Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(1): 317-327, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31852826

RESUMO

Host-pathogen interactions are central to understanding microbial pathogenesis. The staphylococcal pore-forming cytotoxins hijack important immune molecules but little is known about the underlying molecular mechanisms of cytotoxin-receptor interaction and host specificity. Here we report the structures of a staphylococcal pore-forming cytotoxin, leukocidin GH (LukGH), in complex with its receptor (the α-I domain of complement receptor 3, CD11b-I), both for the human and murine homologs. We observe 2 binding interfaces, on the LukG and the LukH protomers, and show that human CD11b-I induces LukGH oligomerization in solution. LukGH binds murine CD11b-I weakly and is inactive toward murine neutrophils. Using a LukGH variant engineered to bind mouse CD11b-I, we demonstrate that cytolytic activity does not only require binding but also receptor-dependent oligomerization. Our studies provide an unprecedented insight into bicomponent leukocidin-host receptor interaction, enabling the development of antitoxin approaches and improved animal models to explore these approaches.


Assuntos
Proteínas de Bactérias/metabolismo , Antígeno CD11b/metabolismo , Leucocidinas/metabolismo , Antígeno de Macrófago 1/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/ultraestrutura , Antígeno CD11b/imunologia , Antígeno CD11b/ultraestrutura , Linhagem Celular , Membrana Celular/metabolismo , Cristalografia por Raios X , Humanos , Leucocidinas/imunologia , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/ultraestrutura , Camundongos , Modelos Moleculares , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Domínios Proteicos/imunologia , Multimerização Proteica/imunologia , Coelhos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Especificidade da Espécie , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/patogenicidade
2.
Genes Immun ; 23(6): 196-204, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36089616

RESUMO

Warts, hypogammaglobulinemia, infections, myelokathexis (WHIM) syndrome is a rare primary immunodeficiency predominantly caused by heterozygous gain-of-function mutations in CXCR4 C-terminus. We assessed genotype-phenotype correlations for known pathogenic CXCR4 variants and in vitro response of each variant to mavorixafor, an investigational CXCR4 antagonist. We used cell-based assays to analyze CXCL12-induced receptor trafficking and downstream signaling of 14 pathogenic CXCR4 variants previously identified in patients with WHIM syndrome. All CXCR4 variants displayed impaired receptor trafficking, hyperactive downstream signaling, and enhanced chemotaxis in response to CXCL12. Mavorixafor inhibited CXCL12-dependent signaling and hyperactivation in cells harboring CXCR4WHIM mutations. A strong correlation was found between CXCR4 internalization defect and severity of blood leukocytopenias and infection susceptibility, and between AKT activation and immunoglobulin A level and CD4+ T-cell counts. This study is the first to show WHIM syndrome clinical phenotype variability as a function of both CXCR4WHIM genotype diversity and associated functional dysregulation. Our findings suggest that CXCR4 internalization may be used to assess the pathogenicity of CXCR4 variants in vitro and also as a potential WHIM-related disease biomarker. The investigational CXCR4 antagonist mavorixafor inhibited CXCL12-dependent signaling in all tested CXCR4-variant cell lines at clinically relevant concentrations.


Assuntos
Agamaglobulinemia , Síndromes de Imunodeficiência , Neutropenia , Verrugas , Agamaglobulinemia/genética , Aminoquinolinas , Benzimidazóis , Biomarcadores , Butilaminas , Estudos de Associação Genética , Humanos , Imunoglobulina A/genética , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/patologia , Neutropenia/genética , Neutropenia/metabolismo , Doenças da Imunodeficiência Primária , Proteínas Proto-Oncogênicas c-akt/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Verrugas/genética , Verrugas/metabolismo , Verrugas/patologia
3.
Biochem J ; 476(2): 275-292, 2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30559327

RESUMO

Host defense against Staphylococcus aureus greatly depends on bacterial clearance by phagocytic cells. LukGH (or LukAB) is the most potent staphylococcal leukocidin towards human phagocytes in vitro, but its role in pathogenesis is obscured by the lack of suitable small animal models because LukGH has limited or no cytotoxicity towards rodent and rabbit compared with human polymorphonuclear cells (PMNs) likely due to an impaired interaction with its cellular receptor, CD11b. We aimed at adapting LukGH for the rabbit host by improving binding to the rabbit homolog of CD11b, specifically its I-domain (CD11b-I). Targeted amino acid substitutions were introduced into the LukH polypeptide to map its receptor interaction site(s). We found that the binding affinity of LukGH variants to the human and rabbit CD11b-I correlated well with their PMN cytotoxicity. Importantly, we identified LukGH variants with significantly improved cytotoxicity towards rabbit PMNs, when expressed recombinantly (10-15-fold) or by engineered S. aureus strains. These findings support the development of small animal models of S. aureus infection with the potential for demonstrating the importance of LukGH in pathogenesis.


Assuntos
Proteínas de Bactérias , Antígeno CD11b/metabolismo , Leucocidinas , Engenharia de Proteínas , Infecções Estafilocócicas , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Leucocidinas/genética , Leucocidinas/metabolismo , Staphylococcus aureus Resistente à Meticilina , Neutrófilos/metabolismo , Neutrófilos/patologia , Coelhos , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia
4.
Adv Exp Med Biol ; 966: 15-35, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28455832

RESUMO

Staphylococcus aureus can produce up to five different bi-component cytotoxins: two gamma-hemolysins HlgAB and HlgCB, and leukocidins SF-PV (Panton Valentine leukocidin), ED (LukED) and GH (LukGH, also called LukAB). Their major function in S. aureus pathogenesis is to evade innate immunity by attacking phagocytic cells and to support bacterial growth by lysing red blood cells. The five cytotoxins display different levels of amino acid sequence conservation (30-82%), but all form a remarkably similar beta-barrel type pore structure (greatly resembling the mono-component toxin alpha-hemolysin) that inserts into the target cell membrane leading to necrotic cell death. This review provides an overview of the culmination of decades of research on the structure of these toxins, their unique sequence and structural features that helps to explain the observed functional differences, such as toxin potency towards different cell types and species, receptor specificity and formation of functional non-cognate toxin pairs. The vast knowledge accumulated in this field supports novel approaches and the design of therapeutics targeting these cytotoxins to tame virulence and fight S. aureus infections.


Assuntos
Antibacterianos/uso terapêutico , Proteínas de Bactérias/antagonistas & inibidores , Desenho de Fármacos , Proteínas Hemolisinas/antagonistas & inibidores , Leucocidinas/antagonistas & inibidores , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Animais , Antibacterianos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Humanos , Leucocidinas/química , Leucocidinas/metabolismo , Modelos Moleculares , Terapia de Alvo Molecular , Conformação Proteica , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade , Relação Estrutura-Atividade , Virulência
5.
Adv Exp Med Biol ; 1053: 119-153, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29549638

RESUMO

The failing efficacy of antibiotics and the high mortality rate among high-risk patients calls for new treatment modalities for bacterial infections. Due to the vastly divergent pathogenesis of human pathogens, each microbe requires a tailored approach. The main modes of action of anti-bacterial antibodies are virulence factor neutralization, complement-mediated bacterial lysis and enhancement of opsonophagocytic uptake and killing (OPK). Gram-positive bacteria cannot be lysed by complement and their pathogenesis often involves secreted toxins, therefore typically toxin-neutralization and OPK activity are required to prevent and ameliorate disease. In fact, the success stories in terms of approved products, in the anti-bacterial mAb field are based on toxin neutralization (Bacillus anthracis, Clostridium difficile). In contrast, Gram-negative bacteria are vulnerable to antibody-dependent complement-mediated lysis, while their pathogenesis rarely relies on secreted exotoxins, and involves the pro-inflammatory endotoxin (lipopolysaccharide). Given the complexity of bacterial pathogenesis, antibody therapeutics are expected to be most efficient upon targeting more than one virulence factor and/or combining different modes of action. The improved understanding of bacterial pathogenesis combined with the versatility and maturity of antibody discovery technologies available today are pivotal for the design of novel anti-bacterial therapeutics. The intensified research generating promising proof-of-concept data, and the increasing number of clinical programs with anti-bacterial mAbs, indicate that the field is ready to fulfill its promise in the coming years.


Assuntos
Antibacterianos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Bactérias/efeitos dos fármacos , Infecções Bacterianas/tratamento farmacológico , Imunoconjugados/uso terapêutico , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Animais , Antibacterianos/efeitos adversos , Antibacterianos/imunologia , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Bactérias/imunologia , Bactérias/patogenicidade , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Citotoxicidade Imunológica , Interações Hospedeiro-Patógeno , Humanos , Imunoconjugados/efeitos adversos , Imunoconjugados/imunologia , Fragmentos Fab das Imunoglobulinas/efeitos adversos , Fragmentos Fab das Imunoglobulinas/imunologia , Virulência
6.
J Biol Chem ; 290(1): 142-56, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25371205

RESUMO

The bi-component leukocidins of Staphylococcus aureus are important virulence factors that lyse human phagocytic cells and contribute to immune evasion. The γ-hemolysins (HlgAB and HlgCB) and Panton-Valentine leukocidin (PVL or LukSF) were shown to assemble from soluble subunits into membrane-bound oligomers on the surface of target cells, creating barrel-like pore structures that lead to cell lysis. LukGH is the most distantly related member of this toxin family, sharing only 30-40% amino acid sequence identity with the others. We observed that, unlike other leukocidin subunits, recombinant LukH and LukG had low solubility and were unable to bind to target cells, unless both components were present. Using biolayer interferometry and intrinsic tryptophan fluorescence we detected binding of LukH to LukG in solution with an affinity in the low nanomolar range and dynamic light scattering measurements confirmed formation of a heterodimer. We elucidated the structure of LukGH by x-ray crystallography at 2.8-Šresolution. This revealed an octameric structure that strongly resembles that reported for HlgAB, but with important structural differences. Structure guided mutagenesis studies demonstrated that three salt bridges, not found in other bi-component leukocidins, are essential for dimer formation in solution and receptor binding. We detected weak binding of LukH, but not LukG, to the cellular receptor CD11b by biolayer interferometry, suggesting that in common with other members of this toxin family, the S-component has the primary contact role with the receptor. These new insights provide the basis for novel strategies to counteract this powerful toxin and Staphylococcus aureus pathogenesis.


Assuntos
Proteínas de Bactérias/química , Antígeno CD11b/química , Proteínas Hemolisinas/química , Leucocidinas/química , Staphylococcus aureus/química , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Antígeno CD11b/metabolismo , Cristalografia por Raios X , Células HL-60 , Proteínas Hemolisinas/metabolismo , Humanos , Leucocidinas/metabolismo , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Staphylococcus aureus/metabolismo , Relação Estrutura-Atividade
7.
Antimicrob Agents Chemother ; 59(6): 3109-16, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25779571

RESUMO

The Escherichia coli sequence type 131 (ST131)-O25b:H4 clone has spread worldwide and become responsible for a significant proportion of multidrug-resistant extraintestinal infections. We generated humanized monoclonal antibodies (MAbs) that target the lipopolysaccharide O25b antigen conserved within this lineage. These MAbs bound to the surface of live bacterial cells irrespective of the capsular type expressed. In a serum bactericidal assay in vitro, MAbs induced >95% bacterial killing in the presence of human serum as the complement source. Protective efficacy at low antibody doses was observed in a murine model of bacteremia. The mode of action in vivo was investigated by using aglycosylated derivatives of the protective MAbs. The significant binding to live E. coli cells and the in vitro and in vivo efficacy were corroborated in assays using bacteria grown in human serum to mimic relevant clinical conditions. Given the dry pipeline of novel antibiotics against multidrug-resistant Gram-negative pathogens, passive immunization with bactericidal antibodies offers a therapeutic alternative to control infections caused by E. coli ST131-O25b:H4.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Antígenos O/metabolismo , Animais , Farmacorresistência Bacteriana Múltipla , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Feminino , Camundongos
8.
Proc Natl Acad Sci U S A ; 108(32): 13007-12, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21778408

RESUMO

Copper is supplied to plastocyanin for photosynthesis and cytochrome c oxidase for respiration in the thylakoids of Synechocystis PCC 6803 by the membrane-bound P-type ATPases CtaA and PacS, and the metallochaperone Atx1. We have determined the Cu(I) affinities of all of the soluble proteins and domains in this pathway. The Cu(I) affinities of the trafficking proteins range from 5 × 10(16) to 5 × 10(17) M(-1) at pH 7.0, consistent with values for homologues. Unusually, Atx1 binds Cu(I) significantly tighter than the metal-binding domains (MBDs) of CtaA and PacS (CtaA(N) and PacS(N)), and equilibrium copper exchange constants of approximately 0.2 are obtained for transfer to the MBDs. Dimerization of Atx1 increases the affinity for Cu(I), but the loop 5 His61 residue has little influence. The MBD of the zinc exporter ZiaA (ZiaA(N)) exhibits an almost identical Cu(I) affinity, and Cu(I) exchange with Atx1, as CtaA(N) and PacS(N), and the relative stabilities of the complexes must enable the metallochaperone to distinguish between the MBDs. The binding of potentially competing zinc to the trafficking proteins has been studied. ZiaA(N) has the highest Zn(II) affinity and thermodynamics could be important for zinc removal from the cell. Plastocyanin has a Cu(I) affinity of 2.6 × 10(17) M(-1), 15-fold tighter than that of the Cu(A) site of cytochrome c oxidase, highlighting the need for specific mechanisms to ensure copper delivery to both of these targets. The narrow range of Cu(I) affinities for the cytoplasmic copper proteins in Synechocystis will facilitate relocation when copper is limiting.


Assuntos
Cobre/metabolismo , Synechocystis/metabolismo , Zinco/metabolismo , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Transporte Biológico , Cinética , Modelos Biológicos , Oxirredução , Plastocianina/metabolismo , Multimerização Proteica , Termodinâmica
9.
Biochemistry ; 52(39): 6816-23, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24050657

RESUMO

Although zinc and copper are required by proteins with very different functions, these metals can be delivered to cellular locations by homologous metal transporters within the same organism, as demonstrated by the cyanobacterial ( Synechocystis PCC 6803) zinc exporter ZiaA and thylakoidal copper importer PacS. The N-terminal metal-binding domains of these transporters (ZiaAN and PacSN, respectively) have related ferredoxin folds also found in the metallochaperone Atx1, which delivers copper to PacS, but differ in the residues found in their M/IXCXXC metal-binding motifs. To investigate the role of the nonconserved residues in this region on metal binding, the sequence from ZiaAN has been introduced into Atx1 and PacSN, and the motifs of Atx1 and PacSN swapped. The motif sequence can tune Cu(I) affinity only approximately 3-fold. However, the introduction of the ZiaAN motif (MDCTSC) dramatically increases the Zn(II) affinity of both Atx1 and PacSN by up to 2 orders of magnitude. The Atx1 mutant with the ZiaAN motif crystallizes as a side-to-side homodimer very similar to that found for [Cu(I)2-Atx1]2 ( Badarau et al. Biochemistry 2010 , 49 , 7798 ). In a crystal structure of the PacSN mutant possessing the ZiaAN motif (PacSN(ZiaAN)), the Asp residue from the metal-binding motif coordinates Zn(II). This demonstrates that the increased Zn(II) affinity of this variant and the high Zn(II) affinity of ZiaAN are due to the ability of the carboxylate to ligate this metal ion. Comparison of the Zn(II) sites in PacSN(ZiaAN) structures provides additional insight into Zn(II) trafficking in cyanobacteria.


Assuntos
Cobre/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Synechocystis/metabolismo , Zinco/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Cobre/química , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Synechocystis/química , Zinco/química
10.
Biochemistry ; 51(7): 1439-48, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22320662

RESUMO

The delivery of copper by the human metallochaperone CCS is a key step in the activation of Cu,Zn-superoxide dismutase (SOD1). CCS is a three-domain protein with Cu(I)-binding CXXC and CXC motifs in domains 1 and 3, respectively. A detailed analysis of the binding of copper to CCS, including variants in which the Cys residues from domains 1 and 3 have been mutated to Ser, and also using separate domain 1 and 3 constructs, demonstrates that CCS is able to bind 1 equiv of Cu(I) in both of these domains. The Cu(I) affinity of domain 1 is approximately 5 × 10(17) M(-1) at pH 7.5, while that of domain 3 is at least 1 order of magnitude weaker. The CXXC site will therefore be preferentially loaded with Cu(I), suggesting that domain 1 plays a role in the acquisition of the metal. The delivery of copper to the target occurs via domain 3 whose structural flexibility and ability to be transiently metalated during copper delivery appear to be more important than the Cu(I) affinity of its CXC motif. The Cu(I) affinity of domain 1 of CCS is comparable to that of HAH1, another cytosolic copper metallochaperone. CCS and HAH1 readily exchange Cu(I), providing a mechanism whereby cross-talk can occur between copper trafficking pathways.


Assuntos
Cobre/química , Escherichia coli/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Cromatografia em Gel , Clonagem Molecular , Cisteína/química , Citosol/química , Relação Dose-Resposta a Droga , Humanos , Concentração de Íons de Hidrogênio , Cinética , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Terciária de Proteína , Serina/química , Espectrofotometria Atômica/métodos , Superóxido Dismutase/química
11.
J Am Chem Soc ; 133(9): 2983-8, 2011 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-21323310

RESUMO

Copper is trafficked to cellular destinations by homeostatic proteins that also prevent adverse reactivity of the metal. The copper metallochaperone HAH1 (human Atx1) binds Cu(I) via a CXXC motif on loop1/α1 of a ßαßßαß ferredoxin-like structure. A similar fold constitutes each of the six metal-binding domains (MBDs) of the two P-type ATPases (Menkes and Wilson disease proteins), the destination for copper bound to HAH1. In this work we have investigated the influence of pH on copper trafficking between HAH1 and the first MBD of the Menkes protein (MNK1). Cu(I) affinities of 5.6 × 10(17) and 3.6 × 10(17) M(-1) have been determined at pH 7.0 for HAH1 and MNK1, respectively, from competition titrations with the chromophoric Cu(I) ligand bathocuproine disulfonate. The mutation of Lys60 on loop5 of HAH1 to Ala (the corresponding residue is Phe67 in MNK1) results in a 3-fold lowering of the affinity for Cu(I) at pH 7.0. The Cu(I) affinity of WT HAH1 exhibits a different pH-dependence compared to MNK1 and Lys60Ala HAH1. This arises because the pK(a) of the second Cys ligand in the CXXC motif of HAH1 is 1.5 pH units lower due to stabilization of the thiolate via a hydrogen-bonding interaction with the side chain of Lys60. The thermodynamic gradient for Cu(I) transfer between HAH1 and MNK1 depends on pH. The decrease in the pK(a) of the Cys ligand in HAH1 can also influence the kinetics of Cu(I) transfer.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Adenosina Trifosfatases/metabolismo , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Transporte de Cobre , ATPases Transportadoras de Cobre , Proteínas Fúngicas/metabolismo , Fungos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Metalochaperonas , Chaperonas Moleculares/metabolismo , Estrutura Terciária de Proteína
12.
Biochemistry ; 49(36): 7798-810, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20726513

RESUMO

Molecular systems have evolved to permit the safe delivery of copper. Despite extensive studies, many copper site structures involved in copper homeostasis, even for the well-studied metallochaperone Atx1, remain unresolved. Cyanobacteria import copper to their thylakoid compartments for use in photosynthesis and respiration and possess an Atx1 that we show can adopt multiple oligomeric states when metalated, capable of binding up to four copper ions. Two-copper- and four-copper-loaded dimers exist in solution at low micromolar concentrations, and head-to-head and side-to-side arrangements, respectively, can be crystallized, with the latter binding a [Cu(4){mu(2)-S(gamma)(Cys)}(4)Cl(2)](2-) cluster. The His61Tyr mutation on loop 5 weakens head-to-head dimerization, yet a side-to-side dimer binding a similar cluster as in the wild-type protein, but with phenolate coordination, is present. The cognate metal-binding domains (MBDs) of the P-type ATPases CtaA and PacS, which are proposed to donate copper to and accept copper from Atx1, respectively, are monomeric in the presence of copper. The structure of the MBD of Cu(I)-PacS shows a crystallographic trimer arrangement around a [Cu(3){mu(2)-S(gamma)(Cys)}(3){S(gamma)(Cys)}(3)](2-) cluster that is very similar to that found for an alternate form of the His61Tyr Atx1 mutant. Copper transfer from the MBD of CtaA to Atx1 is favorable, but delivery from Atx1 to the MBD of PacS is strongly dependent upon the dimeric form of Atx1. A copper-induced switch in Atx1 dimer structure may have a regulatory role with cluster formation helping to buffer copper.


Assuntos
Cobre/química , Metalochaperonas/química , Sítios de Ligação , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Dimerização , Metalochaperonas/metabolismo , Modelos Moleculares , Conformação Proteica
13.
Sci Rep ; 9(1): 5339, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30926865

RESUMO

Staphylococcus aureus pneumonia is associated with high mortality irrespective of antibiotic susceptibility. Both MRSA and MSSA strains produce powerful cytotoxins: alpha-hemolysin(Hla) and up to five leukocidins - LukSF-PV, HlgAB, HlgCB, LukED and LukGH (LukAB) - to evade host innate defense mechanisms. Neutralizing cytotoxins has been shown to provide survival benefit in rabbit S. aureus pneumonia models. We studied the mechanisms of protection of ASN100, a combination of two human monoclonal antibodies (mAbs), ASN-1 and ASN-2, that together neutralize Hla and the five leukocidins, in rabbit MRSA and MSSA pneumonia models. Upon prophylactic passive immunization, ASN100 displayed dose-dependent increase in survival and was fully protective against all S. aureus strains tested at 5 or 20 mg/kg doses. Macroscopic and microscopic lung pathology, edema rate, and bacterial burden were evaluated 12 hours post infection and reduced by ASN100. Pharmacokinetic analysis of ASN100 in bronchoalveolar-lavage fluid from uninfected animals detected efficient penetration to lung epithelial lining fluid reaching peak levels between 24 and 48 hours post dosing that were comparable to the mAb concentration measured in serum. These data confirm that the ASN100 mAbs neutralize the powerful cytotoxins of S. aureus in the lung and prevent damage to the mucosal barrier and innate immune cells.


Assuntos
Anticorpos Neutralizantes/imunologia , Imunoglobulina G/imunologia , Imunotoxinas/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/prevenção & controle , Staphylococcus aureus/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Biópsia , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Imunização Passiva , Imuno-Histoquímica , Imunotoxinas/administração & dosagem , Staphylococcus aureus Resistente à Meticilina/imunologia , Pneumonia Estafilocócica/mortalidade , Pneumonia Estafilocócica/patologia , Prognóstico , Coelhos
15.
Artigo em Inglês | MEDLINE | ID: mdl-18259066

RESUMO

Bacterial resistance to aminoglycoside antibiotics is primarily the result of deactivation of the drugs. Three families of enzymes are responsible for this activity, with one such family being the aminoglycoside phosphotransferases (APHs). The gene encoding one of these enzymes, aminoglycoside-2''-phosphotransferase-Ic [APH(2'')-Ic] from Enterococcus gallinarum, has been cloned and the wild-type protein (comprising 308 amino-acid residues) and three mutants that showed elevated minimum inhibitory concentrations towards gentamicin (F108L, H258L and a double mutant F108L/H258L) were expressed in Escherichia coli and subsequently purified. All APH(2'')-Ic variants were crystallized in the presence of 14-20%(w/v) PEG 4000, 0.25 M MgCl(2), 0.1 M Tris-HCl pH 8.5 and 1 mM Mg(2)GTP. The crystals belong to the monoclinic space group C2, with one molecule in the asymmetric unit. The approximate unit-cell parameters are a = 82.4, b = 54.2, c = 77.0 A, beta = 108.8 degrees. X-ray diffraction data were collected to approximately 2.15 A resolution from an F108L crystal at beamline BL9-2 at SSRL, Stanford, California, USA.


Assuntos
Enterococcus/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/química , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/isolamento & purificação , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação
16.
Biochem J ; 401(1): 197-203, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16961465

RESUMO

Metallo-beta-lactamases are native zinc enzymes that catalyse the hydrolysis of beta-lactam antibiotics, but are also able to function with cobalt(II) and require one or two metal-ions for catalytic activity. The hydrolysis of cefoxitin, cephaloridine and benzylpenicillin catalysed by CoBcII (cobalt-substituted beta-lactamase from Bacillus cereus) has been studied at different pHs and metal-ion concentrations. An enzyme group of pK(a) 6.52+/-0.1 is found to be required in its deprotonated form for metal-ion binding and catalysis. The species that results from the loss of one cobalt ion from the enzyme has no significant catalytic activity and is thought to be the mononuclear CoBcII. It appears that dinuclear CoBcII is the active form of the enzyme necessary for turnover, while the mononuclear CoBcII is only involved in substrate binding. The cobalt-substituted enzyme is a more efficient catalyst than the native enzyme for the hydrolysis of some beta-lactam antibiotics suggesting that the role of the metal-ion is predominantly to provide the nucleophilic hydroxide, rather than to act as a Lewis acid to polarize the carbonyl group and stabilize the oxyanion tetrahedral intermediate.


Assuntos
Bacillus cereus/enzimologia , beta-Lactamases/metabolismo , beta-Lactamas/metabolismo , Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Catálise , Cobalto/metabolismo , Cinética , Metaloproteínas/metabolismo , Modelos Moleculares , Zinco/metabolismo
17.
Virulence ; 9(1): 231-247, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29099326

RESUMO

Pathogenesis of Staphylococcus aureus is increasingly recognized to be driven by powerful toxins. Staphylococcus aureus employs up to six pore-forming toxins to subvert the human host defense and to promote bacterial invasion: alpha-hemolysin that disrupts epithelial and endothelial barriers and five leukocidins that lyse phagocytes involved in bacterial clearance. Previously, we described two human monoclonal antibodies (mAbs), ASN-1 that neutralizes alpha-hemolysin and four leukocidins (LukSF-PV, LukED, HlgAB, HlgCB), and ASN-2 that inactivates the 5th leukocidin, LukGH. In this study we tested the individual and combined effects of ASN-1 and ASN-2 in multiple in vitro models employing relevant human target cells. We found that diverse S. aureus isolates with different genetic backgrounds (based on MLST- and spa-typing) and antibiotic sensitivity (both MRSA and MSSA) displayed greatly different cytotoxin expression patterns influenced by the type of growth medium used. Both mAbs were required to fully prevent the lysis of human neutrophils exposed to the mixture of recombinant cytotoxins or native toxins present in the culture supernatants of S. aureus isolates. Flow cytometry confirmed the protective effects of ASN-1 + ASN-2 (known as ASN100) on granulocytes, monocytes, NK-cells and T-lymphocytes. ASN-1 alone preserved the integrity of a 3D-primary culture of human tracheal/bronchial mucociliary epithelial tissue infected with S. aureus. We conclude that simultaneous inhibition of alpha-hemolysin and five leukocidins by ASN100 blocks cytolytic activity of S. aureus towards human target cells in vitro.


Assuntos
Anticorpos Monoclonais/imunologia , Toxinas Bacterianas/metabolismo , Citotoxinas/metabolismo , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Anticorpos Monoclonais/metabolismo , Toxinas Bacterianas/antagonistas & inibidores , Citotoxinas/antagonistas & inibidores , Proteínas Hemolisinas/antagonistas & inibidores , Proteínas Hemolisinas/metabolismo , Leucocidinas/antagonistas & inibidores , Leucocidinas/metabolismo , Neutrófilos/imunologia , Neutrófilos/microbiologia , Organoides/imunologia , Organoides/microbiologia , Organoides/patologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/química
18.
Virulence ; 9(1): 1521-1538, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30289054

RESUMO

Streptococcus pneumoniae isolates express up to three neuraminidases (sialidases), NanA, NanB and NanC, all of which cleave the terminal sialic acid of glycan-structures that decorate host cell surfaces. Most research has focused on the role of NanA with limited investigations evaluating the roles of all three neuraminidases in host-pathogen interactions. We generated two highly potent monoclonal antibodies (mAbs), one that blocks the enzymatic activity of NanA and one cross-neutralizing NanB and NanC. Total neuraminidase activity of clinical S. pneumoniae isolates could be inhibited by this mAb combination in enzymatic assays. To detect desialylation of cell surfaces by pneumococcal neuraminidases, primary human tracheal/bronchial mucocilial epithelial tissues were infected with S. pneumoniae and stained with peanut lectin. Simultaneous targeting of the neuraminidases was required to prevent desialylation, suggesting that inhibition of NanA alone is not sufficient to preserve terminal lung glycans. Importantly, we also found that all three neuraminidases increased the interaction of S. pneumoniae with human airway epithelial cells. Lectin-staining of lung tissues of mice pre-treated with mAbs before intranasal challenge with S. pneumoniae confirmed that both anti-NanA and anti-NanBC mAbs were required to effectively block desialylation of the respiratory epithelium in vivo. Despite this, no effect on survival, reduction in pulmonary bacterial load, or significant changes in cytokine responses were observed. This suggests that neuraminidases have no pivotal role in this murine pneumonia model that is induced by high bacterial challenge inocula and does not progress from colonization as it happens in the human host.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas de Bactérias/imunologia , Neuraminidase/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/enzimologia , Células A549 , Animais , Anticorpos Antibacterianos/imunologia , Modelos Animais de Doenças , Células Epiteliais/microbiologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Polissacarídeos/metabolismo , Traqueia/citologia , Traqueia/microbiologia
19.
Virulence ; 8(7): 1203-1215, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28103139

RESUMO

Klebsiella pneumoniae ST258 is a globally distributed multi-drug resistant pathogen responsible for severe invasive infections. In this study, the different virulence potential of K. pneumoniae ST258 isolates in endotoxin susceptible versus resistant animal models was shown. Furthermore, ST258 clinical isolates were found highly sensitive to the bactericidal effect of naive animal and human serum. These observations imply that LPS, released from the rapidly lysed bacteria, may contribute to the high mortality associated with ST258 bacteremia cases. A humanized version (mAb A1102) of a previously described murine mAb specific for the conserved LPS O-antigen, was tested for endotoxin neutralization. A1102 was able to neutralize TLR-4 activation by ST258-derived LPS in vitro with an efficacy exceeding that of polymyxin B by 3 orders of magnitude. Passive immunization with A1102 afforded a significant level of protection in a galactosamine-sensitized mouse model of endotoxemia, induced by ST258-derived LPS, or upon challenge with live bacteria. Efficacy was retained using an aglycosylated IgG, as well as upon complement depletion, suggesting that Fc-independent endotoxin neutralization may be the main protective mechanism in this model, in spite of the complement-dependent bactericidal and opsonic activities additionally observed for A1102 in vitro. Furthermore, rabbits that are naturally highly susceptible to endotoxin, were also significantly protected by low doses of A1102 when challenged with an ST258 strain. Given this unique mode of action and the high protective efficacy of this mAb, passive immunization, as prophylactic or adjunct therapeutic approach for the treatment of infections caused by ST258 isolates should be considered.


Assuntos
Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/imunologia , Endotoxinas/imunologia , Infecções por Klebsiella/tratamento farmacológico , Klebsiella pneumoniae/imunologia , Antígenos O/imunologia , Animais , Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Feminino , Humanos , Imunização Passiva , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Coelhos , Ratos , Ratos Sprague-Dawley
20.
J Infect ; 74(5): 473-483, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28237625

RESUMO

OBJECTIVES: Staphylococcus aureus produces up to five bi-component leukocidins - LukSF-PV, gamma-hemolysins AB and CB, LukGH (LukAB) and LukED - to evade innate immunity by lysing phagocytic cells. Species specificity of these leukocidins limits the relevance of animal models, therefore we assessed their individual contribution using human neutrophils. METHODS: Human polymorphonuclear leukocytes (PMNs) were activated with stimuli relevant during bacterial infections and sensitivity to recombinant leukocidins was measured in cell-viability assays. Leukocidin receptor expression was quantified by flow cytometry. RESULTS: We observed greatly variable sensitivities of different PMN preparations towards LukGH. Activation of PMNs by lipopolysaccharide (LPS) or S. aureus culture supernatant (CS) lacking all leukocidins resulted in higher surface expression of CD11b, the LukGH receptor, and greatly enhanced the sensitivity towards LukGH, eliminating the variability observed with unstimulated cells. In contrast, CS induced a decrease in sensitivity of PMNs to the other four leukocidins and reduced surface staining for their cognate receptors (CXCR1, CXCR2, C5aR, C5L2). Delta-toxin and peptidoglycan mimicked the effect of CS. Moreover, IL-8, an important cytokine in neutrophil activation, also selectively increased LukGH sensitivity. Deletion of lukGH, but not other leukocidin genes, prevented PMN killing upon infection with USA300 CA-MRSA. CONCLUSION: Inflammatory signals enhance the susceptibility of human PMNs to lysis by LukGH rendering this toxin dominant among the S. aureus leukocidins in vitro.


Assuntos
Proteínas de Bactérias/imunologia , Leucocidinas/imunologia , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Antígeno CD11b/análise , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Humanos , Interleucina-8/análise , Interleucina-8/imunologia , Interleucina-8/metabolismo , Lipopolissacarídeos/imunologia , Modelos Biológicos , Neutrófilos/microbiologia , Staphylococcus aureus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA