Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 385(6705): 168-174, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38900912

RESUMEN

Intercellular communication in the nervous system occurs through the release of neurotransmitters into the synaptic cleft between neurons. In the presynaptic neuron, the proton pumping vesicular- or vacuolar-type ATPase (V-ATPase) powers neurotransmitter loading into synaptic vesicles (SVs), with the V1 complex dissociating from the membrane region of the enzyme before exocytosis. We isolated SVs from rat brain using SidK, a V-ATPase-binding bacterial effector protein. Single-particle electron cryomicroscopy allowed high-resolution structure determination of V-ATPase within the native SV membrane. In the structure, regularly spaced cholesterol molecules decorate the enzyme's rotor and the abundant SV protein synaptophysin binds the complex stoichiometrically. ATP hydrolysis during vesicle loading results in a loss of the V1 region of V-ATPase from the SV membrane, suggesting that loading is sufficient to induce dissociation of the enzyme.


Asunto(s)
Vesículas Sinápticas , ATPasas de Translocación de Protón Vacuolares , Animales , Ratas , Proteínas Bacterianas/química , Encéfalo/ultraestructura , Encéfalo/enzimología , Colesterol/química , Microscopía por Crioelectrón , Hidrólisis , Vesículas Sinápticas/enzimología , Vesículas Sinápticas/ultraestructura , Sinaptofisina/metabolismo , ATPasas de Translocación de Protón Vacuolares/química , ATPasas de Translocación de Protón Vacuolares/aislamiento & purificación , ATPasas de Translocación de Protón Vacuolares/ultraestructura , Conformación Proteica
2.
ACS Omega ; 8(21): 19081-19098, 2023 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-37273644

RESUMEN

Q203 (telacebec) is an imidazopyridine amide (IPA) targeting the respiratory CIII2CIV2 supercomplex of the mycobacterial electron transport chain (ETC). Aiming for a better understanding of the molecular mechanism of action of IPA, 27 analogues were prepared through a seven-step synthetic scheme. Oxygen consumption assay was designed to test the inhibition of purified Mycobacterium smegmatis CIII2CIV2 by these compounds. The assay results generally supported structure-activity relationship information obtained from the structure of M. smegmatis CIII2CIV2 bound to Q203. The IC50 of Q203 and compound 27 was 99 ± 32 and 441 ± 138 nM, respectively. All IPAs including Q203 showed no inhibition of mitochondrial ETC, proving their selectivity against mycobacteria. In vitro Mycobacterium tuberculosis growth inhibition and M. smegmatis CIII2CIV2 binding did not correlate perfectly. These observations suggest that further investigation into the mechanisms of resistance in different mycobacterial species is needed to understand the lack of the correlation pattern between CIII2CIV2 inhibition and cellular activity.

3.
Proc Natl Acad Sci U S A ; 120(13): e2214949120, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36952383

RESUMEN

Oxidative phosphorylation, the combined activity of the electron transport chain (ETC) and adenosine triphosphate synthase, has emerged as a valuable target for the treatment of infection by Mycobacterium tuberculosis and other mycobacteria. The mycobacterial ETC is highly branched with multiple dehydrogenases transferring electrons to a membrane-bound pool of menaquinone and multiple oxidases transferring electrons from the pool. The proton-pumping type I nicotinamide adenine dinucleotide (NADH) dehydrogenase (Complex I) is found in low abundance in the plasma membranes of mycobacteria in typical in vitro culture conditions and is often considered dispensable. We found that growth of Mycobacterium smegmatis in carbon-limited conditions greatly increased the abundance of Complex I and allowed isolation of a rotenone-sensitive preparation of the enzyme. Determination of the structure of the complex by cryoEM revealed the "orphan" two-component response regulator protein MSMEG_2064 as a subunit of the assembly. MSMEG_2064 in the complex occupies a site similar to the proposed redox-sensing subunit NDUFA9 in eukaryotic Complex I. An apparent purine nucleoside triphosphate within the NuoG subunit resembles the GTP-derived molybdenum cofactor in homologous formate dehydrogenase enzymes. The membrane region of the complex binds acyl phosphatidylinositol dimannoside, a characteristic three-tailed lipid from the mycobacterial membrane. The structure also shows menaquinone, which is preferentially used over ubiquinone by gram-positive bacteria, in two different positions along the quinone channel, comparable to ubiquinone in other structures and suggesting a conserved quinone binding mechanism.


Asunto(s)
Complejo I de Transporte de Electrón , Ubiquinona , Complejo I de Transporte de Electrón/metabolismo , Ubiquinona/metabolismo , Vitamina K 2 , Quinonas/metabolismo , Mycobacterium smegmatis/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(6): e2217181120, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36724250

RESUMEN

Vacuolar-type adenosine triphosphatases (V-ATPases) are rotary proton pumps that acidify specific intracellular compartments in almost all eukaryotic cells. These multi-subunit enzymes consist of a soluble catalytic V1 region and a membrane-embedded proton-translocating VO region. VO is assembled in the endoplasmic reticulum (ER) membrane, and V1 is assembled in the cytosol. However, V1 binds VO only after VO is transported to the Golgi membrane, thereby preventing acidification of the ER. We isolated VO complexes and subcomplexes from Saccharomyces cerevisiae bound to V-ATPase assembly factors Vma12p, Vma21p, and Vma22p. Electron cryomicroscopy shows how the Vma12-22p complex recruits subunits a, e, and f to the rotor ring of VO while blocking premature binding of V1. Vma21p, which contains an ER-retrieval motif, binds the VO:Vma12-22p complex, "mature" VO, and a complex that appears to contain a ring of loosely packed rotor subunits and the proteins YAR027W and YAR028W. The structures suggest that Vma21p binds assembly intermediates that contain a rotor ring and that activation of proton pumping following assembly of V1 with VO removes Vma21p, allowing V-ATPase to remain in the Golgi. Together, these structures show how Vma12-22p and Vma21p function in V-ATPase assembly and quality control, ensuring the enzyme acidifies only its intended cellular targets.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , ATPasas de Translocación de Protón Vacuolares , Proteínas de Saccharomyces cerevisiae/metabolismo , Protones , Proteínas de la Membrana/metabolismo , Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
5.
Life Sci Alliance ; 5(11)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35794005

RESUMEN

V-ATPases are rotary proton pumps that serve as signaling hubs with numerous protein binding partners. CryoEM with exhaustive focused classification allowed detection of endogenous proteins associated with porcine kidney V-ATPase. An extra C subunit was found in ∼3% of complexes, whereas ∼1.6% of complexes bound mEAK-7, a protein with proposed roles in dauer formation in nematodes and mTOR signaling in mammals. High-resolution cryoEM of porcine kidney V-ATPase with recombinant mEAK-7 showed that mEAK-7's TLDc domain interacts with V-ATPase's stator, whereas its C-terminal α helix binds V-ATPase's rotor. This crosslink would be expected to inhibit rotary catalysis. However, unlike the yeast TLDc protein Oxr1p, exogenous mEAK-7 does not inhibit V-ATPase and mEAK-7 overexpression in cells does not alter lysosomal or phagosomal pH. Instead, cryoEM suggests that the mEAK-7:V-ATPase interaction is disrupted by ATP-induced rotation of the rotor. Comparison of Oxr1p and mEAK-7 binding explains this difference. These results show that V-ATPase binding by TLDc domain proteins can lead to effects ranging from strong inhibition to formation of labile interactions that are sensitive to the enzyme's activity.


Asunto(s)
ATPasas de Translocación de Protón Vacuolares , Animales , Microscopía por Crioelectrón , Mamíferos/metabolismo , Unión Proteica , Subunidades de Proteína/química , Porcinos , ATPasas de Translocación de Protón Vacuolares/química , ATPasas de Translocación de Protón Vacuolares/metabolismo
6.
Nat Struct Mol Biol ; 29(5): 430-439, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35469063

RESUMEN

Vacuolar-type ATPases (V-ATPases) are rotary enzymes that acidify intracellular compartments in eukaryotic cells. These multi-subunit complexes consist of a cytoplasmic V1 region that hydrolyzes ATP and a membrane-embedded VO region that transports protons. V-ATPase activity is regulated by reversible dissociation of the two regions, with the isolated V1 and VO complexes becoming autoinhibited on disassembly and subunit C subsequently detaching from V1. In yeast, assembly of the V1 and VO regions is mediated by the regulator of the ATPase of vacuoles and endosomes (RAVE) complex through an unknown mechanism. We used cryogenic-electron microscopy of yeast V-ATPase to determine structures of the intact enzyme, the dissociated but complete V1 complex and the V1 complex lacking subunit C. On separation, V1 undergoes a dramatic conformational rearrangement, with its rotational state becoming incompatible for reassembly with VO. Loss of subunit C allows V1 to match the rotational state of VO, suggesting how RAVE could reassemble V1 and VO by recruiting subunit C.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , ATPasas de Translocación de Protón Vacuolares , Endosomas/metabolismo , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/química , Vacuolas/metabolismo
7.
Elife ; 102021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34590581

RESUMEN

The imidazopyridine telacebec, also known as Q203, is one of only a few new classes of compounds in more than 50 years with demonstrated antituberculosis activity in humans. Telacebec inhibits the mycobacterial respiratory supercomplex composed of complexes III and IV (CIII2CIV2). In mycobacterial electron transport chains, CIII2CIV2 replaces canonical CIII and CIV, transferring electrons from the intermediate carrier menaquinol to the final acceptor, molecular oxygen, while simultaneously transferring protons across the inner membrane to power ATP synthesis. We show that telacebec inhibits the menaquinol:oxygen oxidoreductase activity of purified Mycobacterium smegmatis CIII2CIV2 at concentrations similar to those needed to inhibit electron transfer in mycobacterial membranes and Mycobacterium tuberculosis growth in culture. We then used electron cryomicroscopy (cryoEM) to determine structures of CIII2CIV2 both in the presence and absence of telacebec. The structures suggest that telacebec prevents menaquinol oxidation by blocking two different menaquinol binding modes to prevent CIII2CIV2 activity.


Asunto(s)
Antituberculosos/farmacología , Imidazoles/farmacología , Mycobacterium smegmatis/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Piperidinas/farmacología , Piridinas/farmacología , Transporte de Electrón , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/metabolismo , Oxidación-Reducción
8.
Biochemistry ; 60(23): 1808-1821, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34080844

RESUMEN

Tuberous sclerosis protein complex (pTSC) nucleates a proteinaceous signaling hub that integrates information about the internal and external energy status of the cell in the regulation of growth and energy consumption. Biochemical and cryo-electron microscopy studies of recombinant pTSC have revealed its structure and stoichiometry and hinted at the possibility that the complex may form large oligomers. Here, we have partially purified endogenous pTSC from fasted mammalian brains of rat and pig by leveraging a recombinant antigen binding fragment (Fab) specific for the TSC2 subunit of pTSC. We demonstrate Fab-dependent purification of pTSC from membrane-solubilized fractions of the brain homogenates. Negative stain electron microscopy of the samples purified from pig brain demonstrates rod-shaped protein particles with a width of 10 nm, a variable length as small as 40 nm, and a high degree of conformational flexibility. Larger filaments are evident with a similar 10 nm width and a ≤1 µm length in linear and weblike organizations prepared from pig brain. Immunogold labeling experiments demonstrate linear aggregates of pTSC purified from mammalian brains. These observations suggest polymerization of endogenous pTSC into filamentous superstructures.


Asunto(s)
Proteína 2 del Complejo de la Esclerosis Tuberosa/química , Proteína 2 del Complejo de la Esclerosis Tuberosa/ultraestructura , Esclerosis Tuberosa/metabolismo , Animales , Microscopía por Crioelectrón/métodos , Citoesqueleto/metabolismo , Humanos , Unión Proteica/fisiología , Ratas , Proteínas Recombinantes/metabolismo , Transducción de Señal/genética , Porcinos , Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteínas Supresoras de Tumor/metabolismo
9.
Nat Commun ; 12(1): 3661, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34135340

RESUMEN

SARS-CoV-2, the virus responsible for COVID-19, has caused a global pandemic. Antibodies can be powerful biotherapeutics to fight viral infections. Here, we use the human apoferritin protomer as a modular subunit to drive oligomerization of antibody fragments and transform antibodies targeting SARS-CoV-2 into exceptionally potent neutralizers. Using this platform, half-maximal inhibitory concentration (IC50) values as low as 9 × 10-14 M are achieved as a result of up to 10,000-fold potency enhancements compared to corresponding IgGs. Combination of three different antibody specificities and the fragment crystallizable (Fc) domain on a single multivalent molecule conferred the ability to overcome viral sequence variability together with outstanding potency and IgG-like bioavailability. The MULTi-specific, multi-Affinity antiBODY (Multabody or MB) platform thus uniquely leverages binding avidity together with multi-specificity to deliver ultrapotent and broad neutralizers against SARS-CoV-2. The modularity of the platform also makes it relevant for rapid evaluation against other infectious diseases of global health importance. Neutralizing antibodies are a promising therapeutic for SARS-CoV-2.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , SARS-CoV-2/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Apoferritinas/química , Disponibilidad Biológica , Mapeo Epitopo , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ingeniería de Proteínas/métodos , Subunidades de Proteína/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Distribución Tisular
10.
Nature ; 589(7840): 143-147, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33299175

RESUMEN

Tuberculosis-the world's leading cause of death by infectious disease-is increasingly resistant to current first-line antibiotics1. The bacterium Mycobacterium tuberculosis (which causes tuberculosis) can survive low-energy conditions, allowing infections to remain dormant and decreasing their susceptibility to many antibiotics2. Bedaquiline was developed in 2005 from a lead compound identified in a phenotypic screen against Mycobacterium smegmatis3. This drug can sterilize even latent M. tuberculosis infections4 and has become a cornerstone of treatment for multidrug-resistant and extensively drug-resistant tuberculosis1,5,6. Bedaquiline targets the mycobacterial ATP synthase3, which is an essential enzyme in the obligate aerobic Mycobacterium genus3,7, but how it binds the intact enzyme is unknown. Here we determined cryo-electron microscopy structures of M. smegmatis ATP synthase alone and in complex with bedaquiline. The drug-free structure suggests that hook-like extensions from the α-subunits prevent the enzyme from running in reverse, inhibiting ATP hydrolysis and preserving energy in hypoxic conditions. Bedaquiline binding induces large conformational changes in the ATP synthase, creating tight binding pockets at the interface of subunits a and c that explain the potency of this drug as an antibiotic for tuberculosis.


Asunto(s)
Complejos de ATP Sintetasa/química , Antituberculosos/química , Microscopía por Crioelectrón , Diarilquinolinas/química , Mycobacterium smegmatis/enzimología , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Complejos de ATP Sintetasa/antagonistas & inhibidores , Complejos de ATP Sintetasa/metabolismo , Adenosina Trifosfato/metabolismo , Antituberculosos/metabolismo , Antituberculosos/farmacología , Diarilquinolinas/metabolismo , Diarilquinolinas/farmacología , Hidrólisis/efectos de los fármacos , Modelos Moleculares , Mycobacterium smegmatis/efectos de los fármacos , Rotación
11.
Science ; 367(6483): 1240-1246, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32165585

RESUMEN

In neurons, the loading of neurotransmitters into synaptic vesicles uses energy from proton-pumping vesicular- or vacuolar-type adenosine triphosphatases (V-ATPases). These membrane protein complexes possess numerous subunit isoforms, which complicates their analysis. We isolated homogeneous rat brain V-ATPase through its interaction with SidK, a Legionella pneumophila effector protein. Cryo-electron microscopy allowed the construction of an atomic model, defining the enzyme's ATP:proton ratio as 3:10 and revealing a homolog of yeast subunit f in the membrane region, which we tentatively identify as RNAseK. The c ring encloses the transmembrane anchors for cleaved ATP6AP1/Ac45 and ATP6AP2/PRR, the latter of which is the (pro)renin receptor that, in other contexts, is involved in both Wnt signaling and the renin-angiotensin system that regulates blood pressure. This structure shows how ATP6AP1/Ac45 and ATP6AP2/PRR enable assembly of the enzyme's catalytic and membrane regions.


Asunto(s)
Biomarcadores/química , Encéfalo/enzimología , Receptores de Superficie Celular/química , ATPasas de Translocación de Protón Vacuolares/química , Animales , Proteínas Bacterianas/química , Biocatálisis , Membrana Celular/enzimología , Microscopía por Crioelectrón , Modelos Químicos , Dominios Proteicos , Ratas , Sistema Renina-Angiotensina , Vía de Señalización Wnt
12.
Proc Natl Acad Sci U S A ; 116(15): 7272-7277, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30910982

RESUMEN

Proton-translocating vacuolar-type ATPases (V-ATPases) are necessary for numerous processes in eukaryotic cells, including receptor-mediated endocytosis, protein maturation, and lysosomal acidification. In mammals, V-ATPase subunit isoforms are differentially targeted to various intracellular compartments or tissues, but how these subunit isoforms influence enzyme activity is not clear. In the yeast Saccharomyces cerevisiae, isoform diversity is limited to two different versions of the proton-translocating subunit a: Vph1p, which is targeted to the vacuole, and Stv1p, which is targeted to the Golgi apparatus and endosomes. We show that purified V-ATPase complexes containing Vph1p have higher ATPase activity than complexes containing Stv1p and that the relative difference in activity depends on the presence of lipids. We also show that VO complexes containing Stv1p could be readily purified without attached V1 regions. We used this effect to determine structures of the membrane-embedded VO region with Stv1p at 3.1-Å resolution, which we compare with a structure of the VO region with Vph1p that we determine to 3.2-Å resolution. These maps reveal differences in the surface charge near the cytoplasmic proton half-channel. Both maps also show the presence of bound lipids, as well as regularly spaced densities that may correspond to ergosterol or bound detergent, around the c-ring.

13.
Science ; 358(6365): 936-940, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-29074581

RESUMEN

Mitochondrial adenosine triphosphate (ATP) synthase produces the majority of ATP in eukaryotic cells, and its dimerization is necessary to create the inner membrane folds, or cristae, characteristic of mitochondria. Proton translocation through the membrane-embedded FO region turns the rotor that drives ATP synthesis in the soluble F1 region. Although crystal structures of the F1 region have illustrated how this rotation leads to ATP synthesis, understanding how proton translocation produces the rotation has been impeded by the lack of an experimental atomic model for the FO region. Using cryo-electron microscopy, we determined the structure of the dimeric FO complex from Saccharomyces cerevisiae at a resolution of 3.6 angstroms. The structure clarifies how the protons travel through the complex, how the complex dimerizes, and how the dimers bend the membrane to produce cristae.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales/química , Modelos Moleculares , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Microscopía por Crioelectrón , Cristalografía por Rayos X , Mitocondrias/enzimología , Pliegue de Proteína , Multimerización de Proteína , Estructura Secundaria de Proteína
14.
PLoS Pathog ; 13(6): e1006394, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28570695

RESUMEN

Intracellular pathogenic bacteria evade the immune response by replicating within host cells. Legionella pneumophila, the causative agent of Legionnaires' Disease, makes use of numerous effector proteins to construct a niche supportive of its replication within phagocytic cells. The L. pneumophila effector SidK was identified in a screen for proteins that reduce the activity of the proton pumping vacuolar-type ATPases (V-ATPases) when expressed in the yeast Saccharomyces cerevisae. SidK is secreted by L. pneumophila in the early stages of infection and by binding to and inhibiting the V-ATPase, SidK reduces phagosomal acidification and promotes survival of the bacterium inside macrophages. We determined crystal structures of the N-terminal region of SidK at 2.3 Å resolution and used single particle electron cryomicroscopy (cryo-EM) to determine structures of V-ATPase:SidK complexes at ~6.8 Å resolution. SidK is a flexible and elongated protein composed of an α-helical region that interacts with subunit A of the V-ATPase and a second region of unknown function that is flexibly-tethered to the first. SidK binds V-ATPase strongly by interacting via two α-helical bundles at its N terminus with subunit A. In vitro activity assays show that SidK does not inhibit the V-ATPase completely, but reduces its activity by ~40%, consistent with the partial V-ATPase deficiency phenotype its expression causes in yeast. The cryo-EM analysis shows that SidK reduces the flexibility of the A-subunit that is in the 'open' conformation. Fluorescence experiments indicate that SidK binding decreases the affinity of V-ATPase for a fluorescent analogue of ATP. Together, these results reveal the structural basis for the fine-tuning of V-ATPase activity by SidK.


Asunto(s)
Proteínas Bacterianas/metabolismo , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/microbiología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Regulación Enzimológica de la Expresión Génica , Humanos , Legionella pneumophila/química , Legionella pneumophila/genética , Enfermedad de los Legionarios/enzimología , Enfermedad de los Legionarios/genética , Conformación Proteica , ATPasas de Translocación de Protón Vacuolares/química , ATPasas de Translocación de Protón Vacuolares/genética
15.
Nature ; 539(7627): 118-122, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27776355

RESUMEN

Vacuolar-type ATPases (V-ATPases) are ATP-powered proton pumps involved in processes such as endocytosis, lysosomal degradation, secondary transport, TOR signalling, and osteoclast and kidney function. ATP hydrolysis in the soluble catalytic V1 region drives proton translocation through the membrane-embedded VO region via rotation of a rotor subcomplex. Variability in the structure of the intact enzyme has prevented construction of an atomic model for the membrane-embedded motor of any rotary ATPase. We induced dissociation and auto-inhibition of the V1 and VO regions of the V-ATPase by starving the yeast Saccharomyces cerevisiae, allowing us to obtain a ~3.9-Šresolution electron cryomicroscopy map of the VO complex and build atomic models for the majority of its subunits. The analysis reveals the structures of subunits ac8c'c″de and a protein that we identify and propose to be a new subunit (subunit f). A large cavity between subunit a and the c-ring creates a cytoplasmic half-channel for protons. The c-ring has an asymmetric distribution of proton-carrying Glu residues, with the Glu residue of subunit c″ interacting with Arg735 of subunit a. The structure suggests sequential protonation and deprotonation of the c-ring, with ATP-hydrolysis-driven rotation causing protonation of a Glu residue at the cytoplasmic half-channel and subsequent deprotonation of a Glu residue at a luminal half-channel.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura , Saccharomyces cerevisiae/ultraestructura , ATPasas de Translocación de Protón Vacuolares/química , ATPasas de Translocación de Protón Vacuolares/ultraestructura , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Arginina/química , Arginina/metabolismo , Ácido Glutámico/química , Ácido Glutámico/metabolismo , Hidrólisis , Modelos Moleculares , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Protones , Rotación , Saccharomyces cerevisiae/química
16.
ACS Chem Biol ; 10(11): 2616-23, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26352211

RESUMEN

Most existing antibiotics were discovered through screens of environmental microbes, particularly the streptomycetes, for the capacity to prevent the growth of pathogenic bacteria. This "activity-guided screening" method has been largely abandoned because it repeatedly rediscovers those compounds that are highly expressed during laboratory culture. Most of these metabolites have already been biochemically characterized. However, the sequencing of streptomycete genomes has revealed a large number of "cryptic" secondary metabolic genes that are either poorly expressed in the laboratory or that have biological activities that cannot be discovered through standard activity-guided screens. Methods that reveal these uncharacterized compounds, particularly methods that are not biased in favor of the highly expressed metabolites, would provide direct access to a large number of potentially useful biologically active small molecules. To address this need, we have devised a discovery method in which a chemical elicitor called Cl-ARC is used to elevate the expression of cryptic biosynthetic genes. We show that the resulting change in product yield permits the direct discovery of secondary metabolites without requiring knowledge of their biological activity. We used this approach to identify three rare secondary metabolites and find that two of them target eukaryotic cells and not bacterial cells. In parallel, we report the first paired use of cheminformatic inference and chemical genetic epistasis in yeast to identify the target. In this way, we demonstrate that oxohygrolidin, one of the eukaryote-active compounds we identified through activity-independent screening, targets the V1 ATPase in yeast and human cells and secondarily HSP90.


Asunto(s)
Acetanilidas/química , Actinobacteria/química , Actinobacteria/metabolismo , Antibacterianos/biosíntesis , Antibacterianos/aislamiento & purificación , Productos Biológicos/química , Descubrimiento de Drogas/métodos , Macrólidos/química , Éteres Fenílicos/química , Acetanilidas/farmacología , Actinobacteria/genética , Actinobacteria/crecimiento & desarrollo , Antibacterianos/química , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Productos Biológicos/metabolismo , Cromatografía Liquida , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Macrólidos/farmacología , Éteres Fenílicos/farmacología
17.
Biochemistry ; 54(3): 853-8, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25546637

RESUMEN

Vacuolar-type ATPases (V-ATPases) acidify numerous intracellular compartments in all eukaryotic cells and are responsible for extracellular acidification in some specialized cells. V-ATPases are large macromolecular complexes with at least 15 different subunits, some of which are found in multiple copies. The main roles of all V-ATPase subunits have been established except for the e subunit, encoded by the gene VMA9 in Saccharomyces cerevisiae, and the Ac45 subunit, which is not found in the S. cerevisiae enzyme. Here we demonstrate that when the S. cerevisiae V-ATPase is solubilized with the detergent dodecylmaltoside (DDM), Vma9p is removed. We further demonstrate that after Vma9p has been removed by detergent the purified enzyme is still able to perform fully-coupled ATP-dependent proton pumping. This observation shows that Vma9p is not necessary in vitro for this principal activity of the V-ATPase.


Asunto(s)
Bombas de Protones/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Adenosina Trifosfato/metabolismo , Glucósidos/metabolismo , Hidrólisis , Proteínas Recombinantes de Fusión/metabolismo , Solubilidad
18.
J Biol Chem ; 288(22): 15913-25, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23589308

RESUMEN

α-Catenin is an actin- and vinculin-binding protein that regulates cell-cell adhesion by interacting with cadherin adhesion receptors through ß-catenin, but the mechanisms by which it anchors the cadherin-catenin complex to the actin cytoskeleton at adherens junctions remain unclear. Here we determined crystal structures of αE-catenin in the autoinhibited state and the actin-binding domain of αN-catenin. Together with the small-angle x-ray scattering analysis of full-length αN-catenin, we deduced an elongated multidomain assembly of monomeric α-catenin that structurally and functionally couples the vinculin- and actin-binding mechanisms. Cellular and biochemical studies of αE- and αN-catenins show that αE-catenin recruits vinculin to adherens junctions more effectively than αN-catenin, partly because of its higher affinity for actin filaments. We propose a molecular switch mechanism involving multistate conformational changes of α-catenin. This would be driven by actomyosin-generated tension to dynamically regulate the vinculin-assisted linkage between adherens junctions and the actin cytoskeleton.


Asunto(s)
Citoesqueleto de Actina , Uniones Adherentes , Modelos Biológicos , Proteínas del Tejido Nervioso , Vinculina , alfa Catenina , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Uniones Adherentes/química , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Animales , Línea Celular , Cristalografía por Rayos X , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Vinculina/química , Vinculina/genética , Vinculina/metabolismo , alfa Catenina/química , alfa Catenina/genética , alfa Catenina/metabolismo
19.
J Biol Chem ; 288(8): 5896-913, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23288846

RESUMEN

Previously, we reported an acidification-dependent interaction of the endosomal vacuolar H(+)-ATPase (V-ATPase) with cytohesin-2, a GDP/GTP exchange factor (GEF), suggesting that it functions as a pH-sensing receptor. Here, we have studied the molecular mechanism of signaling between the V-ATPase, cytohesin-2, and Arf GTP-binding proteins. We found that part of the N-terminal cytosolic tail of the V-ATPase a2-subunit (a2N), corresponding to its first 17 amino acids (a2N(1-17)), potently modulates the enzymatic GDP/GTP exchange activity of cytohesin-2. Moreover, this peptide strongly inhibits GEF activity via direct interaction with the Sec7 domain of cytohesin-2. The structure of a2N(1-17) and its amino acids Phe(5), Met(10), and Gln(14) involved in interaction with Sec7 domain were determined by NMR spectroscopy analysis. In silico docking experiments revealed that part of the V-ATPase formed by its a2N(1-17) epitope competes with the switch 2 region of Arf1 and Arf6 for binding to the Sec7 domain of cytohesin-2. The amino acid sequence alignment and GEF activity studies also uncovered the conserved character of signaling between all four (a1-a4) a-subunit isoforms of mammalian V-ATPase and cytohesin-2. Moreover, the conserved character of this phenomenon was also confirmed in experiments showing binding of mammalian cytohesin-2 to the intact yeast V-ATPase holo-complex. Thus, here we have uncovered an evolutionarily conserved function of the V-ATPase as a novel cytohesin-signaling receptor.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Secuencia de Aminoácidos , Animales , Dicroismo Circular , ADN Complementario/metabolismo , Epítopos/química , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética/métodos , Ratones , Microscopía Confocal/métodos , Datos de Secuencia Molecular , Péptidos/química , Isoformas de Proteínas , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes/química , Transducción de Señal , Triptófano/química
20.
Nat Struct Mol Biol ; 19(12): 1356-62, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23142977

RESUMEN

Vacuolar-type ATPases (V-type ATPases) in eukaryotic cells are large membrane protein complexes that acidify various intracellular compartments. The enzymes are regulated by dissociation of the V(1) and V(O) regions of the complex. Here we present the structure of the Saccharomyces cerevisiae V-type ATPase at 11-Å resolution by cryo-EM of protein particles in ice. The structure explains many cross-linking and protein interaction studies. Docking of crystal structures suggests that inhibition of ATPase activity by the dissociated V(1) region involves rearrangement of the N- and C-terminal domains of subunit H and also suggests how this inhibition is triggered upon dissociation. We provide support for this model by demonstrating that mutation of subunit H to increase the rigidity of the linker between its two domains decreases its ability to inhibit ATPase activity.


Asunto(s)
Saccharomyces cerevisiae/enzimología , ATPasas de Translocación de Protón Vacuolares/química , Microscopía por Crioelectrón , Conformación Proteica , ATPasas de Translocación de Protón Vacuolares/metabolismo , ATPasas de Translocación de Protón Vacuolares/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...