Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(2): 105537, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38072042

RESUMEN

The extremophile bacterium D. radiodurans boasts a distinctive cell envelope characterized by the regular arrangement of three protein complexes. Among these, the Type II Secretion System (T2SS) stands out as a pivotal structural component. We used cryo-electron microscopy to reveal unique features, such as an unconventional protein belt (DR_1364) around the main secretin (GspD), and a cap (DR_0940) found to be a separated subunit rather than integrated with GspD. Furthermore, a novel region at the N-terminus of the GspD constitutes an additional second gate, supplementing the one typically found in the outer membrane region. This T2SS was found to contribute to envelope integrity, while also playing a role in nucleic acid and nutrient trafficking. Studies on intact cell envelopes show a consistent T2SS structure repetition, highlighting its significance within the cellular framework.


Asunto(s)
Membrana Celular , Deinococcus , Extremófilos , Sistemas de Secreción Tipo II , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Deinococcus/metabolismo , Extremófilos/metabolismo , Sistemas de Secreción Tipo II/química , Sistemas de Secreción Tipo II/metabolismo , Transporte de Proteínas
2.
Chemphyschem ; : e202400147, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38625051

RESUMEN

We investigated, by using all-atom molecular dynamics simulations, the effect of the outer membrane of Gram-negative bacteria, composed in the outer leaflet by polar/charged lipopolysaccharides (LPS), on the electrostatic properties of general porins from the Enterobacteriaceae family. General porins constitute the main path for the facilitated diffusion of polar antibiotics through the outer membrane. As model system we selected OmpK36 from Klebsiella pneumoniae, the ortholog of OmpC from Escherichia coli. This species presents high variability of amino acid composition of porins, with the effect to increase its resistance to the penetration of antibiotics. The various properties we analyzed seem to indicate that LPS acts as an independent layer without affecting the internal electrostatic properties of OmpK36. The only apparent effect on the microsecond time scale we sampled is the appearance of calcium ions, when present at moderate concentration in solution, inside the pore. However, we noticed increased fluctuations of the polarization density and only minor changes on its average value.

3.
Phys Chem Chem Phys ; 25(39): 26497-26506, 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37772905

RESUMEN

General porins are nature's sieving machinery in the outer membrane of Gram-negative bacteria. Their unique hourglass-shaped architecture is highly conserved among different bacterial membrane proteins and other biological channels. These biological nanopores have been designed to protect the interior of the bacterial cell from leakage of toxic compounds while selectively allowing the entry of the molecules needed for cell growth and function. The mechanism of transport through porins is of utmost and direct interest for drug discovery, extending toward nanotechnology applications for blue energy, separations, and sequencing. Here we present a theoretical framework for analysing the filter of general porins in relation to translocating molecules with the aid of enhanced molecular simulations quantitatively. Using different electrostatic probes in the form of a series of related molecules, we describe the nature of this filter and how to finely tune permeability by exploiting electrostatic interactions between the pore and the translocating molecule. Eventually, we show how enhanced simulations constitute today a valid tool for characterising the mechanism and quantifying energetically the transport of molecules through nanopores.

4.
Phys Chem Chem Phys ; 25(18): 12712-12722, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37098836

RESUMEN

Transmembrane ß-barrel proteins are key systems for transport phenomena in biology. Based on their broad substrate specificity, they represent good candidates for present and future technological applications, such as DNA/RNA and protein sequencing, sensing of biomedical analytes, and production of blue energy. For a better understanding of the process at the molecular level, we applied parallel tempering simulations in the WTE ensemble to compare two ß-barrel porins from Escherichia coli, OmpF and OmpC. Our analysis showed a different behavior of the two highly homologous porins, where subtle amino acid substitutions can modulate critical properties of mass transport. Interestingly, the differences can be mapped to the respective environmental conditions under which the two porins are expressed. Apart from reporting on the advantages of the enhanced sampling methods in assessing the molecular properties of nanopores, our comparative analysis provided new and key results to better understand biological function and technical applications. Eventually, we showed how results from molecular simulations align well with experimental single-channel measurements, thus demonstrating the mature evolution of numerical methodologies for predicting properties in this field crucial for future biomedical applications.


Asunto(s)
Escherichia coli , Porinas , Escherichia coli/metabolismo , Secuencia de Aminoácidos , Porinas/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Bacterianas/metabolismo
5.
J Biol Chem ; 295(13): 4224-4236, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32071085

RESUMEN

In the extremophile bacterium Deinococcus radiodurans, the outermost surface layer is tightly connected with the rest of the cell wall. This integrated organization provides a compact structure that shields the bacterium against environmental stresses. The fundamental unit of this surface layer (S-layer) is the S-layer deinoxanthin-binding complex (SDBC), which binds the carotenoid deinoxanthin and provides both, thermostability and UV radiation resistance. However, the structural organization of the SDBC awaits elucidation. Here, we report the isolation of the SDBC with a gentle procedure consisting of lysozyme treatment and solubilization with the nonionic detergent n-dodecyl-ß-d-maltoside, which preserved both hydrophilic and hydrophobic components of the SDBC and allows the retention of several minor subunits. As observed by low-resolution single-particle analysis, we show that the complex possesses a porin-like structural organization, but is larger than other known porins. We also noted that the main SDBC component, the protein DR_2577, shares regions of similarity with known porins. Moreover, results from electrophysiological assays with membrane-reconstituted SDBC disclosed that it is a nonselective channel that has some peculiar gating properties, but also exhibits behavior typically observed in pore-forming proteins, such as porins and ionic transporters. The functional properties of this system and its porin-like organization provide information critical for understanding ion permeability through the outer cell surface of S-layer-carrying bacterial species.


Asunto(s)
Proteínas Bacterianas/química , Deinococcus/química , Glicoproteínas de Membrana/química , Complejos Multiproteicos/química , Proteínas Bacterianas/genética , Carotenoides/química , Membrana Celular/química , Pared Celular/química , Deinococcus/genética , Complejos Multiproteicos/genética , Porinas/química , Unión Proteica/genética
6.
Phys Chem Chem Phys ; 23(34): 18461-18474, 2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34612386

RESUMEN

Subcellular and organellar mechanisms have manifested a prominent importance for a broad variety of processes that maintain cellular life at its most basic level. Mammalian two-pore channels (TPCs) appear to be cornerstones of these processes in endo-lysosomes by controlling delicate ion-concentrations in their interiors. With evolutionary remarkable architecture and one-of-a-kind selectivity filter, TPCs are an extremely attractive topic per se. In the light of the current COVID-19 pandemic, hTPC2 emerges to be more than attractive. As a key regulator of the endocytosis pathway, it is potentially essential for diverse viral infections in humans, as demonstrated. Here, by means of multiscale molecular simulations, we propose a model of sodium transport from the lumen to the cytosol where the central cavity works as a reservoir. Since the inhibition of hTPC2 is proven to stop SARS-CoV2 in vitro, shedding light on the hTPC2 function and mechanism is the first step towards the selection of potential inhibiting candidates.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos Activados por Ligandos/fisiología , Sodio/metabolismo , COVID-19 , Canales Iónicos Activados por Ligandos/metabolismo , Ligandos , SARS-CoV-2/aislamiento & purificación
7.
Phys Chem Chem Phys ; 22(27): 15664-15674, 2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32618303

RESUMEN

In the last decade two-pore intracellular channels (TPCs) attracted the interest of researchers, still some key questions remain open. Their importance for vacuolar (plants) and endo-lysosomal (animals) function highlights them as a very attractive system to study, both theoretically and experimentally. Indicated as key players in the trafficking of the cell, today they are considered a new potential target for avoiding virus infections, including those from coronaviruses. A particular boost for theoretical examinations has been made with recent high-resolution X-ray and cryo-EM structures. These findings have opened the way for efficient and precise computational studies at the atomistic level. Here we report a set of multiscale-calculations performed on the mTPC1, a ligand- and voltage-gated sodium selective channel. The molecular dynamics and enhanced molecular dynamics simulations were used for a thorough analysis of the mammalian TPC1 behaviour in the presence and absence of the ligand molecule, with a special accent on the supposed bottleneck, the hydrophobic gate. Moreover, from the reconstructed free energy obtained from enhanced simulations, we have calculated the macroscopic conductance of sodium ions through the mTPC1, which we compared with measured single-channel conductance values. The hydrophobic gate works as a steric barrier and the key parameters are its flexibility and the dimension of the sodium first hydration shell.


Asunto(s)
Canales de Calcio/química , Simulación del Acoplamiento Molecular , Animales , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico , Ligandos , Ratones
8.
Molecules ; 25(23)2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-33291474

RESUMEN

Modern medicine relies upon antibiotics, but we have arrived to the point where our inability to come up with new effective molecules against resistant pathogens, together with the declining private investment, is resulting in the number of untreatable infections increasing worldwide at worrying pace. Among other pathogens, widely recognized institutions have indicated Gram-negative bacteria as particularly challenging, due to the presence of the outer membrane. The very first step in the action of every antibiotic or adjuvant is the permeation through this membrane, with small hydrophilic drugs usually crossing through protein channels. Thus, a detailed understanding of their properties at a molecular level is crucial. By making use of Molecular Dynamics simulations, we compared the two main porins of four members of the Enterobacteriaceae family, and, in this paper, we show their shared geometrical and electrostatic characteristics. Then, we used metadynamics simulations to reconstruct the free energy for permeation of selected diazobicyclooctans through OmpF. We demonstrate how porins features are coupled to those of the translocating species, modulating their passive permeation. In particular, we show that the minimal projection area of a molecule is a better descriptor than its molecular mass or the volume. Together with the magnitude and orientation of the electric dipole moment, these are the crucial parameters to gain an efficient compensation between the entropic and enthalpic contributions to the free energy barrier required for permeation. Our results confirm the possibility to predict the permeability of molecules through porins by using a few molecular parameters and bolster the general model according to which the free energy increase is mostly due to the decrease of conformational entropy, and this can be compensated by a favorable alignment of the electric dipole with respect to the channel intrinsic electric field.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Bacterias Gramnegativas/metabolismo , Porinas/metabolismo , Inhibidores de beta-Lactamasas/metabolismo , Antibacterianos/metabolismo , Enterobacteriaceae/metabolismo , Simulación de Dinámica Molecular , Electricidad Estática
9.
Phys Chem Chem Phys ; 21(16): 8457-8463, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-30951074

RESUMEN

Deciphering the transport through outer-membrane porins is crucial to understand how anti-infectives enter Gram-negative bacteria and perform their function. Here we elucidated the transport mechanism of substrates through the Pseudomonads sugar-specific porin OprB by means of multiscale modeling. We used molecular dynamics simulations to quantify the energetics of transport and thus a diffusion model to quantify the macroscopic flux of molecules through OprB. Our results show that Trp171 and several glutamate residues in the constriction region are key for the transport of glucose, the preferred natural substrate, through OprB. The unveiled transport mechanism suggests that 2-acetamido-1,2-dideoxynojirimycin (DNJ-NAc), an anti-infective structurally similar to glucose, can enter the cell via OprB. We quantified its energetics and macroscopic flux through OprB providing a comparative analysis with the natural substrate. Thus this pore can be considered as a promising gateway for exploiting the Trojan Horse strategy in pathogenic bacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glucosa/metabolismo , Porinas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas putida/metabolismo , Antiinfecciosos/metabolismo , Proteínas Bacterianas/química , Transporte Biológico , Humanos , Modelos Moleculares , Porinas/química , Conformación Proteica , Pseudomonas putida/química , Especificidad por Sustrato
10.
Biometals ; 32(1): 155-170, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30635814

RESUMEN

Nuclear magnetic resonance and infrared spectroscopy have been used to investigate the formation of complexes of BAL30072 with Fe3+ and Ga3+ in solution and to collect geometrical parameters supporting reliable 3D structure models. Structural models for the ligand-metal complexes with different stoichiometries have been characterized using density functional theory calculations. Blind ensemble docking to the PiuA receptor from P. aeruginosa was performed for the different complexes to compare binding affinities and statistics of the residues most frequently contacted. When compared to analogues, BAL30072 was found to have an intrinsic propensity to form complexes with low ligand-to-metal stoichiometry. By using one of the sulfate oxygen atoms as a third donor in addition to the bidentate pyridinone moiety, BAL30072 can form a L2M complex, which was predicted to be the one with the best binding affinity to PiuA. The example of BAL30072 strongly suggests that a lower stoichiometry might be the one recognized by the receptor, so that to focus only on the highest stoichiometry might be misleading for siderophores with less than six donors.


Asunto(s)
Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/antagonistas & inhibidores , Monobactamas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Sideróforos/química , Tiazoles/farmacología , Antibacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Simulación del Acoplamiento Molecular , Monobactamas/química , Tiazoles/química
11.
J Chem Phys ; 150(21): 211102, 2019 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-31176312

RESUMEN

We present a statistical model for solving and predicting the transport of large molecules through small flexible channels. The average radius of the channel and the average radius of the molecule are the only two quantities determining the steric part of the potential of mean force for the translocation, in the case of a small rigid particle and a large rigid channel: the barrier is completely entropic and is described by the Fick-Jacobs model. However, the flexibility of the channel's cross section and that of the molecule's size have a significant effect on transport, especially when a large molecule goes through a narrow channel. In this case, the steric barrier changes its statistical nature becoming enthalpic, and we predict a strong temperature enhancement of the diffusion current through the channel. The flexibility is described in terms of the equilibrium fluctuations of the channel and of the molecule. The model is compared with the all-atom MD simulations of the transport of hard spheres of various radii and of drug molecules through a biological nanochannel. For the case of Gaussian fluctuations, we derived a simple analytical expression for the steric barrier, which can be quantified using average size and fluctuations of the channel and of the molecule.

12.
Biochim Biophys Acta Bioenerg ; 1859(4): 270-279, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29408701

RESUMEN

The yeast Saccharomyces cerevisiae genome is endowed with two distinct isoforms of Voltage-Dependent Anion Channel (VDAC). The isoform yVDAC2 is currently understudied with respect to the best known yVDAC1. Yet, since the discovery, the function of yVDAC2 was unclear, leading to the hypothesis that it might be devoid of a channel function. In this work we have elucidated, by bioinformatics modeling and electrophysiological analysis, the functional activity of yVDAC2. The conformation of yVDAC2 and, for comparison, of yVDAC1 were modeled using a multiple template approach involving mouse, human and zebrafish structures and both showed to arrange the sequences as the typical 19-stranded VDAC ß-barrel. Molecular dynamics simulations showed that yVDAC2, in comparison with yVDAC1, has a different number of permeation paths of potassium and chloride ions. yVDAC2 protein was over-expressed in the S. cerevisiae cells depleted of functional yVDAC1 (Δpor1 mutant) and, after purification, it was reconstituted in artificial membranes (planar lipid bilayer (PLB) system). The protein displayed channel-forming activity and the calculated conductance, voltage-dependence and ion selectivity values were similar to those of yVDAC1 and other members of VDAC family. This is the first time that yVDAC2 channel features are detected and characterized.


Asunto(s)
Mitocondrias/química , Membranas Mitocondriales/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 2 Dependiente del Voltaje/química , Animales , Sitios de Unión , Cloruros/química , Cloruros/metabolismo , Biología Computacional , Expresión Génica , Humanos , Transporte Iónico , Cinética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Ratones , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Simulación de Dinámica Molecular , Potasio/química , Potasio/metabolismo , Unión Proteica , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología Estructural de Proteína , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/genética , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Pez Cebra
13.
Phys Chem Chem Phys ; 20(13): 8533-8546, 2018 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-29542754

RESUMEN

One of the greatest health threats facing modern medicine is the emergence of new bacterial strains which are increasingly resistant to almost all currently available antibiotics. According to a CDC (Center for Disease Control and Prevention) report published in 2013, 63% of Acinetobacter species have been identified as Multidrug resistant strains. As for other Gram-negative bacteria, the presence of an outer membrane increases the intrinsic resistance of A. baumannii to most antibiotics. The outer membrane of A. baumannii possesses several specific porins that control the selectivity for different polar substrates in a way that is still poorly understood. Recently, the X-ray crystal structures of 4 related porins, termed OccAB1-4, were solved at high resolution, providing a framework to study the structural and functional characteristics of these porins in filtering natural substrates. Here, we first use molecular dynamics simulations on OccAB proteins to investigate the stability and dynamics of the pores, and to establish their common biophysical features. We then applied metadynamics simulations to evaluate the free energy costs required for polar substrates to overcome the pore. Together, the comparative analysis of the OccAB porins not only sheds light on how these channels could function as potential antibiotic gateways, but also allows identification of putative affinity sites that represent a common path through which other molecules can transit.

14.
J Biol Chem ; 291(6): 2837-47, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26645688

RESUMEN

Integral membrane proteins known as porins are the major pathway by which hydrophilic antibiotics cross the outer membrane of Gram-negative bacteria. Single point mutations in porins can decrease the permeability of an antibiotic, either by reduction of channel size or modification of electrostatics in the channel, and thereby confer clinical resistance. Here, we investigate four mutant OmpC proteins from four different clinical isolates of Escherichia coli obtained sequentially from a single patient during a course of antimicrobial chemotherapy. OmpC porin from the first isolate (OmpC20) undergoes three consecutive and additive substitutions giving rise to OmpC26, OmpC28, and finally OmpC33. The permeability of two zwitterionic carbapenems, imipenem and meropenem, measured using liposome permeation assays and single channel electrophysiology differs significantly between OmpC20 and OmpC33. Molecular dynamic simulations show that the antibiotics must pass through the constriction zone of porins with a specific orientation, where the antibiotic dipole is aligned along the electric field inside the porin. We identify that changes in the vector of the electric field in the mutated porin, OmpC33, create an additional barrier by "trapping" the antibiotic in an unfavorable orientation in the constriction zone that suffers steric hindrance for the reorientation needed for its onward translocation. Identification and understanding the underlying molecular details of such a barrier to translocation will aid in the design of new antibiotics with improved permeation properties in Gram-negative bacteria.


Asunto(s)
Escherichia coli/química , Imipenem/química , Porinas/química , Tienamicinas/química , Resistencia betalactámica , Escherichia coli/genética , Escherichia coli/metabolismo , Imipenem/farmacología , Meropenem , Mutación , Porinas/genética , Porinas/metabolismo , Tienamicinas/farmacología
15.
Biochim Biophys Acta ; 1858(7 Pt B): 1772-7, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26796683

RESUMEN

The number of pathogens developing multiple drug resistance is ever increasing. The impact on healthcare systems is huge and the need for novel antibiotics as well a new way to develop them is urgent, especially against Gram-negative bacteria. The first defense of these bacteria is the outer membrane, where unspecific protein channels (porins) modulate nutrients passive diffusion. Also polar antibiotics enter through this path and down-regulation and/or mutation of porins are very common in drug resistant strains. Our inability to come up with novel effective antibiotics mostly relies upon the insufficient comprehension of the key molecular features enabling better penetration through porins. Molecular dynamics simulations offer an extraordinary tool in the study of the dynamics of biological systems; however, one of the major drawbacks of this method is that its use is currently restricted to study time scales of the order of microsecond. Enhanced sampling methods like Metadynamics have been recently used to investigate the diffusion of antibiotics through bacterial porins. The main limitation is that dynamical properties cannot be estimated because of the different potential that the systems under study are experiencing. Recently, the scope of Metadynamics has been extended. By applying an a posteriori analysis one can obtain rates of transitions and rate-limiting steps of the process under study, directly comparable with kinetic data extracted from electrophysiology experiments. In this work, we apply this method to the study of the permeability of Escherichia coli's OmpF with respect to Meropenem, finding good agreement with the residence time obtained analyzing experimental current noise. This article is part of a Special Issue entitled: Membrane Proteins edited by J.C. Gumbart and Sergei Noskov.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/ultraestructura , Membrana Celular/química , Canales Iónicos/química , Canales Iónicos/ultraestructura , Simulación de Dinámica Molecular , Antibacterianos/química , Sitios de Unión , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular , Difusión , Bacterias Gramnegativas/química , Bacterias Gramnegativas/ultraestructura , Activación del Canal Iónico , Membrana Dobles de Lípidos/química , Modelos Químicos , Porosidad , Unión Proteica , Conformación Proteica
16.
Biochim Biophys Acta ; 1858(4): 813-23, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26806159

RESUMEN

The human VDAC channel exists in three isoforms characterized by high sequence homology and structural similarity. Yet the function and mode of action of hVDAC3 are still elusive. The presence of six surface cysteines exposed to the oxidizing environment of the mitochondrial inter-membrane space suggests the possible establishment of intramolecular disulfide bonds. Two natural candidates for disulfide bridge formation are Cys2 and Cys8 that, located on the flexible N-terminal domain, can easily come in contact. A third potentially important residue is Cys122 that is close to Cys2 in the homology model of VDAC3. Here we analyzed the impact of SS bonds through molecular dynamics simulations of derivatives of hVDAC3 (dubbed SS-2-8, SS-2-122, SS-8-122) including a single disulfide bond. Simulations showed that in SS-8-122, the fragment 1-7 crosses the top part of the barrel partially occluding the pore and causing a 20% drop of conductance. In order to identify other potential channel-occluding disulfide bonds, we used a set of neural networks and structural bioinformatics algorithms, after filtering with the steric constraints imposed by the 3D-structure. We identified other three species, namely SS-8-65, SS-2-36 and SS-8-36. While the conductance of SS-8-65 and SS-2-36 is about 30% lower than that of the species without disulfide bonds, the conductance of SS-8-36 was 40-50% lower. The results show how VDAC3 is able to modulate its pore size and current by exploiting the mobility of the N-terminal and forming, upon external stimuli, disulfide bridges with cysteine residues located on the barrel and exposed to the inter-membrane space.


Asunto(s)
Cisteína/química , Disulfuros/química , Proteínas de Transporte de Membrana Mitocondrial/química , Conformación Proteica , Canales Aniónicos Dependientes del Voltaje/química , Humanos , Transporte Iónico , Simulación de Dinámica Molecular , Isoformas de Proteínas/química
17.
Phys Chem Chem Phys ; 18(13): 8855-64, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26931352

RESUMEN

Multi-drug resistance bacteria are a challenging problem of contemporary medicine. This is particularly critical for Gram-negative bacteria, where antibiotics are hindered by the outer membrane to reach internal targets. Here more polar antibiotics make use of nanometric water-filled channels to permeate inside. We present in this work a computational all-atom approach, using water as a probe, for the calculation of the macroscopic electric field inside water-filled channels. The method allows one to compare not only different systems but also the same system under different conditions, such as pH and ion concentration. This provides a detailed picture of electrostatics in biological nanopores shedding more light on how the charged residues of proteins determine the electric field inside, and also how medium can tune it. These details are central to unveil the filtering mechanism behind the permeation of small polar molecules through nanometric water-filled channels.


Asunto(s)
Electricidad , Nanoporos , Farmacorresistencia Microbiana , Bacterias Gramnegativas/efectos de los fármacos , Concentración de Iones de Hidrógeno , Concentración Osmolar , Agua/química
18.
Phys Chem Chem Phys ; 18(45): 30998-31011, 2016 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-27805179

RESUMEN

Dendrimeric peptides make a versatile group of bioactive peptidomimetics and a potential new class of antimicrobial agents to tackle the pressing threat of multi-drug resistant pathogens. These are branched supramolecular assemblies where multiple copies of the bioactive unit are linked to a central core. Beyond their antimicrobial activity, dendrimeric peptides could also be designed to functionalize the surface of nanoparticles or materials for other medical uses. Despite these properties, however, little is known about the structure-function relationship of such compounds, which is key to unveil the fundamental physico-chemical parameters and design analogues with desired attributes. To close this gap, we focused on a semi-synthetic, two-branched peptide, SB056, endowed with remarkable activity against both Gram-positive and Gram-negative bacteria and limited cytotoxicity. SB056 can be considered the smallest prototypical dendrimeric peptide, with the core restricted to a single lysine residue and only two copies of the same highly cationic 10-mer polypeptide; an octanamide tail is present at the C-terminus. Combining NMR and Molecular Dynamics simulations, we have determined the 3D structure of two analogues. Fluorescence spectroscopy was applied to investigate the water-bilayer partition in the presence of vesicles of variable charge. Vesicle leakage assays were also performed and the experimental data were analyzed by applying an iterative Monte Carlo scheme to estimate the minimum number of bound peptides needed to achieve the release. We unveiled a singular beta hairpin-type structure determined by the peptide chains only, with the octanamide tail available for further functionalization to add new potential properties without affecting the structure.


Asunto(s)
Antibacterianos/química , Bacterias Gramnegativas , Simulación de Dinámica Molecular , Péptidos/química , Farmacorresistencia Bacteriana , Método de Montecarlo , Espectrometría de Fluorescencia
19.
Biochemistry ; 54(36): 5646-56, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26303511

RESUMEN

The voltage-dependent anion channel (VDAC) is the main mitochondrial porin allowing the exchange of ions and metabolites between the cytosol and the mitochondrion. In addition, VDAC was found to actively interact with proteins playing a fundamental role in the regulation of apoptosis and being of central interest in cancer research. VDAC is a large transmembrane ß-barrel channel, whose N-terminal helical fragment adheres to the channel interior, partially closing the pore. This fragment is considered to play a key role in protein stability and function as well as in the interaction with apoptosis-related proteins. Three VDAC isoforms are differently expressed in higher eukaryotes, for which distinct and complementary roles are proposed. In this work, the folding propensity of their N-terminal fragments has been compared. By using multiple spectroscopic techniques, and complementing the experimental results with theoretical computer-assisted approaches, we have characterized their conformational equilibrium. Significant differences were found in the intrinsic helical propensity of the three peptides, decreasing in the following order: hVDAC2 > hVDAC3 > hVDAC1. In light of the models proposed in the literature to explain voltage gating, selectivity, and permeability, as well as interactions with functionally related proteins, our results suggest that the different chemicophysical properties of the N-terminal domain are possibly correlated to different functions for the three isoforms. The overall emerging picture is that a similar transmembrane water accessible conduit has been equipped with not identical domains, whose differences can modulate the functional roles of the three VDAC isoforms.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas Mitocondriales/química , Péptidos/química , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 2 Dependiente del Voltaje/química , Canales Aniónicos Dependientes del Voltaje/química , Dicroismo Circular , Biología Computacional , Humanos , Simulación de Dinámica Molecular , Resonancia Magnética Nuclear Biomolecular , Isoformas de Proteínas/química , Estructura Secundaria de Proteína , Espectrofotometría Infrarroja
20.
Analyst ; 140(14): 4820-7, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-25717496

RESUMEN

For an antibiotic to be effective, it needs to cross the outer membrane barrier and reach the target inside the cell. Hydrophilic antibiotics, e.g.ß-lactams, use porin channels to cross the outer membrane and accumulate in the periplasm. Experimental determination of antibiotic interactions with porin is performed by using electrophysiology on a single channel level by noise analysis or single event analysis methods. We report a novel framework for analyzing the ion-current noise, taking into account the corrections due to the analogous filter and the sampling procedure, with the goal of extending the time resolution to a range previously inaccessible by event analysis or by conventional noise analysis. The new method allows one to analyse fast binding events and/or the case when the single channel is not completely blocked by the substrate. We demonstrate the power of this approach by using as an example the interactions of meropenem, an antibiotic of the carbapenem family, with the OmpF porin that is considered to be one of the main pathways for antibiotics to enter Escherichia coli. The presence of meropenem in OmpF is detected by ion current blockages, and the on and off rates are estimated from the concentration dependence of the average ion current and of its power spectral density. The obtained average residence time of the antibiotic inside the channel is in the range of a few microseconds, i.e. more than 50 times smaller than the inverse cut-off frequency of the analogous filter.


Asunto(s)
Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/metabolismo , Canales Iónicos/fisiología , Modelos Teóricos , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA