Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Biol Chem ; 298(12): 102701, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36395884

RESUMEN

The L-type Ca2+ channel CaV1.2 controls gene expression, cardiac contraction, and neuronal activity. Calmodulin (CaM) governs CaV1.2 open probability (Po) and Ca2+-dependent inactivation (CDI) but the mechanisms remain unclear. Here, we present electrophysiological data that identify a half Ca2+-saturated CaM species (Ca2/CaM) with Ca2+ bound solely at the third and fourth EF-hands (EF3 and EF4) under resting Ca2+ concentrations (50-100 nM) that constitutively preassociates with CaV1.2 to promote Po and CDI. We also present an NMR structure of a complex between the CaV1.2 IQ motif (residues 1644-1665) and Ca2/CaM12', a calmodulin mutant in which Ca2+ binding to EF1 and EF2 is completely disabled. We found that the CaM12' N-lobe does not interact with the IQ motif. The CaM12' C-lobe bound two Ca2+ ions and formed close contacts with IQ residues I1654 and Y1657. I1654A and Y1657D mutations impaired CaM binding, CDI, and Po, as did disabling Ca2+ binding to EF3 and EF4 in the CaM34 mutant when compared to WT CaM. Accordingly, a previously unappreciated Ca2/CaM species promotes CaV1.2 Po and CDI, identifying Ca2/CaM as an important mediator of Ca signaling.


Asunto(s)
Canales de Calcio Tipo L , Calmodulina , Calmodulina/metabolismo , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Unión Proteica , Mutación , Calcio/metabolismo
2.
Nat Immunol ; 10(7): 761-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19525969

RESUMEN

Granule-mediated cytotoxicity is the main effector mechanism of cytotoxic CD8+ T cells. We report that CD8+ T cells from acid sphingomyelinase (ASMase)-deficient (ASMase-KO) mice are defective in exocytosis of cytolytic effector molecules; this defect resulted in attenuated cytotoxic activity of ASMase-KO CD8+ T cells and delayed elimination of lymphocytic choriomeningitis virus from ASMase-KO mice. Cytolytic granules of ASMase-KO and wild-type CD8+ T cells were equally loaded with granzymes and perforin, and correctly directed to the immunological synapse. In wild-type CD8+ T cells, secretory granules underwent shrinkage by 82% after fusion with the plasma membrane. In ASMase-KO CD8+ T cells, the contraction of secretory granules was markedly impaired. Thus, ASMase is required for contraction of secretory granules and expulsion of cytotoxic effector molecules.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Vesículas Secretoras/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Linfocitos T/inmunología , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CCL5/metabolismo , Femenino , Granzimas/genética , Granzimas/metabolismo , Immunoblotting , Sinapsis Inmunológicas/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Perforina/genética , Perforina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vesículas Secretoras/enzimología , Esfingomielina Fosfodiesterasa/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
3.
Neurobiol Dis ; 136: 104721, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31887354

RESUMEN

Voltage-gated calcium-channels (VGCCs) are heteromers consisting of several subunits. Mutations in the genes coding for VGCC subunits have been reported to be associated with autism spectrum disorder (ASD). In a previous study, we identified electrophysiologically relevant missense mutations of CaVß2 subunits of VGCCs. From this, we derived the hypothesis that several CaVß2-mutations associated with ASD show common features sensitizing LTCCs and/or enhancing currents. Using a CaVß2d backbone, we performed extensive whole-cell and single-channel patch-clamp analyses of Ba2+ currents carried by Cav1.2 pore subunits co-transfected with the previously described CaVß2 mutations (G167S, S197F) as well as a recently identified point mutation (V2D). Furthermore, the interaction of the mutated CaVß2d subunits with the RGK protein Gem was analyzed by co-immunoprecipitation assays and electrophysiological studies. Patch-clamp analyses revealed that all mutations increase Ba2+ currents, e.g. by decreasing inactivation or increasing fraction of active sweeps. All CaVß2 mutations interact with Gem, but differ in the extent and characteristics of modulation by this RGK protein (e.g. decrease of fraction of active sweeps: CaVß2d_G167S > CaVß2d_V2D > CaVß2d_S197F). In conclusion, patch-clamp recordings of ASD-associated CaVß2d mutations revealed differential modulation of Ba2+ currents carried by CaV1.2 suggesting kind of an "electrophysiological fingerprint" each. The increase in current finally observed with all CaVß2d mutations analyzed might contribute to the complex pathophysiology of ASD and by this indicate a possible underlying molecular mechanism.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Canales de Calcio Tipo L/fisiología , Proteínas de Unión al GTP Monoméricas/fisiología , Mutación Missense/fisiología , Calcio/fisiología , Células HEK293 , Humanos , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp/métodos
4.
Biophys J ; 116(5): 836-846, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773296

RESUMEN

Voltage-dependent calcium (CaV) 1.3 channels are involved in the control of cellular excitability and pacemaking in neuronal, cardiac, and sensory cells. Various proteins interact with the alternatively spliced channel C-terminus regulating gating of CaV1.3 channels. Binding of a regulatory calcium-binding protein calmodulin (CaM) to the proximal C-terminus leads to the boosting of channel activity and promotes calcium-dependent inactivation (CDI). The C-terminal modulator domain (CTM) of CaV1.3 channels can interfere with the CaM binding, thereby inhibiting channel activity and CDI. Here, we compared single-channel gating behavior of two natural CaV1.3 splice isoforms: the long CaV1.342 with the full-length CTM and the short CaV1.342A with the C-terminus truncated before the CTM. We found that CaM regulation of CaV1.3 channels is dynamic on a minute timescale. We observed that at equilibrium, single CaV1.342 channels occasionally switched from low to high open probability, which perhaps reflects occasional binding of CaM despite the presence of CTM. Similarly, when the amount of the available CaM in the cell was reduced, the short CaV1.342A isoform showed patterns of the low channel activity. CDI also underwent periodic changes with corresponding kinetics in both isoforms. Our results suggest that the competition between CTM and CaM is influenced by calcium, allowing further fine-tuning of CaV1.3 channel activity for particular cellular needs.


Asunto(s)
Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/metabolismo , Calmodulina/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico , Cinética , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo
5.
Acta Myol ; 33(1): 22-33, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24843232

RESUMEN

We studied the consequences of the Nav1.4 mutation R1448H that is situated in the fourth voltage sensor of the channel and causes paramyotonia, a cold-induced myotonia followed by weakness. Previous work showed that the mutation uncouples inactivation from activation. We measured whole-cell Na(+) currents at 10, 15, 20, and 25°C using HEK293 cells stably transfected with wildtype (WT) and R1448H Na(+) channels. A Markov model was developed the parameters of which reproduced the data measured on WT and R1448H channels in the whole voltage and temperature range. It required an additional transient inactivated state and an additional closed-state inactivation transition not previously described. The model was used to predict single-channel properties, free energy barriers and temperature dependence of rates. It allowed us to draw the following conclusions: i) open-state inactivation results from a two-step process; ii) the channel re-openings that cause paramyotonia originate from enhanced deactivation/reactivation and not from destabilized inactivation; iii) the closed-state inactivation of R1448H is strikingly enhanced. We assume that latter explains the episodic weakness following cold-induced myotonia.


Asunto(s)
Frío , Activación del Canal Iónico/fisiología , Miotonía/genética , Humanos , Riñón/citología , Mutación , Miotonía/fisiopatología , Técnicas de Placa-Clamp , Canales de Sodio/genética , Transfección
6.
Naunyn Schmiedebergs Arch Pharmacol ; 395(4): 459-470, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35122502

RESUMEN

Voltage-gated calcium channel (VGCC) subunits have been genetically associated with autism spectrum disorders (ASD). The properties of the pore-forming VGCC subunit are modulated by auxiliary ß-subunits, which exist in four isoforms (CaVß1-4). Our previous findings suggested that activation of L-type VGCCs is a common feature of CaVß2 subunit mutations found in ASD patients. In the current study, we functionally characterized a novel CaVß1b variant (p.R296C) identified in an ASD patient. We used whole-cell and single-channel patch clamp to study the effect of CaVß1b_R296C on the function of L- and N-type VGCCs. Furthermore, we used co-immunoprecipitation followed by Western blot to evaluate the interaction of the CaVß1b-subunits with the RGK-protein Gem. Our data obtained at both, whole-cell and single-channel levels, show that compared to a wild-type CaVß1b, the CaVß1b_R296C variant inhibits L- and N-type VGCCs. Interaction with and modulation by the RGK-protein Gem seems to be intact. Our findings indicate functional effects of the CaVß1b_R296C variant differing from that attributed to CaVß2 variants found in ASD patients. Further studies have to detail the effects on different VGCC subtypes and on VGCC expression.


Asunto(s)
Trastorno del Espectro Autista , Canales de Calcio Tipo L , Canales de Calcio Tipo N , Trastorno del Espectro Autista/genética , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Humanos
7.
Biophys J ; 101(11): 2661-70, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22261054

RESUMEN

Voltage-dependent Ca(2+) channels are heteromultimers of Ca(V)α(1) (pore), Ca(V)ß- and Ca(V)α(2)δ-subunits. The stoichiometry of this complex, and whether it is dynamically regulated in intact cells, remains controversial. Fortunately, Ca(V)ß-isoforms affect gating differentially, and we chose two extremes (Ca(V)ß(1a) and Ca(V)ß(2b)) regarding single-channel open probability to address this question. HEK293α(1C) cells expressing the Ca(V)1.2 subunit were transiently transfected with Ca(V)α(2)δ1 alone or with Ca(V)ß(1a), Ca(V)ß(2b), or (2:1 or 1:1 plasmid ratio) combinations. Both Ca(V)ß-subunits increased whole-cell current and shifted the voltage dependence of activation and inactivation to hyperpolarization. Time-dependent inactivation was accelerated by Ca(V)ß(1a)-subunits but not by Ca(V)ß(2b)-subunits. Mixtures induced intermediate phenotypes. Single channels sometimes switched between periods of low and high open probability. To validate such slow gating behavior, data were segmented in clusters of statistically similar open probability. With Ca(V)ß(1a)-subunits alone, channels mostly stayed in clusters (or regimes of alike clusters) of low open probability. Increasing Ca(V)ß(2b)-subunits (co-)expressed (1:2, 1:1 ratio or alone) progressively enhanced the frequency and total duration of high open probability clusters and regimes. Our analysis was validated by the inactivation behavior of segmented ensemble averages. Hence, a phenotype consistent with mutually exclusive and dynamically competing binding of different Ca(V)ß-subunits is demonstrated in intact cells.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Activación del Canal Iónico/fisiología , Subunidades de Proteína/metabolismo , Células HEK293 , Humanos , Cinética , Unión Proteica , Factores de Tiempo
8.
Pflugers Arch ; 459(3): 399-411, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19821165

RESUMEN

Voltage-dependent calcium channel (Ca(v)) pores are modulated by cytosolic beta subunits. Four beta-subunit genes and their splice variants offer a wide structural array for tissue- or disease-specific biophysical gating phenotypes. For instance, the length of the N terminus of beta(2) subunits has major effects on activation and inactivation rates. We tested whether a similar mechanism principally operates in a beta(1) subunit. Wild-type beta(1a) subunit (N terminus length 60 aa) and its newly generated N-terminal deletion mutants (51, 27 and 18 aa) were examined within recombinant L-type calcium channel complexes (Ca(v)1.2 and alpha(2)delta2) in HEK293 cells at the whole-cell and single-channel level. Whole-cell currents were enhanced by co-transfection of the full-length beta(1a) subunit and by all truncated constructs. Voltage dependence of steady-state activation and inactivation did not depend on N terminus length, but inactivation rate was diminished by N terminus truncation. This was confirmed at the single-channel level, using ensemble average currents. Additionally, gating properties were estimated by Markov modeling. In confirmation of the descriptive analysis, inactivation rate, but none of the other transition rates, was reduced by shortening of the beta(1a) subunit N terminus. Our study shows that the length-dependent mechanism of modulating inactivation kinetics of beta(2) calcium channel subunits can be confirmed and extended to the beta(1) calcium channel subunit.


Asunto(s)
Canales de Calcio Tipo L , Activación del Canal Iónico/fisiología , Mutación , Isoformas de Proteínas , Subunidades de Proteína , Empalme Alternativo , Secuencia de Aminoácidos , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Línea Celular , Humanos , Cadenas de Markov , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
9.
J Physiol ; 586(22): 5349-66, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18818244

RESUMEN

The eight members of the calcium channel gamma subunit family are integral membrane proteins that regulate the expression and behaviour of voltage and ligand gated ion channels. While a subgroup consisting of gamma(2), gamma(3), gamma(4) and gamma(8) (the TARPs) modulate AMPA receptor localization and function, the gamma(1) and gamma(6) subunits conform to the original description of these proteins as regulators of voltage gated calcium channels. We have previously shown that the gamma(6) subunit is highly expressed in atrial myocytes and that it is capable of acting as a negative modulator of low voltage activated calcium current. In this study we extend our understanding of gamma(6) subunit modulation of low voltage activated calcium current. Using engineered chimeric constructs, we demonstrate that the first transmembrane domain (TM1) of gamma(6) is necessary for its inhibitory effect on Cav3.1 current. Mutational analysis is then used to identify a unique GxxxA motif within TM1 that is required for the function of the subunit strongly suggesting the involvement of helix-helix interactions in its effects. Results from co-immunoprecipitation experiments confirm a physical association of gamma(6) with the Cav3.1 channel in both HEK cells and atrial myocytes. Single channel analysis reveals that binding of gamma(6) reduces channel availability for activation. Taken together, the results of this study provide both a molecular and a mechanistic framework for understanding the unique ability of the gamma(6) calcium channel subunit to modulate low voltage activated (Cav3.1) calcium current density.


Asunto(s)
Canales de Calcio/química , Canales de Calcio/metabolismo , Secuencias de Aminoácidos , Animales , Canales de Calcio/genética , Canales de Calcio Tipo T/química , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Línea Celular , Electrofisiología , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos , Miocitos Cardíacos/metabolismo , Subunidades de Proteína , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido
10.
J Mol Biol ; 357(1): 313-24, 2006 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-16426636

RESUMEN

Single-molecule fluorescence (Förster) resonance energy transfer (FRET) experiments were performed on surface-immobilized RNase H molecules as a function of the concentration of the chemical denaturant guanidinium chloride (GdmCl). For comparison, we measured ensemble FRET on RNase H solutions. The single-molecule approach allowed us to study FRET distributions of the subpopulation of unfolded molecules without interference from the folded population. The unfolded ensemble experienced a continuous shift of the FRET efficiency distribution with increasing concentration of GdmCl, indicating a heterogeneous population of expanding, unfolded polypeptide chains. We have analyzed the behavior of the unfolded state quantitatively with a model in which the unfolded state is described by a continuum of substates, with the free energy of each substate linearly coupled to its m-value, the proportionality coefficient between free energy and denaturant activity. By fitting this model to the data, we have derived energetic and structural parameters that describe the unfolded state ensemble. Specifically, we have found that the average size of the unfolded state increases from 23-38 A between 0 and 6 M denaturant. Excellent agreement was achieved between the fitted model and our FRET measurements, and with previously published nuclear magnetic resonance and small-angle X-ray scattering data.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , Guanidina/química , Desnaturalización Proteica , Ribonucleasa H/química , Interpretación Estadística de Datos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Matemática , Modelos Químicos , Conformación Proteica , Ribonucleasa H/metabolismo
11.
Pflugers Arch ; 454(4): 605-14, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17333247

RESUMEN

L-type calcium-channel mutations causing hypokalemic periodic paralysis type 1 (HypoPP-1) have pronounced "loss-of-function" features and stabilize the less-selective second open state O(2), as we demonstrated in the companion paper. Here, we compared the effects of the L-type calcium-channel activator (+/-)BayK 8644 (BayK) on the heterologously expressed wild-type (WT) calcium channel, rabbit Cav1.2 HypoPP-1 analogs, and two double mutants (R650H/R1362H, R650H/R1362G). Our goal was to elucidate (1) whether the "loss-of-function" in HypoPP-1 can be compensated by BayK application, (2) how the less-selective open state is affected by BayK in WT and HypoPP-1 mutants, as well as (3) to gain an insight into BayK mechanism of action. Ionic currents were examined by whole-cell patch-clamp and analyzed by the global-fitting procedure. Our results imply that (1) BayK promotes channel activation, but equalized the differences among the WT and mutants, thus attenuating HypoPP-related effects on activation and deactivation; (2) BayK binds to the first open state O(1), and then serves as a catalyst for O(2) formation; (3) binding of BayK is impaired in the HypoPP mutants, thus affecting the formation of the less-selective second open state; (4) BayK affects cooperativity between the single HypoPP-1 mutations at all stages of the channel gating; and (5) BayK favoring of O(2) lowers calcium-channel selectivity.


Asunto(s)
Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/genética , Parálisis Periódica Hipopotasémica/genética , Activación del Canal Iónico/efectos de los fármacos , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/fisiología , Línea Celular , Humanos , Parálisis Periódica Hipopotasémica/fisiopatología , Activación del Canal Iónico/fisiología , Oxígeno/metabolismo , Técnicas de Placa-Clamp
12.
Pflugers Arch ; 454(3): 495-505, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17333249

RESUMEN

Hypokalemic periodic paralysis type 1 (HypoPP-1) is a hereditary muscular disorder caused by point mutations in the gene encoding the voltage-gated Ca(2+) channel alpha subunit (Ca(v)1.1). Despite extensive research, the results on HypoPP-1 mutations are minor and controversial, as it is difficult to analyse Ca(2+) channel activation macroscopically due to an existence of two open states. In this study, we heterologously expressed the wild-type and HypoPP-1 mutations introduced into the rabbit cardiac Ca(2+) channel (R650H, R1362H, R1362G) in HEK-293 cells. To examine the cooperative effects of the mutations on channel gating, we expressed two double mutants (R650H/R1362H, R650H/R1362G). We performed whole-cell patch-clamp and, to obtain more information, applied a global fitting procedure whereby several current traces elicited by different potentials were simultaneously fit to the kinetic model containing four closed, two open and two inactivated states. We found that all HypoPP-1 mutations have "loss-of-function" features: D4/S4 mutations shift the equilibrium to the closed states, which results in reduced open probability, shorter openings and, therefore, in smaller currents, and the D2/S4 mutant slows the activation. In addition, HypoPP-1 histidine mutants favored the second open state O(2) with a possibly lower channel selectivity. Cooperativity between the D2/S4 and D4/S4 HypoPP-1 mutations manifested in dominant effects of the D4/S4 mutations on kinetics of the double mutants, suggesting different roles of D2/S4 and D4/S4 voltage sensors in the gating of voltage-gated calcium channels.


Asunto(s)
Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Parálisis Periódica Hipopotasémica/genética , Parálisis Periódica Hipopotasémica/metabolismo , Mutación Puntual , Sustitución de Aminoácidos , Animales , Línea Celular , Humanos , Técnicas In Vitro , Activación del Canal Iónico , Cinética , Potenciales de la Membrana , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Am J Pathol ; 170(2): 755-63, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17255341

RESUMEN

C-reactive protein (CRP), the prototype human acute phase protein, is widely regarded as a key player in cardiovascular disease, but the identity of its cellular receptor is still under debate. By using ultrasensitive confocal imaging analysis, we have studied CRP binding to transfected COS-7 cells expressing the high-affinity IgG receptor FcgammaRI. Here we show that CRP binds to FcgammaRI on intact cells, with a kd of 10+/-3 micromol/L. Transfection of COS-7 cells with a plasmid coding for both FcgammaRI and its functional counterpart, the gamma-chain, markedly increases CRP affinity to FcgammaRI, resulting in a kd of 0.35+/-0.10 micromol/L. The affinity increase results from an approximately 30-fold enhanced association rate coefficient. The pronounced enhancement of affinity by the gamma-chain suggests its crucial involvement in the CRP receptor interaction, possibly by mediating interactions between the transmembrane moieties of the receptors. Dissociation of CRP from the cell surfaces cannot be detected throughout the time course of several hours and is thus extremely slow. Considering the pentameric structure of CRP, this result indicates that multivalent binding and receptor clustering are crucially involved in the interaction of CRP with nucleated cells.


Asunto(s)
Proteína C-Reactiva/metabolismo , Agregación de Receptores , Receptores de IgG/metabolismo , Animales , Proteína C-Reactiva/química , Células COS , Chlorocebus aethiops , Humanos , Unión Proteica , Estructura Cuaternaria de Proteína , Receptores de IgG/química , Receptores de IgG/genética
14.
Proc Natl Acad Sci U S A ; 102(43): 15471-6, 2005 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-16221762

RESUMEN

Proteins are highly complex systems, exhibiting a substantial degree of structural variability in their folded state. In the presence of denaturants, the heterogeneity is greatly enhanced, and fluctuations among vast numbers of folded and unfolded conformations occur via many different pathways. Here, we have studied the structure and dynamics of the small enzyme ribonuclease HI (RNase H) in the presence of the chemical denaturant guanidinium chloride (GdmCl) using single-molecule fluorescence microscopy, with a particular focus on the characterization of the unfolded-state ensemble. A dye pair was specifically attached to the enzyme to measure structural changes through Förster resonance energy transfer (FRET). Enzyme immobilization on star-polymer surfaces that were specially developed for negligible interaction with folded and unfolded proteins enabled us to monitor conformational changes of individual proteins for several hundred seconds. FRET efficiency histograms were calculated from confocal scan images. They showed an expansion of the unfolded proteins with increasing GdmCl concentration. Cross-correlation analysis of donor and acceptor fluorescence intensity time traces from single molecules revealed reconfiguration of the polypeptide chain on a timescale of approximately equal to 20 micros at 1.7 M GdmCl. Slow conformational dynamics gave rise to characteristic, stepwise FRET efficiency changes. Transitions between folded and unfolded enzyme molecules occurred on the 100-s timescale, in excellent agreement with bulk denaturation experiments. Transitions between unfolded conformations were more frequent, with characteristic times of approximately equal to 2 s. These data were analyzed to obtain information on the free energy landscape of RNase H in the presence of chemical denaturants.


Asunto(s)
Ribonucleasa H/química , Fluorescencia , Transferencia Resonante de Energía de Fluorescencia , Desnaturalización Proteica , Pliegue de Proteína , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA