ligasas/química , Ligasas/genética , Ligasas/metabolismo , Conformación Proteica , Dominios Proteicos , Multimerización de Proteína , Pirofosfatasas/genética , Dispersión del Ángulo Pequeño , Difracción de Rayos X
7.
Biochim Biophys Acta Gen Subj ; 1861(9): 2354-2366, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28499823

RESUMEN

Mycobacteria employ a versatile machinery of the mycothiol-dependent system, containing the proteins mycothiol disulfide reductase (Mtr), the oxido-reductase Mycoredoxin-1 (Mrx-1) and the alkyl-hydroperoxide subunit E (AhpE). The mycothiol-dependent protein ensemble regulates the balance of oxidized-reduced mycothiol, to ensure a reductive intracellular environment for optimal functioning of its proteins even upon exposure to oxidative stress. Here, we determined the first low-resolution solution structure of Mycobacterium tuberculosis Mtr (MtMtr) derived from small-angle X-ray scattering data, which provides insight into its dimeric state. The solution shape reveals the two NADPH-binding domains inside the dimeric MtMtr in different conformations. NMR-titration shows that the MtMtr-MtMrx-1 interaction is characterized by a fast exchange regime and critical residues involved in the protein-protein interaction were identified. Using NMR spectroscopy and docking studies, the epitopes of MtMrx-1 and MtAhpE interaction are described, shedding new light into the interaction interface and mechanism of action. Finally, the essential residue of MtMrx-1 identified in the interaction with MtMtr and MtAhpE form a platform for structure-guided drug design against the versatile enzyme machinery of the mycothiol-dependent system inside M. tuberculosis.


Asunto(s)
Mycobacterium tuberculosis/enzimología , NADH NADPH Oxidorreductasas/química , Peroxirredoxinas/química , Descubrimiento de Drogas , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Conformación Proteica
8.
Free Radic Biol Med ; 97: 588-601, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27417938

RESUMEN

Mycobacterium tuberculosis (Mtb) has the ability to persist within the human host for a long time in a dormant stage and re-merges when the immune system is compromised. The pathogenic bacterium employs an elaborate antioxidant defence machinery composed of the mycothiol- and thioredoxin system in addition to a superoxide dismutase, a catalase, and peroxiredoxins (Prxs). Among the family of Peroxiredoxins, Mtb expresses a 1-cysteine peroxiredoxin, known as alkylhydroperoxide reductase E (MtAhpE), and defined as a potential tuberculosis drug target. The reduced MtAhpE (MtAhpE-SH) scavenges peroxides to become converted to MtAhpE-SOH. To provide continuous availability of MtAhpE-SH, MtAhpE-SOH has to become reduced. Here, we used NMR spectroscopy to delineate the reduced (MtAhpE-SH), sulphenic (MtAhpE-SOH) and sulphinic (MtAhpE-SO2H) states of MtAhpE through cysteinyl-labelling, and provide for the first time evidence of a mycothiol-dependent mechanism of MtAhpE reduction. This is confirmed by crystallographic studies, wherein MtAhpE was crystallized in the presence of mycothiol and the structure was solved at 2.43Å resolution. Combined with NMR-studies, the crystallographic structures reveal conformational changes of important residues during the catalytic cycle of MtAhpE. In addition, alterations of the overall protein in solution due to redox modulation are observed by small angle X-ray scattering (SAXS) studies. Finally, by employing SAXS and dynamic light scattering, insight is provided into the most probable physiological oligomeric state of MtAhpE necessary for activity, being also discussed in the context of concerted substrate binding inside the dimeric MtAhpE.


Asunto(s)
Proteínas Bacterianas/química , Cisteína/química , Glicopéptidos/química , Inositol/química , Mycobacterium tuberculosis/enzimología , Peroxirredoxinas/química , Dominio Catalítico , Cristalografía por Rayos X , Enlace de Hidrógeno , Redes y Vías Metabólicas , Simulación del Acoplamiento Molecular , Resonancia Magnética Nuclear Biomolecular , Oxidación-Reducción , Unión Proteica , Conformación Proteica en Hélice alfa , Estructura Cuaternaria de Proteína , Dispersión del Ángulo Pequeño , Soluciones , Tiorredoxinas/química
9.
Prog Biophys Mol Biol ; 119(1): 84-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26033199

RESUMEN

The V1VO-ATPase (V-ATPase) is the important proton-pump in eukaryotic cells, responsible for pH-homeostasis, pH-sensing and amino acid sensing, and therefore essential for cell growths and metabolism. ATP-cleavage in the catalytic A3B3-hexamer of V1 has to be communicated via several so-called central and peripheral stalk units to the proton-pumping VO-part, which is membrane-embedded. A unique feature of V1VO-ATPase regulation is its reversible disassembly of the V1 and VO domain. Actin provides a network to hold the V1 in proximity to the VO, enabling effective V1VO-assembly to occur. Besides binding to actin, the 14-subunit V-ATPase interacts with multi-subunit machineries to form cellular sensors, which regulate the pH in cellular compartments or amino acid signaling in lysosomes. Here we describe a variety of subunit-subunit interactions within the V-ATPase enzyme during catalysis and its protein-protein assembling with key cellular machineries, essential for cellular function.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Eucariontes/citología , Eucariontes/enzimología , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Animales , Biocatálisis , Humanos , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Protones
10.
Biochim Biophys Acta ; 1847(10): 1139-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26092085

RESUMEN

Redox homeostasis is significant for the survival of pro- and eukaryotic cells and is crucial for defense against reactive oxygen species like superoxide and hydrogen peroxide. In Escherichia coli, the reduction of peroxides occurs via the redox active disulfide center of the alkyl hydroperoxide reductase C subunit (AhpC), whose reduced state becomes restored by AhpF. The 57kDa EcAhpF contains an N-terminal domain (NTD), which catalyzes the electron transfer from NADH via an FAD of the C-terminal domain into EcAhpC. The NTD is connected to the C-terminal domain via a linker. Here, the first crystal structure of E. coli AhpF bound with NADH and NAD(+) has been determined at 2.5Å and 2.4Å resolution, respectively. The NADH-bound form of EcAhpF reveals that the NADH-binding domain is required to alter its conformation to bring a bound NADH to the re-face of the isoalloxazine ring of the flavin, and thereby render the NADH-domain dithiol center accessible to the NTD disulfide center for electron transfer. The NAD(+)-bound form of EcAhpF shows conformational differences for the nicotinamide end moieties and its interacting residue M467, which is proposed to represent an intermediate product-release conformation. In addition, the structural alterations in EcAhpF due to NADH- and NAD(+)-binding in solution are shown by small angle X-ray scattering studies. The EcAhpF is revealed to adopt many intermediate conformations in solution to facilitate the electron transfer from the substrate NADH to the C-terminal domain, and subsequently to the NTD of EcAhpF for the final step of AhpC reduction.

11.
J Biol Chem ; 290(6): 3183-96, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25505269

RESUMEN

Eukaryotic V1VO-ATPases hydrolyze ATP in the V1 domain coupled to ion pumping in VO. A unique mode of regulation of V-ATPases is the reversible disassembly of V1 and VO, which reduces ATPase activity and causes silencing of ion conduction. The subunits D and F are proposed to be key in these enzymatic processes. Here, we describe the structures of two conformations of the subunit DF assembly of Saccharomyces cerevisiae (ScDF) V-ATPase at 3.1 Å resolution. Subunit D (ScD) consists of a long pair of α-helices connected by a short helix ((79)IGYQVQE(85)) as well as a ß-hairpin region, which is flanked by two flexible loops. The long pair of helices is composed of the N-terminal α-helix and the C-terminal helix, showing structural alterations in the two ScDF structures. The entire subunit F (ScF) consists of an N-terminal domain of four ß-strands (ß1-ß4) connected by four α-helices (α1-α4). α1 and ß2 are connected via the loop (26)GQITPETQEK(35), which is unique in eukaryotic V-ATPases. Adjacent to the N-terminal domain is a flexible loop, followed by a C-terminal α-helix (α5). A perpendicular and extended conformation of helix α5 was observed in the two crystal structures and in solution x-ray scattering experiments, respectively. Fitted into the nucleotide-bound A3B3 structure of the related A-ATP synthase from Enterococcus hirae, the arrangements of the ScDF molecules reflect their central function in ATPase-coupled ion conduction. Furthermore, the flexibility of the terminal helices of both subunits as well as the loop (26)GQITPETQEK(35) provides information about the regulatory step of reversible V1VO disassembly.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , ATPasas de Translocación de Protón Vacuolares/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química
12.
Adv Exp Med Biol ; 827: 85-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25387961

RESUMEN

The process of islet amyloid polypeptide (IAPP) formation and the prefibrillar oligomers are supposed to be one of the pathogenic agents causing pancreatic ß-cell dysfunction. The human IAPP (hIAPP) aggregates easily and therefore, it is difficult to characterize its structural features by standard biophysical tools. The rat version of IAPP (rIAPP) that differs by six amino acids when compared with hIAPP, is not prone to aggregation and does not form amyloid fibrils. Similar to hIAPP it also demonstrates random-coiled nature in solution. The structural propensity of rIAPP has been studied as a hIAPP mimic in recent works. However, the overall shape of it in solution still remains elusive. Using small angle X-ray scattering (SAXS) measurements combined with nuclear magnetic resonance (NMR) and molecular dynamics simulations (MD) the solution structure of rIAPP was studied. An unambiguously extended structural model with a radius of gyration of 1.83 nm was determined from SAXS data. Consistent with previous studies, an overall random-coiled feature with residual helical propensity in the N-terminus was confirmed. Combined efforts are necessary to unambiguously resolve the structural features of intrinsic disordered proteins.


Asunto(s)
Polipéptido Amiloide de los Islotes Pancreáticos/química , Animales , Espectroscopía de Resonancia Magnética , Conformación Proteica , Ratas , Dispersión de Radiación , Soluciones
13.
Biochim Biophys Acta ; 1837(6): 940-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24650628

RESUMEN

Archaea live under different environmental conditions, such as high salinity, extreme pHs and cold or hot temperatures. How energy is conserved under such harsh environmental conditions is a major question in cellular bioenergetics of archaea. The key enzymes in energy conservation are the archaeal A1AO ATP synthases, a class of ATP synthases distinct from the F1FO ATP synthase ATP synthase found in bacteria, mitochondria and chloroplasts and the V1VO ATPases of eukaryotes. A1AO ATP synthases have distinct structural features such as a collar-like structure, an extended central stalk, and two peripheral stalks possibly stabilizing the A1AO ATP synthase during rotation in ATP synthesis/hydrolysis at high temperatures as well as to provide the storage of transient elastic energy during ion-pumping and ATP synthesis/-hydrolysis. High resolution structures of individual subunits and subcomplexes have been obtained in recent years that shed new light on the function and mechanism of this unique class of ATP synthases. An outstanding feature of archaeal A1AO ATP synthases is their diversity in size of rotor subunits and the coupling ion used for ATP synthesis with H(+), Na(+) or even H(+) and Na(+) using enzymes. The evolution of the H(+) binding site to a Na(+) binding site and its implications for the energy metabolism and physiology of the cell are discussed.


Asunto(s)
Archaea/enzimología , Proteínas Arqueales/fisiología , ATPasas de Translocación de Protón Mitocondriales/fisiología , Proteínas Motoras Moleculares/fisiología , Proteínas Arqueales/química , Sitios de Unión , Biocatálisis , Dominio Catalítico , ATPasas de Translocación de Protón Mitocondriales/química , Modelos Moleculares , Proteínas Motoras Moleculares/química
14.
J Biol Chem ; 288(17): 11930-9, 2013 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-23476018

RESUMEN

Subunit F of V-ATPases is proposed to undergo structural alterations during catalysis and reversible dissociation from the V1VO complex. Recently, we determined the low resolution structure of F from Saccharomyces cerevisiae V-ATPase, showing an N-terminal egg shape, connected to a C-terminal hook-like segment via a linker region. To understand the mechanistic role of subunit F of S. cerevisiae V-ATPase, composed of 118 amino acids, the crystal structure of the major part of F, F(1-94), was solved at 2.3 Å resolution. The structural features were confirmed by solution NMR spectroscopy using the entire F subunit. The eukaryotic F subunit consists of the N-terminal F(1-94) domain with four-parallel ß-strands, which are intermittently surrounded by four α-helices, and the C terminus, including the α5-helix encompassing residues 103 to 113. Two loops (26)GQITPETQEK(35) and (60)ERDDI(64) are described to be essential in mechanistic processes of the V-ATPase enzyme. The (26)GQITPETQEK(35) loop becomes exposed when fitted into the recently determined EM structure of the yeast V1VO-ATPase. A mechanism is proposed in which the (26)GQITPETQEK(35) loop of subunit F and the flexible C-terminal domain of subunit H move in proximity, leading to an inhibitory effect of ATPase activity in V1. Subunits D and F are demonstrated to interact with subunit d. Together with NMR dynamics, the role of subunit F has been discussed in the light of its interactions in the processes of reversible disassembly and ATP hydrolysis of V-ATPases by transmitting movements of subunit d and H of the VO and V1 sector, respectively.


Asunto(s)
Subunidades de Proteína/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , ATPasas de Translocación de Protón Vacuolares/química , Adenosina Trifosfato/química , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Cristalografía por Rayos X , Hidrólisis , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo
15.
J Bioenerg Biomembr ; 45(1-2): 121-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104121

RESUMEN

Tuberculosis, caused by the strain Mycobacterium tuberculosis, is in focus of interest due to the emergence of multi- and extensive drug-resistant TB strains. The F(1)F(O) ATP synthase is one of the essential enzymes in energy requirement of both proliferating aerobic and hypoxic dormant stage of mycobacterium life cycle, and therefore a potential TB drug target. Subunit γ of F-ATP synthases plays an important role in coupling and catalysis via conformational transitions of its N- and C-termini as well as the bottom segment of the globular domain of γ, which is in close proximity to the rotating and ion-pumping c-ring. Here we describe the first production, purification and low resolution solution structure of subunit γ (γ(1-204), Mtγ(1-204)) of the M. tuberculosis F-ATP synthase. Mtγ(1-204) is a pear-like shaped protein with a molecular weight of 23 ± 2 kDa. Protein sequence analysis of Mtγ revealed differences in the amino acid composition to γ subunits from other sources, in particular the presence of a unique stretch of 13 amino acid residues (Mtγ(165-178)). NMR studies showed that Mtγ(165-178) forms a loop of polar residues. Mtγ(165-178) has been aligned at the bottom of the globular domain of the Escherichia coli subunit γ, being in close vicinity to the polar residues R41, Q42, E44 and Q46 (M. tuberculosis nomenclature) of the c-ring. The putative role(s) of Mtγ(165-178) in coupling and as a potential drug target are discussed.


Asunto(s)
Proteínas Bacterianas/química , Mycobacterium tuberculosis/enzimología , Subunidades de Proteína/química , ATPasas de Translocación de Protón/química , Antituberculosos/química , Antituberculosos/uso terapéutico , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catálisis , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Mycobacterium tuberculosis/genética , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , ATPasas de Translocación de Protón/antagonistas & inhibidores , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tuberculosis/tratamiento farmacológico , Tuberculosis/enzimología
16.
Antimicrob Agents Chemother ; 57(1): 168-76, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23089752

RESUMEN

The subunit ε of bacterial F(1)F(O) ATP synthases plays an important regulatory role in coupling and catalysis via conformational transitions of its C-terminal domain. Here we present the first low-resolution solution structure of ε of Mycobacterium tuberculosis (Mtε) F(1)F(O) ATP synthase and the nuclear magnetic resonance (NMR) structure of its C-terminal segment (Mtε(103-120)). Mtε is significantly shorter (61.6 Å) than forms of the subunit in other bacteria, reflecting a shorter C-terminal sequence, proposed to be important in coupling processes via the catalytic ß subunit. The C-terminal segment displays an α-helical structure and a highly positive surface charge due to the presence of arginine residues. Using NMR spectroscopy, fluorescence spectroscopy, and mutagenesis, we demonstrate that the new tuberculosis (TB) drug candidate TMC207, proposed to bind to the proton translocating c-ring, also binds to Mtε. A model for the interaction of TMC207 with both ε and the c-ring is presented, suggesting that TMC207 forms a wedge between the two rotating subunits by interacting with the residues W15 and F50 of ε and the c-ring, respectively. T19 and R37 of ε provide the necessary polar interactions with the drug molecule. This new model of the mechanism of TMC207 provides the basis for the design of new drugs targeting the F(1)F(O) ATP synthase in M. tuberculosis.


Asunto(s)
Antituberculosos/química , Proteínas Bacterianas/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , Subunidades de Proteína/antagonistas & inhibidores , Quinolinas/química , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Diarilquinolinas , Escherichia coli/genética , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/genética , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Resonancia Magnética Nuclear Biomolecular , Subunidades de Proteína/química , Subunidades de Proteína/genética , Protones , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alineación de Secuencia , Espectrometría de Fluorescencia
17.
J Struct Biol ; 180(3): 509-18, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23063756

RESUMEN

The nucleotide binding sites in A-ATP synthases are located at the interfaces of subunit A and B, which is proposed to play a regulatory role. Differential binding of MgATP and -ADP to subunit B has been described, which does not exist in the related α and B subunits of F-ATP synthases and V-ATPases, respectively. The conserved phosphate loop residues, histidine and asparagine, of the A-ATP synthase subunit B have been proposed to be essential for γ-phosphate interaction. To investigate the role of these conserved P-loop residues in nucleotide-binding, subunit B residues H156 and N157 of the Methanosarcina mazei Gö1 A-ATP synthase were separately substituted with alanine. In addition, N157 was mutated to threonine, because it is the corresponding amino acid in the P-loop of F-ATP synthase subunit α. The structures of the subunit B mutants H156A, N157A/T were solved up to a resolution of 1.75 and 1.7 Å. The binding constants for MgATP and -ADP were determined, demonstrating that the H156A and N157A mutants have a preference to the nucleotide over the wild type and N157T proteins. Importantly, the ability to distinguish MgATP or -ADP was lost, demonstrating that the histidine and asparagine residues are crucial for nucleotide differentiation in subunit B. The structures reveal that the enhanced binding of the alanine mutants is attributed to the increased accessibility of the nucleotide binding cavity, explaining that the structural arrangement of the conserved H156 and N157 define the nucleotide-binding characteristics of the regulatory subunit B of A-ATP synthases.


Asunto(s)
Adenosina Difosfato/química , Adenosina Trifosfato/química , Proteínas Arqueales/química , Asparagina/química , Histidina/química , Methanosarcina/genética , Subunidades de Proteína/química , ATPasas de Translocación de Protón/química , Alanina/química , Alanina/genética , Sustitución de Aminoácidos , Proteínas Arqueales/genética , Asparagina/genética , Sitios de Unión , Cristalografía por Rayos X , Escherichia coli/genética , Histidina/genética , Methanosarcina/enzimología , Simulación del Acoplamiento Molecular , Unión Proteica , Subunidades de Proteína/genética , ATPasas de Translocación de Protón/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Especificidad por Sustrato , Treonina/química , Treonina/genética
18.
Artículo en Inglés | MEDLINE | ID: mdl-22949193

RESUMEN

V-ATPases are very complex multi-subunit enzymes which function as proton-pumping rotary nanomotors. The rotary and coupling subunit F (F(1-94)) was crystallized by the hanging-drop vapour-diffusion method. The native crystals diffracted to a resolution of 2.64 Å and belonged to space group C222(1), with unit-cell parameters a = 47.21, b = 160.26, c = 102.49 Å. The selenomethionyl form of the F(1-94) I69M mutant diffracted to a resolution of 2.3 Å and belonged to space group C222(1), with unit-cell parameters a = 47.22, b = 160.83, c = 102.74 Å. Initial phasing and model building suggested the presence of four molecules in the asymmetric unit.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , ATPasas de Translocación de Protón Vacuolares/química , Cristalización , Cristalografía por Rayos X
19.
PLoS One ; 7(12): e53051, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300861

RESUMEN

HAMLET (Human Alpha-lactalbumin Made LEthal to Tumor cells) is the first member in a new family of protein-lipid complexes with broad tumoricidal activity. Elucidating the molecular structure and the domains crucial for HAMLET formation is fundamental for understanding its tumoricidal function. Here we present the low-resolution solution structure of the complex of oleic acid bound HAMLET, derived from small angle X-ray scattering data. HAMLET shows a two-domain conformation with a large globular domain and an extended part of about 2.22 nm in length and 1.29 nm width. The structure has been superimposed into the related crystallographic structure of human α-lactalbumin, revealing that the major part of α-lactalbumin accommodates well in the shape of HAMLET. However, the C-terminal residues from L105 to L123 of the crystal structure of the human α-lactalbumin do not fit well into the HAMLET structure, resulting in an extended conformation in HAMLET, proposed to be required to form the tumoricidal active HAMLET complex with oleic acid. Consistent with this low resolution structure, we identified biologically active peptide epitopes in the globular as well as the extended domains of HAMLET. Peptides covering the alpha1 and alpha2 domains of the protein triggered rapid ion fluxes in the presence of sodium oleate and were internalized by tumor cells, causing rapid and sustained changes in cell morphology. The alpha peptide-oleate bound forms also triggered tumor cell death with comparable efficiency as HAMLET. In addition, shorter peptides corresponding to those domains are biologically active. These findings provide novel insights into the structural prerequisites for the dramatic effects of HAMLET on tumor cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Lactalbúmina/metabolismo , Lactalbúmina/farmacología , Ácidos Oléicos/metabolismo , Ácidos Oléicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Humanos , Lactalbúmina/química , Ácidos Oléicos/química , Conformación Proteica , Pliegue de Proteína
20.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 12): 1485-91, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22139149

RESUMEN

A reporter tryptophan residue was individually introduced by site-directed mutagenesis into the adenine-binding pocket of the catalytic subunit A (F427W and F508W mutants) of the motor protein A(1)A(O) ATP synthase from Pyrococcus horikoshii OT3. The crystal structures of the F427W and F508W mutant proteins were determined to 2.5 and 2.6 Å resolution, respectively. The tryptophan substitution caused the fluorescence signal to increase by 28% (F427W) and 33% (F508W), with a shift from 333 nm in the wild-type protein to 339 nm in the mutant proteins. Tryptophan emission spectra showed binding of Mg-ATP to the F427W mutant with a K(d) of 8.5 µM. In contrast, no significant binding of nucleotide could be observed for the F508W mutant. A closer inspection of the crystal structure of the F427W mutant showed that the adenine-binding pocket had widened by 0.7 Å (to 8.70 Å) in comparison to the wild-type subunit A (8.07 Å) owing to tryptophan substitution, as a result of which it was able to bind ATP. In contrast, the adenine-binding pocket had narrowed in the F508W mutant. The two mutants presented demonstrate that the exact volume of the adenine ribose binding pocket is essential for nucleotide binding and even minor narrowing makes it unfit for nucleotide binding. In addition, structural and fluorescence data confirmed the viability of the fluorescently active mutant F427W, which had ideal tryptophan spectra for future structure-based time-resolved dynamic measurements of the catalytic subunit A of the ATP-synthesizing enzyme A-ATP synthase.


Asunto(s)
Adenosina Trifosfatasas/química , Dominio Catalítico , Pyrococcus horikoshii/enzimología , Adenina/química , Adenina/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Mutación , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Espectrometría de Fluorescencia , Triptófano/química , Triptófano/genética , Triptófano/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA