Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.150
Filtrar
1.
Nat Commun ; 15(1): 3731, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702306

RESUMEN

Molecular recognition of proteins is key to their biological functions and processes such as protein-protein interactions (PPIs). The large binding interface involved and an often relatively flat binding surface make the development of selective protein-binding materials extremely challenging. A general method is reported in this work to construct protein-binding polymeric nanoparticles from cross-linked surfactant micelles. Preparation involves first dynamic covalent chemistry that encodes signature surface lysines on a protein template. A double molecular imprinting procedure fixes the binding groups on the nanoparticle for these lysine groups, meanwhile creating a binding interface complementary to the protein in size, shape, and distribution of acidic groups on the surface. These water-soluble nanoparticles possess excellent specificities for target proteins and sufficient affinities to inhibit natural PPIs such as those between cytochrome c (Cytc) and cytochrome c oxidase (CcO). With the ability to enter cells through a combination of energy-dependent and -independent pathways, they intervene apoptosis by inhibiting the PPI between Cytc and the apoptotic protease activating factor-1 (APAF1). Generality of the preparation and the excellent molecular recognition of the materials have the potential to make them powerful tools to probe protein functions in vitro and in cellulo.


Asunto(s)
Citocromos c , Complejo IV de Transporte de Electrones , Nanopartículas , Polímeros , Nanopartículas/química , Citocromos c/metabolismo , Citocromos c/química , Humanos , Polímeros/química , Polímeros/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/química , Impresión Molecular/métodos , Unión Proteica , Apoptosis , Micelas , Células HeLa , Animales
2.
J Inorg Biochem ; 249: 112367, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37742491

RESUMEN

Cytochrome c oxidase (CcO), also widely known as mitochondrial electron-transport-chain complex IV, is a multi-subunit transmembrane protein responsible for catalyzing the last step of the electron transport chain, dioxygen reduction to water, which is essential to the establishment and maintenance of the membrane proton gradient that drives ATP synthesis. Although many intermediates in the CcO catalytic cycle have been spectroscopically and/or computationally authenticated, the specifics regarding the IP intermediate, hypothesized to be a heme-Cu (hydro)peroxo species whose O-O bond homolysis is supported by a hydrogen-bonding network of water molecules, are largely obscured by the fast kinetics of the A (FeIII-O2•-/CuI/Tyr) â†’ PM (FeIV=O/CuII-OH/Tyr•) step. In this review, we have focused on the recent advancements in the design, development, and characterization of synthetic heme-peroxo­copper model complexes, which can circumvent the abovementioned limitation, for the investigation of the formation of IP and its O-O cleavage chemistry. Novel findings regarding (a) proton and electron transfer (PT/ET) processes, together with their contributions to exogenous phenol induced O-O cleavage, (b) the stereo-electronic tunability of the secondary coordination sphere (especially hydrogen-bonding) on the geometric and spin state alteration of the heme-peroxo­copper unit, and (c) a plausible mechanism for the Tyr-His cofactor biogenesis, are discussed in great detail. Additionally, since the ferric-superoxide and the ferryl-oxo (Compound II) species are critically involved in the CcO catalytic cycle, this review also highlights a few fundamental aspects of these heme-only (i.e., without copper) species, including the structural and reactivity influences of electron-donating trans-axial ligands and Lewis acid-promoted H-bonding.


Asunto(s)
Complejo IV de Transporte de Electrones , Oxígeno , Oxígeno/química , Complejo IV de Transporte de Electrones/química , Cobre/química , Compuestos Férricos/química , Protones , Hemo/química , Agua/metabolismo , Oxidación-Reducción
3.
J Am Chem Soc ; 145(41): 22305-22309, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37695261

RESUMEN

Cytochrome c oxidase (CcO) is a large membrane-bound hemeprotein that catalyzes the reduction of dioxygen to water. Unlike classical dioxygen binding hemeproteins with a heme b group in their active sites, CcO has a unique binuclear center (BNC) composed of a copper atom (CuB) and a heme a3 iron, where O2 binds and is reduced to water. CO is a versatile O2 surrogate in ligand binding and escape reactions. Previous time-resolved spectroscopic studies of the CO complexes of bovine CcO (bCcO) revealed that photolyzing CO from the heme a3 iron leads to a metastable intermediate (CuB-CO), where CO is bound to CuB, before it escapes out of the BNC. Here, with a pump-probe based time-resolved serial femtosecond X-ray crystallography, we detected a geminate photoproduct of the bCcO-CO complex, where CO is dissociated from the heme a3 iron and moved to a temporary binding site midway between the CuB and the heme a3 iron, while the locations of the two metal centers and the conformation of Helix-X, housing the proximal histidine ligand of the heme a3 iron, remain in the CO complex state. This new structure, combined with other reported structures of bCcO, allows for a clearer definition of the ligand dissociation trajectory as well as the associated protein dynamics.


Asunto(s)
Cobre , Complejo IV de Transporte de Electrones , Bovinos , Animales , Complejo IV de Transporte de Electrones/química , Oxidación-Reducción , Cobre/química , Ligandos , Oxígeno/química , Cristalografía por Rayos X , Hierro/química , Agua/metabolismo
4.
J Gen Appl Microbiol ; 69(2): 68-78, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37394433

RESUMEN

In thermophilic microorganisms, c-type cytochrome (cyt) proteins mainly function in the respiratory chain as electron carriers. Genome analyses at the beginning of this century revealed a variety of genes harboring the heme c motif. Here, we describe the results of surveying genes with the heme c motif, CxxCH, in a genome database comprising four strains of Thermus thermophilus, including strain HB8, and the confirmation of 19 c-type cytochromes among 27 selected genes. We analyzed the 19 genes, including the expression of four, by a bioinformatics approach to elucidate their individual attributes. One of the approaches included an analysis based on the secondary structure alignment pattern between the heme c motif and the 6th ligand. The predicted structures revealed many cyt c domains with fewer ß-strands, such as mitochondrial cyt c, in addition to the ß-strand unique to Thermus inserted in cyt c domains, as in T. thermophilus cyt c552 and caa3 cyt c oxidase subunit IIc. The surveyed thermophiles harbor potential proteins with a variety of cyt c folds. The gene analyses led to the development of an index for the classification of cyt c domains. Based on these results, we propose names for T. thermophilus genes harboring the cyt c fold.


Asunto(s)
Citocromos , Thermus thermophilus , Thermus thermophilus/genética , Thermus thermophilus/metabolismo , Transporte de Electrón , Citocromos/metabolismo , Thermus/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo
5.
Phys Chem Chem Phys ; 25(37): 25105-25115, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37461851

RESUMEN

As the terminal oxidase of cell respiration in mitochondria and aerobic bacteria, the proton pumping mechanism of ba3-type cytochrome c oxidase (CcO) of Thermus thermophiles is still not fully understood. Especially, the functions of key residues which were considered as the possible proton loading sites (PLSs) above the catalytic center, as well as water located above and within the catalytic center, remain unclear. In this work, molecular dynamic simulations were performed on a set of designed mutants of key residues (Asp287, Asp372, His376, and Glu126II). The results showed that Asp287 may not be a PLS, but it could modulate the ability of the proton transfer pathway to transfer protons through its salt bridge with Arg225. Maintaining the closed state of the water pool above the catalytic center is necessary for the participation of inside water molecules in proton transfer. Water molecules inside the water pool can form hydrogen bond chains with PLS to facilitate proton transfer. Additional quantum cluster models of the Fe-Cu metal catalytic center are established, indicating that when the proton is transferred from Tyr237, it is more likely to reach the OCu atom directly through only one water molecule. This work provides a more profound understanding of the functions of important residues and specific water molecules in the proton pumping mechanism of CcO.


Asunto(s)
Complejo IV de Transporte de Electrones , Bombas de Protones , Complejo IV de Transporte de Electrones/química , Protones , Agua/química , Simulación de Dinámica Molecular , Oxidación-Reducción
6.
Exp Parasitol ; 251: 108572, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37348640

RESUMEN

Copper can exist in an oxidized and a reduced form, which enables the metal to play essential roles as a catalytic co-factor in redox reactions in many organisms. Copper confers redox activity to the terminal electron transport chain cytochrome c oxidase protein. Cytochrome c oxidase in yeast obtains copper for the CuB site in the Cox1 subunit from Cox11 in association with Cox19. When copper is chelated in growth medium, Plasmodium falciparum parasite development in infected red blood cells is inhibited and excess copper is toxic for the parasite. The gene of a 26 kDa Plasmodium falciparum PfCox19 protein with two Cx9C Cox19 copper binding motifs, was cloned and expressed as a 66 kDa fusion protein with maltose binding protein and affinity purified (rMBP-PfCox19). rMBP-PfCox19 bound copper measured by: a bicinchoninic acid release assay; an in vivo bacterial host growth inhibition assay; ascorbate oxidation inhibition and differential scanning fluorimetry. The native protein was detected by antibodies raised against rMBP-PfCox19. PfCox19 binds copper and is predicted to associate with PfCox11 in the insertion of copper into the CuB site of Plasmodium cytochrome c oxidase. Characterisation of the proteins involved in Plasmodium spp. copper metabolism will help us understand the role of cytochrome c oxidase and this essential metal in Plasmodium homeostasis.


Asunto(s)
Cobre , Plasmodium falciparum , Plasmodium falciparum/genética , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/química
7.
Nature ; 615(7954): 934-938, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36949187

RESUMEN

Mitochondrial energy conversion requires an intricate architecture of the inner mitochondrial membrane1. Here we show that a supercomplex containing all four respiratory chain components contributes to membrane curvature induction in ciliates. We report cryo-electron microscopy and cryo-tomography structures of the supercomplex that comprises 150 different proteins and 311 bound lipids, forming a stable 5.8-MDa assembly. Owing to subunit acquisition and extension, complex I associates with a complex IV dimer, generating a wedge-shaped gap that serves as a binding site for complex II. Together with a tilted complex III dimer association, it results in a curved membrane region. Using molecular dynamics simulations, we demonstrate that the divergent supercomplex actively contributes to the membrane curvature induction and tubulation of cristae. Our findings highlight how the evolution of protein subunits of respiratory complexes has led to the I-II-III2-IV2 supercomplex that contributes to the shaping of the bioenergetic membrane, thereby enabling its functional specialization.


Asunto(s)
Microscopía por Crioelectrón , Complejo III de Transporte de Electrones , Complejo II de Transporte de Electrones , Complejo IV de Transporte de Electrones , Complejo I de Transporte de Electrón , Mitocondrias , Membranas Mitocondriales , Transporte de Electrón , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/ultraestructura , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/ultraestructura , Mitocondrias/química , Mitocondrias/enzimología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Membranas Mitocondriales/química , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/ultraestructura , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/ultraestructura , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Simulación de Dinámica Molecular , Sitios de Unión , Evolución Molecular
8.
Proc Natl Acad Sci U S A ; 119(38): e2207761119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095184

RESUMEN

Aerobic life is powered by membrane-bound enzymes that catalyze the transfer of electrons to oxygen and protons across a biological membrane. Cytochrome c oxidase (CcO) functions as a terminal electron acceptor in mitochondrial and bacterial respiratory chains, driving cellular respiration and transducing the free energy from O2 reduction into proton pumping. Here we show that CcO creates orientated electric fields around a nonpolar cavity next to the active site, establishing a molecular switch that directs the protons along distinct pathways. By combining large-scale quantum chemical density functional theory (DFT) calculations with hybrid quantum mechanics/molecular mechanics (QM/MM) simulations and atomistic molecular dynamics (MD) explorations, we find that reduction of the electron donor, heme a, leads to dissociation of an arginine (Arg438)-heme a3 D-propionate ion-pair. This ion-pair dissociation creates a strong electric field of up to 1 V Å-1 along a water-mediated proton array leading to a transient proton loading site (PLS) near the active site. Protonation of the PLS triggers the reduction of the active site, which in turn aligns the electric field vectors along a second, "chemical," proton pathway. We find a linear energy relationship of the proton transfer barrier with the electric field strength that explains the effectivity of the gating process. Our mechanism shows distinct similarities to principles also found in other energy-converting enzymes, suggesting that orientated electric fields generally control enzyme catalysis.


Asunto(s)
Complejo IV de Transporte de Electrones , Protones , Aerobiosis , Transporte de Electrón , Complejo IV de Transporte de Electrones/química , Oxígeno/metabolismo , Agua/química
9.
J Phys Chem Lett ; 13(39): 9165-9170, 2022 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-36166647

RESUMEN

Effects of interfacial interactions on the electrocatalytic activity of protein-tethered bilayer lipid membranes (ptBLMs) containing cytochrome c oxidase (CcO) for the oxygen reduction reaction are studied by using protein film electrochemistry and surface-enhanced infrared absorption (SEIRA) spectroscopy. Mammalian CcO was immobilized on a gold electrode via self-assembled monolayers (SAMs) of mixed alkanethiols. The protein orientation on the electrode is controlled by SAM-CcO interactions and is critical to the cytochrome c (cyt c) binding. The CcO-phospholipid and CcO-cyt c interactions modulate the electrocatalytic activity of CcO, and more densely packed ptBLMs show higher electrocatalytic activity. Our study indicates that spectroscopic and electrochemical studies of ptBLMs can provide insights into the effects of relatively weak protein-protein and protein-lipid interactions on the enzymatic activity of transmembrane enzymes.


Asunto(s)
Complejo IV de Transporte de Electrones , Oro , Animales , Biomimética , Citocromos c , Electrodos , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Oro/química , Membrana Dobles de Lípidos , Mamíferos/metabolismo , Oxígeno/metabolismo , Fosfolípidos
10.
J Mol Graph Model ; 117: 108309, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36037732

RESUMEN

In Acidithiobacillus ferrooxidans, proteins such as CcO are present in the electron transport pathway. They cause ferrous iron oxidation to ferric leading to the electron release. CcO has two copper atoms (CuA, CuB). CuA plays an important role in electron transfer. According to previous studies, the conversion of histidine to methionine in a similar protein increased the redox potential and was directly related to the number of electrons received. Also, the binding of methionine 233 to CuA and CuB in the wild protein structure is the reason for the selection of the H230 M mutation in the CuA site. Then, wild-type and H230 M mutant were simulated in the presence of a bilayer membrane POPC using the gromacs version 5.1.4. The changes performed in the H230 M mutant were evaluated by MD simulations analyzes. CcO and CoxA proteins are the last two proteins in the chain and were docked by the PatchDock server. By H230 M mutation, the connection between CuA and M230 weakens. The M230 moves further away from CuA, resulting become more flexible. Therefore, the Methionine gets closer to E149 of the CoxA leading to the higher stability of the CcO/CoxA complex. The results of RMSF analysis at the mutation point showed a significant increase. This indicates more flexibility in the active site. And leads to an increase in E0 in the mutation point, an increase in the rate of electron reception, and an improved bioleaching process.


Asunto(s)
Cobre , Complejo IV de Transporte de Electrones , Cobre/química , Transporte de Electrón , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Electrones , Histidina , Hierro/metabolismo , Metionina/genética , Metionina/metabolismo , Oxidación-Reducción , Mutación Puntual
11.
Proc Natl Acad Sci U S A ; 119(30): e2205228119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858451

RESUMEN

The mitochondrial electron transport chain maintains the proton motive force that powers adenosine triphosphate (ATP) synthesis. The energy for this process comes from oxidation of reduced nicotinamide adenine dinucleotide (NADH) and succinate, with the electrons from this oxidation passed via intermediate carriers to oxygen. Complex IV (CIV), the terminal oxidase, transfers electrons from the intermediate electron carrier cytochrome c to oxygen, contributing to the proton motive force in the process. Within CIV, protons move through the K and D pathways during turnover. The former is responsible for transferring two protons to the enzyme's catalytic site upon its reduction, where they eventually combine with oxygen and electrons to form water. CIV is the main site for respiratory regulation, and although previous studies showed that steroid binding can regulate CIV activity, little is known about how this regulation occurs. Here, we characterize the interaction between CIV and steroids using a combination of kinetic experiments, structure determination, and molecular simulations. We show that molecules with a sterol moiety, such as glyco-diosgenin and cholesteryl hemisuccinate, reversibly inhibit CIV. Flash photolysis experiments probing the rapid equilibration of electrons within CIV demonstrate that binding of these molecules inhibits proton uptake through the K pathway. Single particle cryogenic electron microscopy (cryo-EM) of CIV with glyco-diosgenin reveals a previously undescribed steroid binding site adjacent to the K pathway, and molecular simulations suggest that the steroid binding modulates the conformational dynamics of key residues and proton transfer kinetics within this pathway. The binding pose of the sterol group sheds light on possible structural gating mechanisms in the CIV catalytic cycle.


Asunto(s)
Diosgenina , Complejo IV de Transporte de Electrones , Esteroides , Animales , Sitios de Unión , Dominio Catalítico/efectos de los fármacos , Bovinos , Diosgenina/farmacología , Transporte de Electrón , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/química , Oxidación-Reducción , Oxígeno/metabolismo , Conformación Proteica , Protones , Esteroides/química , Esteroides/farmacología , Esteroles
12.
Nat Commun ; 13(1): 3615, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35750769

RESUMEN

Mitochondrial cytochrome c oxidase (CcO) or respiratory chain complex IV is a heme aa3-copper oxygen reductase containing metal centers essential for holo-complex biogenesis and enzymatic function that are assembled by subunit-specific metallochaperones. The enzyme has two copper sites located in the catalytic core subunits. The COX1 subunit harbors the CuB site that tightly associates with heme a3 while the COX2 subunit contains the binuclear CuA site. Here, we report that in human cells the CcO copper chaperones form macromolecular assemblies and cooperate with several twin CX9C proteins to control heme a biosynthesis and coordinate copper transfer sequentially to the CuA and CuB sites. These data on CcO illustrate a mechanism that regulates the biogenesis of macromolecular enzymatic assemblies with several catalytic metal redox centers and prevents the accumulation of cytotoxic reactive assembly intermediates.


Asunto(s)
Complejo IV de Transporte de Electrones , Oxidorreductasas , Cobre/metabolismo , Citocromos c/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Hemo/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxidorreductasas/metabolismo
14.
J Biol Chem ; 298(4): 101799, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35257742

RESUMEN

Cytochrome c oxidase (CcO) is the terminal enzyme in the electron transfer chain in the inner membrane of mitochondria. It contains four metal redox centers, two of which, CuB and heme a3, form the binuclear center (BNC), where dioxygen is reduced to water. Crystal structures of CcO in various forms have been reported, from which ligand-binding states of the BNC and conformations of the protein matrix surrounding it have been deduced to elucidate the mechanism by which the oxygen reduction chemistry is coupled to proton translocation. However, metal centers in proteins can be susceptible to X-ray-induced radiation damage, raising questions about the reliability of conclusions drawn from these studies. Here, we used microspectroscopy-coupled X-ray crystallography to interrogate how the structural integrity of bovine CcO in the fully oxidized state (O) is modulated by synchrotron radiation. Spectroscopic data showed that, upon X-ray exposure, O was converted to a hybrid O∗ state where all the four metal centers were reduced, but the protein matrix was trapped in the genuine O conformation and the ligands in the BNC remained intact. Annealing the O∗ crystal above the glass transition temperature induced relaxation of the O∗ structure to a new R∗ structure, wherein the protein matrix converted to the fully reduced R conformation with the exception of helix X, which partly remained in the O conformation because of incomplete dissociation of the ligands from the BNC. We conclude from these data that reevaluation of reported CcO structures obtained with synchrotron light sources is merited.


Asunto(s)
Complejo IV de Transporte de Electrones , Metales , Rayos X , Animales , Bovinos , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/efectos de la radiación , Ligandos , Metales/química , Oxidación-Reducción , Estructura Terciaria de Proteína/efectos de la radiación , Reproducibilidad de los Resultados , Temperatura
15.
Chemphyschem ; 23(7): e202100831, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35142420

RESUMEN

Mössbauer isomer shift and quadrupole splitting properties have been calculated using the OLYP-D3(BJ) density functional method on previously obtained (W.-G. Han Du, et al., Inorg Chem. 2020, 59, 8906-8915) geometry optimized Fea33+ -H2 O-CuB2+ dinuclear center (DNC) clusters of the resting oxidized (O state) "as-isolated" cytochrome c oxidase (CcO). The calculated results are highly consistent with the available experimental observations. The calculations have also shown that the structural heterogeneities of the O state DNCs implicated by the Mössbauer experiments are likely consequences of various factors, particularly the variable positions of the central H2 O molecule between the Fea33+ and CuB2+ sites in different DNCs, whether or not this central H2 O molecule has H-bonding interaction with another H2 O molecule, the different spin states having similar energies for the Fea33+ sites, and whether the Fea33+ and CuB2+ sites are ferromagnetically or antiferromagnetically spin-coupled.


Asunto(s)
Complejo IV de Transporte de Electrones , Teoría Funcional de la Densidad , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Oxidación-Reducción
16.
Parasit Vectors ; 15(1): 26, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35033159

RESUMEN

BACKGROUND: Microsporidia is a large group of eukaryotic obligate intracellular spore-forming parasites, of which 17 species can cause microsporidiosis in humans. Most human-infecting microsporidians belong to the genera Enterocytozoon and Encephalitozoon. To date, only five microsporidian species, including Encephalitozoon-like, have been found in hard ticks (Ixodidae) using microscopic methods, but no sequence data are available for them. Furthermore, no widespread screening for microsporidian-infected ticks based on DNA analysis has been carried out to date. Thus, in this study, we applied a recently developed DNA metabarcoding method for efficient microsporidian DNA identification to assess the role of ticks as potential vectors of microsporidian species causing diseases in humans. METHODS: In total, 1070 (493 juvenile and 577 adult) unfed host-seeking Ixodes ricinus ticks collected at urban parks in the city of Poznan, Poland, and 94 engorged tick females fed on dogs and cats were screened for microsporidian DNA. Microsporidians were detected by PCR amplification and sequencing of the hypervariable V5 region of 18S rRNA gene (18S profiling) using the microsporidian-specific primer set. Tick species were identified morphologically and confirmed by amplification and sequencing of the shortened fragment of cytochrome c oxidase subunit I gene (mini-COI). RESULTS: All collected ticks were unambiguously assigned to I. ricinus. Potentially zoonotic Encephalitozoon intestinalis was identified in three fed ticks (3.2%) collected from three different dogs. In eight unfed host-seeking ticks (0.8%), including three males (1.1%), two females (0.7%) and three nymphs (0.7%), the new microsporidian sequence representing a species belonging to the genus Endoreticulatus was identified. CONCLUSIONS: The lack of zoonotic microsporidians in host-seeking ticks suggests that I. ricinus is not involved in transmission of human-infecting microsporidians. Moreover, a very low occurrence of the other microsporidian species in both fed and host-seeking ticks implies that mechanisms exist to defend ticks against infection with these parasites.


Asunto(s)
Vectores Arácnidos/microbiología , Ixodes/microbiología , Microsporidios/fisiología , Animales , Secuencia de Bases , Enfermedades de los Gatos/parasitología , Gatos , Código de Barras del ADN Taxonómico , ADN de Hongos/química , ADN de Hongos/aislamiento & purificación , ADN Ribosómico/química , Enfermedades de los Perros/parasitología , Perros , Complejo IV de Transporte de Electrones/química , Femenino , Masculino , Microsporidios/clasificación , Parques Recreativos , Filogenia , Polonia , Prevalencia , ARN Ribosómico 18S/genética , Análisis de Secuencia de ADN , Infestaciones por Garrapatas/parasitología , Infestaciones por Garrapatas/veterinaria
17.
PLoS One ; 17(1): e0262122, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35025933

RESUMEN

Due to the lack of visible barriers to gene flow, it was a long-standing assumption that marine coastal species are widely distributed, until molecular studies revealed geographically structured intraspecific genetic differentiation in many taxa. Historical events of sea level changes during glacial periods are known to have triggered sequential disjunctions and genetic divergences among populations, especially of coastal organisms. The Parasesarma bidens species complex so far includes three named plus potentially cryptic species of estuarine brachyuran crabs, distributed along East to Southeast Asia. The aim of the present study is to address phylogeography and uncover real and hidden biological diversity within this complex, by revealing the underlying genetic structure of populations and species throughout their distribution ranges from Japan to West Papua, with a comparison of mitochondrial COX1 and 16S rRNA gene sequences. Our results reveal that the P. bidens species complex consists of at least five distinct clades, resulting from four main cladogenesis events during the mid to late Pleistocene. Among those clades, P. cricotum and P. sanguimanus are recovered as monophyletic taxa. Geographically restricted endemic clades are encountered in southeastern Indonesia, Japan and China respectively, whereas the Philippines and Taiwan share two clades. As individuals of the Japanese clade can also be found in Taiwan, we provide evidence of a third lineage and the occurrence of a potential cryptic species on this island. Ocean level retreats during Pleistocene ice ages and present oceanic currents appear to be the main triggers for the divergences of the five clades that are here addressed as the P. bidens complex. Secondary range expansions converted Taiwan into the point of maximal overlap, sharing populations with Japan and the Philippines, but not with mainland China.


Asunto(s)
Biodiversidad , Braquiuros/clasificación , Animales , Braquiuros/genética , China , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Fósiles/historia , Genética de Población , Historia Antigua , Indonesia , Japón , Filipinas , Filogenia , Filogeografía , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Análisis de Secuencia de ADN , Taiwán
18.
Nat Commun ; 13(1): 545, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35087070

RESUMEN

Proton-translocating respiratory complexes assemble into supercomplexes that are proposed to increase the efficiency of energy conversion and limit the production of harmful reactive oxygen species during aerobic cellular respiration. Cytochrome bc complexes and cytochrome aa3 oxidases are major drivers of the proton motive force that fuels ATP generation via respiration, but how wasteful electron- and proton transfer is controlled to enhance safety and efficiency in the context of supercomplexes is not known. Here, we address this question with the 2.8 Å resolution cryo-EM structure of the cytochrome bcc-aa3 (III2-IV2) supercomplex from the actinobacterium Corynebacterium glutamicum. Menaquinone, substrate mimics, lycopene, an unexpected Qc site, dioxygen, proton transfer routes, and conformational states of key protonable residues are resolved. Our results show how safe and efficient energy conversion is achieved in a respiratory supercomplex through controlled electron and proton transfer. The structure may guide the rational design of drugs against actinobacteria that cause diphtheria and tuberculosis.


Asunto(s)
Actinobacteria/metabolismo , Corynebacterium glutamicum/metabolismo , Citocromos/química , Citocromos/metabolismo , Oxidorreductasas/metabolismo , Benzoquinonas/química , Sitios de Unión , Microscopía por Crioelectrón , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético , Modelos Moleculares , Oxígeno/metabolismo , Fuerza Protón-Motriz
19.
Biochemistry ; 61(22): 2506-2521, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-21028883

RESUMEN

The functioning of cytochrome c oxidases involves orchestration of long-range electron transfer (ET) events among the four redox active metal centers. We report the temperature dependence of electron transfer from the CuAr site to the low-spin heme-(a)bo site, i.e., CuAr + heme-a(b)o → CuAo + heme-a(b)r in three structurally characterized enzymes: A-type aa3 from Paracoccus denitrificans (PDB code 3HB3) and bovine heart tissue (PDB code 2ZXW), and the B-type ba3 from T. thermophilus (PDB codes 1EHK and 1XME). k,T data sets were obtained with the use of pulse radiolysis as described previously. Semiclassical Marcus theory revealed that λ varies from 0.74 to 1.1 eV, Hab, varies from ∼2 × 10-5 eV (0.16 cm-1) to ∼24 × 10-5 eV (1.9 cm-1), and ßD varies from 9.3 to 13.9. These parameters are consistent with diabatic electron tunneling. The II-Asp111Asn CuA mutation in cytochrome ba3 had no effect on the rate of this reaction whereas the II-Met160Leu CuA-mutation was slower by an amount corresponding to a decreased driving force of ∼0.06 eV. The structures support the presence of a common, electron-conducting "wire" between CuA and heme-a(b). The transfer of an electron from the low-spin heme to the high-spin heme, i.e., heme-a(b)r + heme-a3o → heme-a(b)o + heme-a3r, was not observed with the A-type enzymes in our experiments but was observed with the Thermus ba3; its Marcus parameters are λ = 1.5 eV, Hab = 26.6 × 10-5 eV (2.14 cm-1), and ßD = 9.35, consistent also with diabatic electron tunneling between the two hemes. The II-Glu15Ala mutation of the K-channel structure, ∼ 24 Å between its CA and Fe-a3, was found to completely block heme-br to heme-a3o electron transfer. A structural mechanism is suggested to explain these observations.


Asunto(s)
Complejo IV de Transporte de Electrones , Thermus thermophilus , Bovinos , Animales , Complejo IV de Transporte de Electrones/química , Grupo Citocromo b/química , Electrones , Radiólisis de Impulso , Temperatura , Oxidación-Reducción , Hemo/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...