Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Microbiol Biol Educ ; 24(2)2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37614897

RESUMO

Undergraduate microbiology students are exposed to the theory of the scientific method throughout their undergraduate coursework, but laboratory course curricula often focus on technical skills rather than fully integrating scientific thinking as a component of competencies addressed. Here, we have designed a six-session inquiry-based laboratory (IBL) curriculum for an upper-level microbiology laboratory course that fully involves students in the scientific process using bacterial conjugation as the model system, including both online discussions and in-person laboratory sessions. The student learning objectives focus on the scientific method, experimental design, data analysis, bacterial conjugation mechanisms, and scientific communication. We hypothesized students would meet these learning objectives after completing this IBL and tracked student learning and surveyed students to provide an assessment of the structure of the IBL using pre- and post-IBL quizzes and the Laboratory Course Assessment Survey. Overall, our results show this IBL results in positive student learning gains.

2.
mBio ; 10(2)2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015325

RESUMO

The cholesterol-dependent cytolysin (CDC) genes are present in bacterial species that span terrestrial, vertebrate, and invertebrate niches, which suggests that they have evolved to function under widely different environmental conditions. Using a combination of biophysical and crystallographic approaches, we reveal that the relative stability of an intramolecular interface in the archetype CDC perfringolysin O (PFO) plays a central role in regulating its pore-forming properties. The disruption of this interface allows the formation of the membrane spanning ß-barrel pore in all CDCs. We show here that the relative strength of the stabilizing forces at this interface directly impacts the energy barrier posed by the transition state for pore formation, as reflected in the Arrhenius activation energy (Ea) for pore formation. This change directly impacts the kinetics and temperature dependence of pore formation. We further show that the interface structure in a CDC from a terrestrial species enables it to function efficiently across a wide range of temperatures by minimizing changes in the strength of the transition state barrier to pore formation. These studies establish a paradigm that CDCs, and possibly other ß-barrel pore-forming proteins/toxins, can evolve significantly different pore-forming properties by altering the stability of this transitional interface, which impacts the kinetic parameters and temperature dependence of pore formation.IMPORTANCE The cholesterol-dependent cytolysins (CDCs) are the archetype for the superfamily of oligomeric pore-forming proteins that includes the membrane attack complex/perforin (MACPF) family of immune defense proteins and the stonefish venom toxins (SNTX). The CDC/MACPF/SNTX family exhibits a common protein fold, which forms a membrane-spanning ß-barrel pore. We show that changing the relative stability of an extensive intramolecular interface within this fold, which is necessarily disrupted to form the large ß-barrel pore, dramatically alters the kinetic and temperature-dependent properties of CDC pore formation. These studies show that the CDCs and other members of the CDC/MACPF/SNTX superfamily have the capacity to significantly alter their pore-forming properties to function under widely different environmental conditions encountered by these species.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Toxinas Bacterianas/genética , Fenômenos Químicos , Cristalografia por Raios X , Análise Mutacional de DNA , Proteínas Hemolisinas/genética , Cinética , Simulação de Dinâmica Molecular , Proteínas Citotóxicas Formadoras de Poros/genética , Temperatura
3.
Annu Rev Microbiol ; 69: 323-40, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26488276

RESUMO

The mechanism by which the cholesterol-dependent cytolysins (CDCs) assemble their giant ß-barrel pore in cholesterol-rich membranes has been the subject of intense study in the past two decades. A combination of structural, biophysical, and biochemical analyses has revealed deep insights into the series of complex and highly choreographed secondary and tertiary structural transitions that the CDCs undergo to assemble their ß-barrel pore in eukaryotic membranes. Our knowledge of the molecular details of these dramatic structural changes in CDCs has transformed our understanding of how giant pore complexes are assembled and has been critical to our understanding of the mechanisms of other important classes of pore-forming toxins and proteins across the kingdoms of life. Finally, there are tantalizing hints that the CDC pore-forming mechanism is more sophisticated than previously imagined and that some CDCs are employed in pore-independent processes.


Assuntos
Bactérias Gram-Positivas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/química , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Citotoxinas/química , Humanos , Modelos Moleculares , Estrutura Secundária de Proteína
4.
J Biol Chem ; 290(29): 17733-17744, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26032415

RESUMO

The majority of cholesterol-dependent cytolysins (CDCs) utilize cholesterol as a membrane receptor, whereas a small number are restricted to the GPI-anchored protein CD59 for initial membrane recognition. Two cholesterol-binding CDCs, perfringolysin O (PFO) and streptolysin O (SLO), were found to exhibit strikingly different binding properties to cholesterol-rich natural and synthetic membranes. The structural basis for this difference was mapped to one of the loops (L3) in the membrane binding interface that help anchor the toxin monomers to the membrane after receptor (cholesterol) binding by the membrane insertion of its amino acid side chains. A single point mutation in this loop conferred the binding properties of SLO to PFO and vice versa. Our studies strongly suggest that changing the side chain structure of this loop alters its equilibrium between membrane-inserted and uninserted states, thereby affecting the overall binding affinity and total bound toxin. Previous studies have shown that the lipid environment of cholesterol has a dramatic effect on binding and activity. Combining this data with the results of our current studies on L3 suggests that the structure of this loop has evolved in the different CDCs to preferentially direct binding to cholesterol in different lipid environments. Finally, the efficiency of ß-barrel pore formation was inversely correlated with the increased binding and affinity of the PFO L3 mutant, suggesting that selection of a compatible lipid environment impacts the efficiency of membrane insertion of the ß-barrel pore.


Assuntos
Fenômenos Fisiológicos Bacterianos , Toxinas Bacterianas/metabolismo , Membrana Celular/microbiologia , Colesterol/metabolismo , Citotoxinas/metabolismo , Proteínas Hemolisinas/metabolismo , Estreptolisinas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/química , Linhagem Celular , Membrana Celular/metabolismo , Citotoxinas/química , Proteínas Hemolisinas/química , Lipossomos/metabolismo , Camundongos , Modelos Moleculares , Ligação Proteica , Estrutura Secundária de Proteína , Estreptolisinas/química
5.
PLoS Biol ; 13(2): e1002049, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25654333

RESUMO

Membrane attack complex/perforin-like (MACPF) proteins comprise the largest superfamily of pore-forming proteins, playing crucial roles in immunity and pathogenesis. Soluble monomers assemble into large transmembrane pores via conformational transitions that remain to be structurally and mechanistically characterised. Here we present an 11 Å resolution cryo-electron microscopy (cryo-EM) structure of the two-part, fungal toxin Pleurotolysin (Ply), together with crystal structures of both components (the lipid binding PlyA protein and the pore-forming MACPF component PlyB). These data reveal a 13-fold pore 80 Å in diameter and 100 Å in height, with each subunit comprised of a PlyB molecule atop a membrane bound dimer of PlyA. The resolution of the EM map, together with biophysical and computational experiments, allowed confident assignment of subdomains in a MACPF pore assembly. The major conformational changes in PlyB are a ∼70° opening of the bent and distorted central ß-sheet of the MACPF domain, accompanied by extrusion and refolding of two α-helical regions into transmembrane ß-hairpins (TMH1 and TMH2). We determined the structures of three different disulphide bond-trapped prepore intermediates. Analysis of these data by molecular modelling and flexible fitting allows us to generate a potential trajectory of ß-sheet unbending. The results suggest that MACPF conformational change is triggered through disruption of the interface between a conserved helix-turn-helix motif and the top of TMH2. Following their release we propose that the transmembrane regions assemble into ß-hairpins via top down zippering of backbone hydrogen bonds to form the membrane-inserted ß-barrel. The intermediate structures of the MACPF domain during refolding into the ß-barrel pore establish a structural paradigm for the transition from soluble monomer to pore, which may be conserved across the whole superfamily. The TMH2 region is critical for the release of both TMH clusters, suggesting why this region is targeted by endogenous inhibitors of MACPF function.


Assuntos
Membrana Celular/química , Complexo de Ataque à Membrana do Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Pleurotus/química , Proteínas Recombinantes de Fusão/química , Animais , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , Eritrócitos/química , Eritrócitos/citologia , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Expressão Gênica , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Modelos Moleculares , Ligação Proteica , Dobramento de Proteína , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ovinos
6.
Proc Natl Acad Sci U S A ; 112(7): 2204-9, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25646411

RESUMO

ß-Barrel pore-forming toxins (ßPFTs) form an obligatory oligomeric prepore intermediate before the formation of the ß-barrel pore. The molecular components that control the critical prepore-to-pore transition remain unknown for ßPFTs. Using the archetype ßPFT perfringolysin O, we show that E183 of each monomer within the prepore complex forms an intermolecular electrostatic interaction with K336 of the adjacent monomer on completion of the prepore complex. The signal generated throughout the prepore complex by this interaction irrevocably commits it to the formation of the membrane-inserted giant ß-barrel pore. This interaction supplies the free energy to overcome the energy barrier (determined here to be ∼ 19 kcal/mol) to the prepore-to-pore transition by the coordinated disruption of a critical interface within each monomer. These studies provide the first insight to our knowledge into the molecular mechanism that controls the prepore-to-pore transition for a ßPFT.


Assuntos
Colesterol/metabolismo , Eletricidade Estática , Estreptolisinas/metabolismo , Proteínas de Bactérias/metabolismo , Simulação de Dinâmica Molecular , Mutação , Espectrometria de Fluorescência , Temperatura
7.
PLoS Pathog ; 10(9): e1004353, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25188225

RESUMO

Streptococcus pneumoniae produces the pore-forming toxin pneumolysin (PLY), which is a member of the cholesterol-dependent cytolysin (CDC) family of toxins. The CDCs recognize and bind the 3ß-hydroxyl group of cholesterol at the cell surface, which initiates membrane pore formation. The cholesterol transport lipoproteins, which carry cholesterol in their outer monolayer, are potential off-pathway binding targets for the CDCs and are present at significant levels in the serum and the interstitial spaces of cells. Herein we show that cholesterol carried specifically by the ApoB-100-containing lipoprotein particles (CH-ApoB-100) in the mouse, but not that carried by human or guinea pig particles, is a potent inhibitor of the PLY pore-forming mechanism. Cholesterol present in the outer monolayer of mouse ApoB-100 particles is recognized and bound by PLY, which stimulates premature assembly of the PLY oligomeric complex thereby inactivating PLY. These studies further suggest that the vast difference in the inhibitory capacity of mouse CH-ApoB-100 and that of the human and the guinea pig is due to differences in the presentation of cholesterol in the outer monolayer of their ApoB-100 particles. Therefore mouse CH-ApoB-100 represents a significant innate CDC inhibitor that is absent in humans, which may underestimate the contribution of CDCs to human disease when utilizing mouse models of disease.


Assuntos
Apolipoproteína B-100/metabolismo , Colesterol/metabolismo , Hemólise/efeitos dos fármacos , Lipoproteínas/metabolismo , Estreptolisinas/antagonistas & inibidores , Estreptolisinas/farmacologia , Animais , Anticorpos Neutralizantes/sangue , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/farmacologia , Membrana Celular/metabolismo , Cobaias , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
Infect Immun ; 81(1): 216-25, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23115036

RESUMO

The cholesterol-dependent cytolysins (CDCs) are pore-forming toxins that have been exclusively associated with a wide variety of bacterial pathogens and opportunistic pathogens from the Firmicutes and Actinobacteria, which exhibit a Gram-positive type of cell structure. We have characterized the first CDCs from Gram-negative bacterial species, which include Desulfobulbus propionicus type species Widdel 1981 (DSM 2032) (desulfolysin [DLY]) and Enterobacter lignolyticus (formerly Enterobacter cloacae) SCF1 (enterolysin [ELY]). The DLY and ELY primary structures show that they maintain the signature motifs of the CDCs but lack an obvious secretion signal. Recombinant, purified DLY (rDLY) and ELY (rELY) exhibited cholesterol-dependent binding and cytolytic activity and formed the typical large CDC membrane oligomeric pore complex. Unlike the CDCs from Gram-positive species, which are human- and animal-opportunistic pathogens, neither D. propionicus nor E. lignolyticus is known to be a pathogen or commensal of humans or animals: the habitats of both organisms appear to be restricted to anaerobic soils and/or sediments. These studies reveal for the first time that the genes for functional CDCs are present in bacterial species that exhibit a Gram-negative cell structure. These are also the first bacterial species containing a CDC gene that are not known to inhabit or cause disease in humans or animals, which suggests a role of these CDCs in the defense against eukaryote bacterial predators.


Assuntos
Colesterol/metabolismo , Citotoxinas/genética , Citotoxinas/metabolismo , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Ligação Proteica/genética , Proteobactérias/genética , Proteobactérias/metabolismo , RNA Mensageiro/genética
9.
J Biol Chem ; 287(29): 24534-43, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22645132

RESUMO

The assembly of the cholesterol-dependent cytolysin (CDC) oligomeric pore complex requires a complex choreography of secondary and tertiary structural changes in domain 3 (D3) of the CDC monomer structure. A point mutation was identified in the archetype CDC, perfringolysin O, that blocks detectable D3 structural changes and traps the membrane-bound monomers in an early and reversible stage of oligomer assembly. Using this and other mutants we show that specific D3 structural changes are propagated from one membrane-bound monomer to another. Propagation of these structural changes results in the exposure of a ß-strand in D3 that allows it to pair and form edge-on interactions with a second ß-strand of a free membrane-bound monomer. Pairing of these strands establishes the final geometry of the pore complex and is necessary to drive the formation of the ß-barrel pore. These studies provide new insights into how structural information is propagated between membrane-bound monomers of a self-assembling system and the interactions that establish the geometry of the final pore complex.


Assuntos
Colesterol/metabolismo , Perforina/química , Perforina/metabolismo , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Transferência Ressonante de Energia de Fluorescência , Hemólise , Humanos , Microscopia Eletrônica , Perforina/genética , Mutação Puntual/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
10.
Structure ; 20(2): 248-58, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22325774

RESUMO

The cholesterol-dependent cytolysins (CDCs) punch holes in target cell membranes through a highly regulated process. Streptococcus mitis lectinolysin (LLY) exhibits another layer of regulation with a lectin domain that enhances the pore-forming activity of the toxin. We have determined the crystal structures of the lectin domain by itself and in complex with various glycans that reveal the molecular basis for the Lewis antigen specificity of LLY. A small-angle X-ray scattering study of intact LLY reveals the molecule is flat and elongated with the lectin domain oriented so that the Lewis antigen-binding site is exposed. We suggest that the lectin domain enhances the pore-forming activity of LLY by concentrating toxin molecules at fucose-rich sites on membranes, thus promoting the formation of prepore oligomers on the surface of susceptible cells.


Assuntos
Proteínas de Bactérias/química , Lectinas/química , Antígenos do Grupo Sanguíneo de Lewis/química , Proteínas Citotóxicas Formadoras de Poros/química , Streptococcus mitis , Sítios de Ligação , Cristalografia por Raios X , Fucose/química , Ligação de Hidrogênio , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Espalhamento a Baixo Ângulo , Difração de Raios X
11.
Biochim Biophys Acta ; 1818(4): 1028-38, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21835159

RESUMO

The cholesterol-dependent cytolysins (CDCs) are a large family of pore-forming toxins that are produced, secreted and contribute to the pathogenesis of many species of Gram-positive bacteria. The assembly of the CDC pore-forming complex has been under intense study for the past 20 years. These studies have revealed a molecular mechanism of pore formation that exhibits many novel features. The CDCs form large ß-barrel pore complexes that are assembled from 35 to 40 soluble CDC monomers. Pore formation is dependent on the presence of membrane cholesterol, which functions as the receptor for most CDCs. Cholesterol binding initiates significant secondary and tertiary structural changes in the monomers, which lead to the assembly of a large membrane embedded ß-barrel pore complex. This review will focus on the molecular mechanism of assembly of the CDC membrane pore complex and how these studies have led to insights into the mechanism of pore formation for other pore-forming proteins. This article is part of a Special Issue entitled: Protein Folding in Membranes.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Perforina/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Humanos , Perforina/química , Estrutura Secundária de Proteína
12.
J Biol Chem ; 286(23): 20952-62, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21507937

RESUMO

CD59 is a glycosylphosphatidylinositol-anchored protein that inhibits the assembly of the terminal complement membrane attack complex (MAC) pore, whereas Streptococcus intermedius intermedilysin (ILY), a pore forming cholesterol-dependent cytolysin (CDC), specifically binds to human CD59 (hCD59) to initiate the formation of its pore. The identification of the residues of ILY and hCD59 that form their binding interface revealed a remarkably deep correspondence between the hCD59 binding site for ILY and that for the MAC proteins C8α and C9. ILY disengages from hCD59 during the prepore to pore transition, suggesting that loss of this interaction is necessary to accommodate specific structural changes associated with this transition. Consistent with this scenario, mutants of hCD59 or ILY that increased the affinity of this interaction decreased the cytolytic activity by slowing the transition of the prepore to pore but not the assembly of the prepore oligomer. A signature motif was also identified in the hCD59 binding CDCs that revealed a new hCD59-binding member of the CDC family. Although the binding site on hCD59 for ILY, C8α, and C9 exhibits significant homology, no similarity exists in their binding sites for hCD59. Hence, ILY and the MAC proteins interact with common amino acids of hCD59 but lack detectable conservation in their binding sites for hCD59.


Assuntos
Bacteriocinas/metabolismo , Antígenos CD59/metabolismo , Complemento C8/metabolismo , Motivos de Aminoácidos , Animais , Bacteriocinas/química , Bacteriocinas/genética , Sítios de Ligação , Antígenos CD59/química , Antígenos CD59/genética , Células CHO , Complemento C8/química , Complemento C8/genética , Complemento C9/química , Complemento C9/genética , Complemento C9/metabolismo , Cricetinae , Cricetulus , Humanos , Mutação , Mapeamento de Peptídeos/métodos , Streptococcus intermedius/química , Streptococcus intermedius/genética , Streptococcus intermedius/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(9): 4341-6, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20145114

RESUMO

The recognition and binding of cholesterol is an important feature of many eukaryotic, viral, and prokaryotic proteins, but the molecular details of such interactions are understood only for a few proteins. The pore-forming cholesterol-dependent cytolysins (CDCs) contribute to the pathogenic mechanisms of a large number of Gram-positive bacteria. Cholesterol dependence of the CDC mechanism is a hallmark of these toxins, yet the identity of the CDC cholesterol recognition motif has remained elusive. A detailed analysis of membrane interactive structures at the tip of perfringolysin O (PFO) domain 4 reveals that a threonine-leucine pair mediates CDC recognition of and binding to membrane cholesterol. This motif is conserved in all known CDCs and conservative changes in its sequence or order are not well tolerated. Thus, the Thr-Leu pair constitutes a common structural basis for mediating CDC-cholesterol recognition and binding, and defines a unique paradigm for membrane cholesterol recognition by surface-binding proteins.


Assuntos
Toxinas Bacterianas/metabolismo , Colesterol/metabolismo , Proteínas Hemolisinas/metabolismo , Leucina/metabolismo , Lipídeos de Membrana/metabolismo , Treonina/metabolismo , Sítios de Ligação , Western Blotting , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Bactérias Gram-Positivas/patogenicidade , Hemólise , Humanos , Ressonância de Plasmônio de Superfície
14.
J Biol Chem ; 284(19): 12719-26, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19293153

RESUMO

Intermedilysin (ILY) is an unusual member of the family of cholesterol-dependent cytolysins because it binds to human CD59 (hCD59) rather than directly to cholesterol-rich membranes. Binding of ILY to hCD59 initiates a series of conformational changes within the toxin that result in the conversion of the soluble monomer into an oligomeric membrane-embedded pore complex. In this study the association of ILY with its membrane receptor has been examined throughout the assembly and formation of the pore complex. Using ILY mutants trapped at various stages of pore assembly, we show ILY remains engaged with hCD59 throughout the assembly of the prepore oligomer, but it disengages from the receptor upon the conversion to the pore complex. We further show that the assembly intermediates increase the sensitivity of the host cell to lysis by its complement membrane attack complex, apparently by blocking the hCD59-binding site for complement proteins C8alpha and C9.


Assuntos
Bacteriocinas/metabolismo , Antígenos CD59/metabolismo , Complemento C8/metabolismo , Complemento C9/metabolismo , Citotoxinas/metabolismo , Membrana Eritrocítica/química , Animais , Bacteriocinas/imunologia , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Dimerização , Hemólise , Humanos , Imunoprecipitação , Ligação Proteica , Coelhos
15.
Proc Natl Acad Sci U S A ; 104(51): 20226-31, 2007 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18077338

RESUMO

The pore-forming mechanism of the cholesterol-dependent cytolysins (CDCs) exhibits an absolute requirement for membrane cholesterol. The structural elements of the CDCs that mediate this interaction are not well understood. Three short hydrophobic loops (L1-L3) and a highly conserved undecapeptide sequence at the tip of domain 4 of the CDC structure are known to anchor the CDC to the membrane. It has been thought that the undecapeptide directly mediates the interaction of the CDCs with a cholesterol-rich cell surface. Herein we show that the L1-L3 loops, not the undecapeptide, are responsible for mediating the specific interaction of the CDCs with cholesterol-rich membranes. The membrane insertion of the undecapeptide was uncoupled from membrane binding by the covalent modification of the undecapeptide cysteine thiol. Modification of the cysteine prevented prepore to pore conversion, but did not affect membrane binding, thus demonstrating that undecapeptide membrane insertion follows that of the L1-L3 loops. These studies provide an example of a structural motif that specifically mediates the interaction of a bacterial toxin with a cholesterol-rich membrane.


Assuntos
Proteínas de Bactérias/química , Toxinas Bacterianas/química , Bacteriocinas/química , Colesterol/química , Citotoxinas/química , Proteínas Hemolisinas/química , Membranas Artificiais , Substituição de Aminoácidos , Ácido Aspártico/química , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Bacteriocinas/genética , Cristalografia por Raios X , Cisteína/química , Citotoxinas/genética , Glicina/química , Proteínas Hemolisinas/genética , Lipossomos/química , Porosidade , Conformação Proteica , Triptofano/química
16.
J Biol Chem ; 282(21): 15709-16, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17412689

RESUMO

Three short hydrophobic loops and a conserved undecapeptide at the tip of domain 4 (D4) of the cholesterol-dependent cytolysins (CDCs) mediate the binding of the CDC monomers to cholesterol-rich cell membranes. But intermedilysin (ILY), from Streptococcus intermedius, does not bind to cholesterol-rich membranes unless they contain the human protein CD59. This observation suggested that the D4 loops, which include loops L1-L3 and the undecapeptide, of ILY were no longer required for its cell binding. However, we show here that membrane insertion of the D4 loops is required for the cytolysis by ILY. Receptor binding triggers changes in the structure of ILY that are necessary for oligomerization, but membrane insertion of the D4 loops is critical for oligomer assembly and pore formation. Defects that prevent membrane insertion of the undecapeptide also block assembly of the prepore oligomer, while defects in the membrane insertion of the L1-L3 loops prevent the conversion of the prepore oligomer to the pore complex. These studies reveal that pore formation by ILY, and probably other CDCs, is affected by an intricate and coupled sequence of interactions between domain 4 and the membrane.


Assuntos
Proteínas de Bactérias/química , Bacteriocinas/química , Antígenos CD59/química , Colesterol/química , Membrana Eritrocítica/química , Streptococcus intermedius/química , Proteínas de Bactérias/metabolismo , Bacteriocinas/metabolismo , Antígenos CD59/metabolismo , Colesterol/metabolismo , Membrana Eritrocítica/metabolismo , Humanos , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Streptococcus intermedius/metabolismo , Relação Estrutura-Atividade
17.
J Struct Biol ; 150(1): 100-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15797734

RESUMO

Perfringolysin O (PFO), a soluble toxin secreted by the pathogenic Clostridium perfringens, forms large homo-oligomeric pore complexes comprising up to 50 PFO molecules in cholesterol-containing membranes. In this study, electron microscopy (EM) and single-particle image analysis were used to reconstruct two-dimensional (2D) projection maps from images of oligomeric PFO prepore and pore complexes formed on cholesterol-rich lipid layers. The projection maps are characterized by an outer and an inner ring of density peaks. The outer rings of the prepore and pore complexes are very similar; however, the protein densities that make up the inner ring of the pore complex are more intense and discretely resolved than they are for the prepore complex. The change in inner-ring protein density is consistent with a mechanism in which the monomers within the prepore complex make a transition from a partially disordered state to a more ordered transmembrane beta-barrel in the pore complex. Finally, the orientation of the monomers within the oligomeric complexes was determined by visualization of streptavidin (SA) molecules bound to biotinylated cysteine-substituted residues predicted to face either the inner or outer surface of the oligomeric pore complex. This study provides an unprecedented view of the conversion of the PFO prepore to pore complex.


Assuntos
Toxinas Bacterianas/química , Colesterol/química , Clostridium perfringens/metabolismo , Proteínas Hemolisinas , Microscopia Eletrônica , Estrutura Molecular
18.
EMBO J ; 23(16): 3206-15, 2004 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-15297878

RESUMO

Perfringolysin O (PFO) is a prototype of the large family of pore-forming cholesterol-dependent cytolysins (CDCs). A central enigma of the cytolytic mechanism of the CDCs is that their membrane-spanning beta-hairpins (the transmembrane amphipathic beta-hairpins (TMHs)) appear to be approximately 40 A too far above the membrane surface to cross the bilayer and form the pore. We now present evidence, using atomic force microscopy (AFM), of a significant difference in the height by which the prepore and pore protrude from the membrane surface: 113+/-5 A for the prepore but only 73+/-5 A for the pore. Time-lapse AFM micrographs show this change in height in real time. Moreover, the monomers in both complexes exhibit nearly identical surface features and these results in combination with those of spectrofluorimetric analyses indicate that the monomers remain in a perpendicular orientation to the bilayer plane during this transition. Therefore, the PFO undergoes a vertical collapse that brings its TMHs to the membrane surface so that they can extend across the bilayer to form the beta-barrel pore.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Clostridium perfringens/química , Citotoxinas/química , Citotoxinas/metabolismo , Membrana Celular/ultraestrutura , Clostridium perfringens/metabolismo , Proteínas Hemolisinas , Lipossomos/química , Lipossomos/metabolismo , Microscopia de Força Atômica , Modelos Moleculares , Estrutura Terciária de Proteína , Transporte Proteico , Fatores de Tempo
19.
J Biol Chem ; 277(13): 11597-605, 2002 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-11799121

RESUMO

Perfringolysin O (PFO), a cholesterol-dependent cytolysin, forms large oligomeric pore complexes comprised of up to 50 PFO molecules. In the present studies a mutant of PFO (PFO(Y181A)) has been identified that traps PFO in a multimeric prepore complex that cannot insert its transmembrane beta-hairpins and therefore cannot form a pore. Remarkably, PFO(Y181A) can be induced to insert its transmembrane beta-hairpins if functional PFO is incorporated into the PFO(Y181A) oligomeric prepore complex. Furthermore, the transition from prepore to pore appears to be an "all or none" process; partial insertion of the transmembrane beta-barrel does not occur. Therefore, cooperative interactions between the monomers of the prepore drive the prepore to pore conversion that results in the formation of the transmembrane beta-barrel.


Assuntos
Toxinas Bacterianas/metabolismo , Colesterol/metabolismo , Transferência de Energia , Glutationa/metabolismo , Proteínas Hemolisinas , Espectrometria de Fluorescência , beta-Amilase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...