Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Rev Mol Cell Biol ; 16(6): 375-88, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25991374

RESUMO

The mitochondrial respiratory chain, also known as the electron transport chain (ETC), is crucial to life, and energy production in the form of ATP is the main mitochondrial function. Three proton-translocating enzymes of the ETC, namely complexes I, III and IV, generate proton motive force, which in turn drives ATP synthase (complex V). The atomic structures and basic mechanisms of most respiratory complexes have previously been established, with the exception of complex I, the largest complex in the ETC. Recently, the crystal structure of the entire complex I was solved using a bacterial enzyme. The structure provided novel insights into the core architecture of the complex, the electron transfer and proton translocation pathways, as well as the mechanism that couples these two processes.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias , Complexo III da Cadeia de Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons , Complexo I de Transporte de Elétrons , Força Próton-Motriz/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Complexo I de Transporte de Elétrons/química , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Estrutura Quaternária de Proteína
2.
Proc Natl Acad Sci U S A ; 116(13): 6329-6334, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30850546

RESUMO

The ancient reductive acetyl-CoA pathway is employed by acetogenic bacteria to form acetate from inorganic energy sources. Since the central pathway does not gain net ATP by substrate-level phosphorylation, chemolithoautotrophic growth relies on the additional formation of ATP via a chemiosmotic mechanism. Genome analyses indicated that some acetogens only have an energy-converting, ion-translocating hydrogenase (Ech) as a potential respiratory enzyme. Although the Ech-encoding genes are widely distributed in nature, the proposed function of Ech as an ion-translocating chemiosmotic coupling site has neither been demonstrated in bacteria nor has it been demonstrated that it can be the only energetic coupling sites in microorganisms that depend on a chemiosmotic mechanism for energy conservation. Here, we show that the Ech complex of the thermophilic acetogenic bacterium Thermoanaerobacter kivui is indeed a respiratory enzyme. Experiments with resting cells prepared from T. kivui cultures grown on carbon monoxide (CO) revealed CO oxidation coupled to H2 formation and the generation of a transmembrane electrochemical ion gradient ([Formula: see text]). Inverted membrane vesicles (IMVs) prepared from CO-grown cells also produced H2 and ATP from CO (via a loosely attached CO dehydrogenase) or a chemical reductant. Finally, we show that Ech activity led to the translocation of both H+ and Na+ across the membrane of the IMVs. The H+ gradient was then used by the ATP synthase for energy conservation. These data demonstrate that the energy-converting hydrogenase in concert with an ATP synthase may be the simplest form of respiration; it combines carbon dioxide fixation with the synthesis of ATP in an ancient pathway.


Assuntos
Fenômenos Bioquímicos , Redes e Vias Metabólicas , Oxirredutases/metabolismo , Força Próton-Motriz/fisiologia , Thermoanaerobacter/metabolismo , Trifosfato de Adenosina/metabolismo , Aldeído Oxirredutases/metabolismo , Ciclo do Carbono , Monóxido de Carbono/metabolismo , Membrana Celular/metabolismo , Hidrogênio/metabolismo , Complexos Multienzimáticos/metabolismo , Família Multigênica , Oxirredução , Vesículas Secretórias/metabolismo , Sódio/metabolismo , Thermoanaerobacter/enzimologia , Thermoanaerobacter/genética
3.
J Bacteriol ; 202(2)2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31636109

RESUMO

Filamentous phages are nonlytic viruses that specifically infect bacteria, establishing a persistent association with their host. The phage particle has no machinery for generating energy and parasitizes its host's existing structures in order to cross the bacterial envelope and deliver its genetic material. The import of filamentous phages across the bacterial periplasmic space requires some of the components of a macrocomplex of the envelope known as the Tol system. This complex uses the energy provided by the proton motive force (pmf) of the inner membrane to perform essential and highly energy-consuming functions of the cell, such as envelope integrity maintenance and cell division. It has been suggested that phages take advantage of pmf-driven conformational changes in the Tol system to transit across the periplasm. However, this hypothesis has not been formally tested. In order to decouple the role of the Tol system in cell physiology and during phage parasitism, we used mutations on conserved essential residues known for inactivating pmf-dependent functions of the Tol system. We identified impaired Tol complexes that remain fully efficient for filamentous phage uptake. We further demonstrate that the TolQ-TolR homologous motor ExbB-ExbD, normally operating with the TonB protein, is able to promote phage infection along with full-length TolA.IMPORTANCE Filamentous phages are widely distributed symbionts of Gram-negative bacteria, with some of them being linked to genome evolution and virulence of their host. However, the precise mechanism that permits their uptake across the cell envelope is poorly understood. The canonical phage model Fd requires the TolQRA protein complex in the host envelope, which is suspected to translocate protons across the inner membrane. In this study, we show that phage uptake proceeds in the presence of the assembled but nonfunctional TolQRA complex. Moreover, our results unravel an alternative route for phage import that relies on the ExbB-ExbD proteins. This work provides new insights into the fundamental mechanisms of phage infection and might be generalized to other filamentous phages responsible for pathogen emergence.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriófagos/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Membrana/genética , Força Próton-Motriz/genética , Força Próton-Motriz/fisiologia
4.
J Biol Chem ; 294(13): 4867-4877, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30683696

RESUMO

The yeast mitochondrial proteins Rcf1 and Rcf2 are associated with a subpopulation of the cytochrome bc1-cytochrome c oxidase supercomplex and have been proposed to play a role in the assembly and/or modulation of the activity of the cytochrome c oxidase (complex IV, CIV). Yeast mutants deficient in either Rcf1 or Rcf2 proteins can use aerobic respiration-based metabolism for growth, but the absence of both proteins results in a strong growth defect. In this study, using assorted biochemical and biophysical analyses of Rcf1/Rcf2 single and double null-mutant yeast cells and mitochondria, we further explored how Rcf1 and Rcf2 support aerobic respiration and growth. We show that the absence of Rcf1 physically reduces the levels of CIV and diminishes the ability of the CIV that is present to maintain a normal mitochondrial proton motive force (PMF). Although the absence of Rcf2 did not noticeably affect the physical content of CIV, the PMF generated by CIV was also lower than normal. Our results indicate that the detrimental effects of the absence of Rcf1 and Rcf2 proteins on the CIV complex are distinct in terms of CIV assembly/accumulation and additive in terms of the ability of CIV to generate PMF. Thus, the combined absence of Rcf1 and Rcf2 alters both CIV physiology and assembly. We conclude that the slow aerobic growth of the Rcf1/Rcf2 double null mutant results from diminished generation of mitochondrial PMF by CIV and limits the level of CIV activity required for maintenance of the PMF and growth under aerobic conditions.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Consumo de Oxigênio/fisiologia , Força Próton-Motriz/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Complexo IV da Cadeia de Transporte de Elétrons/genética , Mutação , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
5.
J Bacteriol ; 201(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30692169

RESUMO

Bacterial type 4 pili (T4P) belong to the strongest molecular machines. The gonococcal T4P retraction ATPase PilT supports forces exceeding 100 pN during T4P retraction. Here, we address the question of whether gonococcal T4P retract in the absence of PilT. We show that pilT deletion strains indeed retract their T4P, but the maximum force is reduced to 5 pN. Similarly, the speed of T4P retraction is lower by orders of magnitude compared to that of T4P retraction driven by PilT. Deleting the pilT paralogue pilT2 further reduces the speed of T4P retraction, yet T4P retraction is detectable in the absence of all three pilT paralogues. Furthermore, we show that depletion of proton motive force (PMF) slows but does not inhibit pilT-independent T4P retraction. We conclude that the retraction ATPase is not essential for gonococcal T4P retraction. However, the force generated in the absence of PilT is too low to support important functions of T4P, including twitching motility, fluidization of colonies, and induction of host cell response.IMPORTANCE Bacterial type 4 pili (T4P) have been termed the "Swiss Army knives" of bacteria because they perform numerous functions, including host cell interaction, twitching motility, colony formation, DNA uptake, protein secretion, and surface sensing. The pilus fiber continuously elongates or retracts, and these dynamics are functionally important. Curiously, only a subset of T4P systems employ T4P retraction ATPases to power T4P retraction. Here, we show that one of the strongest T4P machines, the gonococcal T4P, retracts without a retraction ATPase. Biophysical characterization reveals strongly reduced force and speed compared to retraction with ATPase. We propose that bacteria encode retraction ATPases when T4P have to generate high-force-supporting functions like twitching motility, triggering host cell response, or fluidizing colonies.


Assuntos
Proteínas de Bactérias/metabolismo , Fímbrias Bacterianas/metabolismo , Proteínas Motores Moleculares/metabolismo , Neisseria gonorrhoeae/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Fímbrias/metabolismo , Força Próton-Motriz/fisiologia
6.
Mol Microbiol ; 105(4): 637-651, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28586527

RESUMO

The generation of a membrane potential (Δψ), the major constituent of the proton motive force (pmf), is crucial for ATP synthesis, transport of nutrients and flagellar rotation. Campylobacter jejuni harbors a branched electron transport chain, enabling respiration with different electron donors and acceptors. Here, we demonstrate that a relatively high Δψ is only generated in the presence of either formate as electron donor or oxygen as electron acceptor, in combination with an acceptor/donor respectively. We show the necessity of the pmf for motility and growth of C. jejuni. ATP generation is not only accomplished by oxidative phosphorylation via the pmf, but also by substrate-level phosphorylation via the enzyme AckA. In response to a low oxygen tension, C. jejuni increases the transcription and activity of the donor complexes formate dehydrogenase (FdhABC) and hydrogenase (HydABCD) as well as the transcription of the alternative respiratory acceptor complexes. Our findings suggest that in the gut of warm-blooded animals, C. jejuni depends on at least formate or hydrogen as donor (in the anaerobic lumen) or oxygen as acceptor (near the epithelial cells) to generate a pmf that sustains efficient motility and growth for colonization and pathogenesis.


Assuntos
Campylobacter jejuni/metabolismo , Força Próton-Motriz/fisiologia , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Formiatos/metabolismo , Hidrogênio , Potenciais da Membrana , Oxirredução , Oxigênio , Fosforilação
7.
J Membr Biol ; 251(3): 329-343, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29330604

RESUMO

This review focusses on the energetics of protein translocation via the Sec translocation machinery. First we complement structural data about SecYEG's conformational rearrangements by insight obtained from functional assays. These include measurements of SecYEG permeability that allow assessment of channel gating by ligand binding and membrane voltage. Second we will discuss the power stroke and Brownian ratcheting models of substrate translocation and the role that the two models assign to the putative driving forces: (i) ATP (SecA) and GTP (ribosome) hydrolysis, (ii) interaction with accessory proteins, (iii) membrane partitioning and folding, (iv) proton motive force (PMF), and (v) entropic contributions. Our analysis underlines how important energized membranes are for unravelling the translocation mechanism in future experiments.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Canais de Translocação SEC/química , Canais de Translocação SEC/metabolismo , Trifosfato de Adenosina/metabolismo , Eletrofisiologia , Guanosina Trifosfato/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Methanocaldococcus/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Transporte Proteico/fisiologia , Força Próton-Motriz/fisiologia
8.
Photosynth Res ; 138(2): 207-218, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30056561

RESUMO

In chloroplast, proton motive force (pmf) is critical for ATP synthesis and photoprotection. To prevent photoinhibition of photosynthetic apparatus, proton gradient (ΔpH) across the thylakoid membranes needs to be built up to minimize the production of reactive oxygen species (ROS) in thylakoid membranes. However, the regulation of thylakoid pmf in immature leaves is little known. In this study, we compared photosynthetic electron sinks, P700 redox state, non-photochemical quenching (NPQ), and electrochromic shift (ECS) signal in immature and mature leaves of a cultivar of Camellia. The immature leaves displayed lower linear electron flow and cyclic electron flow, but higher levels of NPQ and P700 oxidation ratio under high light. Meanwhile, we found that pmf and ΔpH were higher in the immature leaves. Furthermore, the immature leaves showed significantly lower thylakoid proton conductivity than mature leaves. These results strongly indicated that immature leaves can build up enough ΔpH by modulating proton efflux from the lumenal side to the stromal side of thylakoid membranes, which is essential to prevent photoinhibition via thermal energy dissipation and photosynthetic control of electron transfer. This study highlights that the activity of chloroplast ATP synthase is a key safety valve for photoprotection in immature leaves.


Assuntos
Camellia/fisiologia , Folhas de Planta , Força Próton-Motriz/fisiologia , Tilacoides , Camellia/classificação , Clorofila/fisiologia , Fluorescência , Fenótipo , Fotossíntese/fisiologia , Complexo de Proteína do Fotossistema I/fisiologia , Complexo de Proteína do Fotossistema II/fisiologia , Folhas de Planta/fisiologia , Transpiração Vegetal , ATPases Translocadoras de Prótons/fisiologia , Tilacoides/fisiologia
9.
AAPS PharmSciTech ; 19(7): 2898-2907, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30209787

RESUMO

This study described a pH-gradient dissolution method combined with flux measurements as an in vitro tool for assessing the risk of bioavailability reduction due to drug-drug interactions (DDI) caused by acid reducing agents (ARAs). The device incorporates absorption chambers into USP II dissolution vessels, with fiber optic UV-probes monitoring concentration in situ. Dosage forms of Genentech BCS class II drugs, GDC-0810, GDC-0941, and compound A, were tested by starting the dissolution in either pH 1.6 or pH 4.0 media then converting to FaSSIF after 30 min. GDC-0810 showed no significant difference in flux between the two conversion experiments. A supersaturation phase was observed for GDC-0941 in the pH 1.6 experiments after media conversion to FaSSIF; however, it did not appear to occur in the pH 4.0 experiment due to low drug solubility at pH 4.0, resulting in a 95% decrease in flux compared to pH 1.6 experiment. The extent of flux reduction and the total accumulated API mass in the absorption chamber agreed well with the 89% reduction in mean Cmax and the 82% reduction in mean AUC from dog PK study between animals treated with pentagastrin and famotidine. Testing of the compound A optimized formulation tablets showed a 25% reduction in flux and in vitro absorbed amount by changing pH 1.6 to 4.0, correlating well with the AUC decrease in clinical studies. Good correlation between in vitro data and in vivo PK data demonstrated the applicability of the method for formulators to develop drug products mitigating DDI from ARAs.


Assuntos
Cinamatos/química , Cinamatos/farmacocinética , Indazóis/química , Indazóis/farmacocinética , Sulfonamidas/química , Sulfonamidas/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Cães , Interações Medicamentosas/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Força Próton-Motriz/efeitos dos fármacos , Força Próton-Motriz/fisiologia , Solubilidade , Comprimidos
10.
Biochim Biophys Acta ; 1857(3): 190-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26546715

RESUMO

Aerobic respiratory chains from all life kingdoms are composed by several complexes that have been deeply characterized in their isolated form. These membranous complexes link the oxidation of reducing substrates to the reduction of molecular oxygen, in a process that conserves energy by ion translocation between both sides of the mitochondrial or prokaryotic cytoplasmatic membranes. In recent years there has been increasing evidence that those complexes are organized as supramolecular structures, the so-called supercomplexes and respirasomes, being available for eukaryotes strong data namely obtained by electron microscopy and single particle analysis. A parallel study has been developed for prokaryotes, based on blue native gels and mass spectrometry analysis, showing that in these more simple unicellular organisms such supercomplexes also exist, involving not only typical aerobic-respiration associated complexes, but also anaerobic-linked enzymes. After a short overview of the data on eukaryotic supercomplexes, we will analyse comprehensively the different types of prokaryotic aerobic respiratory supercomplexes that have been thus far suggested, in both bacteria and archaea. This article is part of a Special Issue entitled Organization and dynamics of bioenergetic systems in bacteria, edited by Prof Conrad Mullineaux.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Membrana Celular/enzimologia , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Força Próton-Motriz/fisiologia , Aerobiose/fisiologia , Transporte de Elétrons/fisiologia
11.
Proc Natl Acad Sci U S A ; 111(19): E2027-36, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778208

RESUMO

Pathogenic Gram-negative bacteria use syringe-like type III secretion systems (T3SS) to inject effector proteins directly into targeted host cells. Effector secretion is triggered by host cell contact, and before contact is prevented by a set of conserved regulators. How these regulators interface with the T3SS apparatus to control secretion is unclear. We present evidence that the proton motive force (pmf) drives T3SS secretion in Pseudomonas aeruginosa, and that the cytoplasmic regulator PcrG interacts with distinct components of the T3SS apparatus to control two important aspects of effector secretion: (i) It coassembles with a second regulator (Pcr1) on the inner membrane T3SS component PcrD to prevent effectors from accessing the T3SS, and (ii) In conjunction with PscO, it controls protein secretion activity by modulating the ability of T3SS to convert pmf.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Citoplasma/metabolismo , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Fenótipo , Estrutura Terciária de Proteína , Força Próton-Motriz/fisiologia , Especificidade por Substrato
12.
J Bioenerg Biomembr ; 48(1): 87-96, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26749514

RESUMO

To elucidate the energy production mechanism of alkaliphiles, the relationship between the H(+) extrusion rate by the respiratory chain and the corresponding ATP synthesis rate was determined in the facultative alkaliphile Bacillus cohnii YN-2000 and compared with those in the obligate alkaliphile Bacillus clarkii DSM 8720(T) and the neutralophile Bacillus subtilis IAM 1026. Under high aeration condition, much higher ATP synthesis rates and larger Δψ in the alkaliphilic Bacillus spp. grown at pH 10 than those in the neutralophilic B. subtilis grown at pH 7 were observed. This high ATP productivity could be attributed to the larger Δψ in alkaliphiles than in B. subtilis because the H(+) extrusion rate in alkaliphiles cannot account for the high ATP productivity. However, the large Δψ in the alkaliphiles could not be explained only by the H(+) translocation rate in the respiratory chain in alkaliphiles. There is a possibility that the Donnan effect across the membrane has the potential to contribute to the large Δψ. To estimate the contribution of the Donnan effect to the large Δψ in alkaliphilic Bacillus spp. grown at pH 10, intracellular negative ion capacity was examined. The intracellular negative ion capacities in alkaliphiles grown at pH 10 under high aeration condition corresponding to their intracellular pH (pH 8.1) were much higher than those in alkaliphiles grown under low aeration condition. A proportional relationship is revealed between the negative ion capacity and Δψ in alkaliphiles grown under different aeration conditions. This relationship strongly suggests that the intracellular negative ion capacity contributes to the formation of Δψ through the Donnan effect in alkaliphilic Bacillus spp. grown at pH 10.


Assuntos
Trifosfato de Adenosina/metabolismo , Bacillus/metabolismo , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Potenciais da Membrana/fisiologia , Força Próton-Motriz/fisiologia , Transporte de Elétrons/fisiologia
13.
Proc Natl Acad Sci U S A ; 110(38): E3650-9, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-24003141

RESUMO

The twin-arginine translocation (Tat) machinery transports folded proteins across the cytoplasmic membrane of bacteria and the thylakoid membrane of chloroplasts. It has been inferred that the Tat translocation site is assembled on demand by substrate-induced association of the protein TatA. We tested this model by imaging YFP-tagged TatA expressed at native levels in living Escherichia coli cells in the presence of low levels of the TatA paralogue TatE. Under these conditions the TatA-YFP fusion supports full physiological Tat transport activity. In agreement with the TatA association model, raising the number of transport-competent substrate proteins within the cell leads to an increase in the number of large TatA complexes present. Formation of these complexes requires both a functional TatBC substrate receptor and the transmembrane proton motive force (PMF). Removing the PMF causes TatA complexes to dissociate, except in strains with impaired Tat transport activity. Based on these observations we propose that TatA assembly reaches a critical point at which oligomerization can be reversed only by substrate transport. In contrast to TatA-YFP, the oligomeric states of TatB-YFP and TatC-YFP fusions are not affected by substrate or the PMF, although TatB-YFP oligomerization does require TatC.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Bactérias , Escherichia coli/metabolismo , Proteínas Luminescentes , Microscopia de Fluorescência , Transporte Proteico/fisiologia , Força Próton-Motriz/fisiologia
14.
Proc Natl Acad Sci U S A ; 110(22): E2054-63, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23671069

RESUMO

Lambdoid phage 21 uses a pinholin-signal anchor release endolysin strategy to effect temporally regulated host lysis. In this strategy, the pinholin S(21)68 accumulates harmlessly in the bilayer until suddenly triggering to form lethal membrane lesions, consisting of S(21)68 heptamers with central pores <2 nm in diameter. The membrane depolarization caused by these pores activates the muralytic endolysin, R(21), leading immediately to peptidoglycan degradation. The lethal S(21)68 complexes have been designated as pinholes to distinguish from the micrometer-scale holes formed by canonical holins. Here, we used GFP fusions of WT and mutant forms of S(21)68 to show that the holin accumulates uniformly throughout the membrane until the time of triggering, when it suddenly redistributes into numerous small foci (rafts). Raft formation correlates with the depletion of the proton motive force, which is indicated by the potential-sensitive dye bis-(1,3-dibutylbarbituric acid)pentamethine oxonol. By contrast, GFP fusions of either antiholin variant irsS(21)68, which only forms inactive dimers, or nonlethal mutant S(21)68(S44C), which is blocked at an activated dimer stage of the pinhole formation pathway, were both blocked in a state of uniform distribution. In addition, fluorescence recovery after photobleaching revealed that, although the antiholin irsS(21)68-GFP fusion was highly mobile in the membrane (even when the proton motive force was depleted), more than one-half of the S(21)68-GFP molecules were immobile, and the rest were in mobile states with a much lower diffusion rate than the rate of irsS(21)68-GFP. These results suggest a model in which, after transiting into an oligomeric state, S(21)68 migrates into rafts with heterogeneous sizes, within which the final pinholes form.


Assuntos
Bacteriólise/genética , Membrana Celular/metabolismo , Escherichia coli/virologia , Siphoviridae/genética , Proteínas Virais/metabolismo , Bacteriólise/fisiologia , Barbitúricos , Membrana Celular/ultraestrutura , Escherichia coli/fisiologia , Recuperação de Fluorescência Após Fotodegradação , Proteínas de Fluorescência Verde/metabolismo , Isoxazóis , Microscopia de Fluorescência , Força Próton-Motriz/fisiologia , Siphoviridae/metabolismo , Siphoviridae/fisiologia , Imagem com Lapso de Tempo
15.
Proc Natl Acad Sci U S A ; 110(22): 8912-7, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23674679

RESUMO

Proton transfer across biological membranes underpins central processes in biological systems, such as energy conservation and transport of ions and molecules. In the membrane proteins involved in these processes, proton transfer takes place through specific pathways connecting the two sides of the membrane via control elements within the protein. It is commonly believed that acidic residues are required near the orifice of such proton pathways to facilitate proton uptake. In cytochrome c oxidase, one such pathway starts near a conserved Asp-132 residue. Results from earlier studies have shown that replacement of Asp-132 by, e.g., Asn, slows proton uptake by a factor of ∼5,000. Here, we show that proton uptake at full speed (∼10(4) s(-1)) can be restored in the Asp-132-Asn oxidase upon introduction of a second structural modification further inside the pathway (Asn-139-Thr) without compensating for the loss of the negative charge. This proton-uptake rate was insensitive to Zn(2+) addition, which in the wild-type cytochrome c oxidase slows the reaction, indicating that Asp-132 is required for Zn(2+) binding. Furthermore, in the absence of Asp-132 and with Thr at position 139, at high pH (>9), proton uptake was significantly accelerated. Thus, the data indicate that Asp-132 is not strictly required for maintaining rapid proton uptake. Furthermore, despite the rapid proton uptake in the Asn-139-Thr/Asp-132-Asn mutant cytochrome c oxidase, proton pumping was impaired, which indicates that the segment around these residues is functionally linked to pumping.


Assuntos
Ácido Aspártico/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/química , Modelos Moleculares , Conformação Proteica , Bombas de Próton/química , Força Próton-Motriz/fisiologia , Absorção , Cristalografia por Raios X , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Oxirredução , Bombas de Próton/genética , Bombas de Próton/metabolismo , Força Próton-Motriz/genética , Rhodobacter sphaeroides , Zinco
16.
J Integr Plant Biol ; 58(10): 848-858, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26947269

RESUMO

During photosynthesis, photosynthetic electron transport generates a proton motive force (pmf) across the thylakoid membrane, which is used for ATP biosynthesis via ATP synthase in the chloroplast. The pmf is composed of an electric potential (ΔΨ) and an osmotic component (ΔpH). Partitioning between these components in chloroplasts is strictly regulated in response to fluctuating environments. However, our knowledge of the molecular mechanisms that regulate pmf partitioning is limited. Here, we report a bestrophin-like protein (AtBest), which is critical for pmf partitioning. While the ΔpH component was slightly reduced in atbest, the ΔΨ component was much greater in this mutant than in the wild type, resulting in less efficient activation of nonphotochemical quenching (NPQ) upon both illumination and a shift from low light to high light. Although no visible phenotype was observed in the atbest mutant in the greenhouse, this mutant exhibited stronger photoinhibition than the wild type when grown in the field. AtBest belongs to the bestrophin family proteins, which are believed to function as chloride (Cl- ) channels. Thus, our findings reveal an important Cl- channel required for ion transport and homeostasis across the thylakoid membrane in higher plants. These processes are essential for fine-tuning photosynthesis under fluctuating environmental conditions.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Tilacoides/metabolismo , Concentração de Íons de Hidrogênio , Fotossíntese/fisiologia , Força Próton-Motriz/fisiologia
17.
Proc Natl Acad Sci U S A ; 109(37): 14876-81, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22927379

RESUMO

The molecular origin of the action of the F(0) proton gradient-driven rotor presents a major puzzle despite significant structural advances. Although important conceptual models have provided guidelines of how such systems should work, it has been challenging to generate a structure-based molecular model using physical principles that will consistently lead to the unidirectional proton-driven rotational motion during ATP synthesis. This work uses a coarse-grained (CG) model to simulate the energetics of the F(0)-ATPase system in the combined space defined by the rotational coordinate and the proton transport (PTR) from the periplasmic side (P) to the cytoplasmic side (N). The model establishes the molecular origin of the rotation, showing that this effect is due to asymmetry in the energetics of the proton path rather than only the asymmetry of the interaction of the Asp on the c-ring helices and Arg on the subunit-a. The simulation provides a clear conceptual background for further exploration of the electrostatic basis of proton-driven mechanochemical systems.


Assuntos
Trifosfato de Adenosina/biossíntese , Força Próton-Motriz/fisiologia , ATPases Translocadoras de Prótons/química , Rotação , Simulação de Dinâmica Molecular , Eletricidade Estática
18.
J Bacteriol ; 196(17): 3074-81, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24936052

RESUMO

The gastric pathogen Helicobacter pylori possesses a highly active urease to support acid tolerance. Urea hydrolysis occurs inside the cytoplasm, resulting in the production of NH3 that is immediately protonated to form NH4 (+). This ammonium must be metabolized or effluxed because its presence within the cell is counterproductive to the goal of raising pH while maintaining a viable proton motive force (PMF). Two compatible hypotheses for mitigating intracellular ammonium toxicity include (i) the exit of protonated ammonium outward via the UreI permease, which was shown to facilitate diffusion of both urea and ammonium, and/or (ii) the assimilation of this ammonium, which is supported by evidence that H. pylori assimilates urea nitrogen into its amino acid pools. We investigated the second hypothesis by constructing strains with altered expression of the ammonium-assimilating enzymes glutamine synthetase (GS) and glutamate dehydrogenase (GDH) and the ammonium-evolving periplasmic enzymes glutaminase (Ggt) and asparaginase (AsnB). H. pylori strains expressing elevated levels of either GS or GDH are more acid tolerant than the wild type, exhibit enhanced ammonium production, and are able to alkalize the medium faster than the wild type. Strains lacking the genes for either Ggt or AsnB are acid sensitive, have 8-fold-lower urea-dependent ammonium production, and are more acid sensitive than the parent. Additionally, we found that purified H. pylori GS produces glutamine in the presence of Mg(2+) at a rate similar to that of unadenylated Escherichia coli GS. These data reveal that all four enzymes contribute to whole-cell acid resistance in H. pylori and are likely important for assimilation and/or efflux of urea-derived ammonium.


Assuntos
Ácidos/farmacologia , Compostos de Amônio/metabolismo , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/enzimologia , Asparaginase/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Glutamato Desidrogenase/metabolismo , Glutamato-Amônia Ligase/metabolismo , Glutaminase/metabolismo , Concentração de Íons de Hidrogênio , Força Próton-Motriz/fisiologia , Urease/metabolismo
19.
J Biol Chem ; 288(15): 10567-77, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23457298

RESUMO

Superoxide flashes are transient bursts of superoxide production within the mitochondrial matrix that are detected using the superoxide-sensitive biosensor, mitochondria-targeted circularly permuted YFP (mt-cpYFP). However, due to the pH sensitivity of mt-cpYFP, flashes were suggested to reflect transient events of mitochondrial alkalinization. Here, we simultaneously monitored flashes with mt-cpYFP and mitochondrial pH with carboxy-SNARF-1. In intact cardiac myocytes and purified skeletal muscle mitochondria, robust mt-cpYFP flashes were accompanied by only a modest increase in SNARF-1 ratio (corresponding to a pH increase of <0.1), indicating that matrix alkalinization is minimal during an mt-cpYFP flash. Individual flashes were also accompanied by stepwise increases of MitoSOX signal and decreases of NADH autofluorescence, supporting the superoxide origin of mt-cpYFP flashes. Transient matrix alkalinization induced by NH4Cl only minimally influenced flash frequency and failed to alter flash amplitude. However, matrix acidification modulated superoxide flash frequency in a bimodal manner. Low concentrations of nigericin (< 100 nM) that resulted in a mild dissipation of the mitochondrial pH gradient increased flash frequency, whereas a maximal concentration of nigericin (5 µm) collapsed the pH gradient and abolished flash activity. These results indicate that mt-cpYFP flash events reflect a burst in electron transport chain-dependent superoxide production that is coincident with a modest increase in matrix pH. Furthermore, flash activity depends strongly on a combination of mitochondrial oxidation and pH gradient.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Superóxidos/metabolismo , Cloreto de Amônio/farmacologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Benzopiranos/farmacologia , Células Cultivadas , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Corantes Fluorescentes/farmacologia , Concentração de Íons de Hidrogênio , Ionóforos/farmacologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Naftóis/farmacologia , Nigericina/farmacologia , Oxirredução/efeitos dos fármacos , Força Próton-Motriz/efeitos dos fármacos , Força Próton-Motriz/fisiologia , Ratos , Ratos Sprague-Dawley , Rodaminas/farmacologia
20.
Antimicrob Agents Chemother ; 58(5): 2491-503, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24614376

RESUMO

The inherent drug susceptibility of microorganisms is determined by multiple factors, including growth state, the rate of drug diffusion into and out of the cell, and the intrinsic vulnerability of drug targets with regard to the corresponding antimicrobial agent. Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), remains a significant source of global morbidity and mortality, further exacerbated by its ability to readily evolve drug resistance. It is well accepted that drug resistance in M. tuberculosis is driven by the acquisition of chromosomal mutations in genes encoding drug targets/promoter regions; however, a comprehensive description of the molecular mechanisms that fuel drug resistance in the clinical setting is currently lacking. In this context, there is a growing body of evidence suggesting that active extrusion of drugs from the cell is critical for drug tolerance. M. tuberculosis encodes representatives of a diverse range of multidrug transporters, many of which are dependent on the proton motive force (PMF) or the availability of ATP. This suggests that energy metabolism and ATP production through the PMF, which is established by the electron transport chain (ETC), are critical in determining the drug susceptibility of M. tuberculosis. In this review, we detail advances in the study of the mycobacterial ETC and highlight drugs that target various components of the ETC. We provide an overview of some of the efflux pumps present in M. tuberculosis and their association, if any, with drug transport and concomitant effects on drug resistance. The implications of inhibiting drug extrusion, through the use of efflux pump inhibitors, are also discussed.


Assuntos
Metabolismo Energético/fisiologia , Mycobacterium tuberculosis/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Transporte Biológico/fisiologia , Força Próton-Motriz/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA