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.
Nat Commun ; 15(1): 899, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38321013

RESUMO

Antigen-specific regulatory T cells (Tregs) suppress pathogenic autoreactivity and are potential therapeutic candidates for autoimmune diseases such as systemic lupus erythematosus (SLE). Lupus nephritis is associated with autoreactivity to the Smith (Sm) autoantigen and the human leucocyte antigen (HLA)-DR15 haplotype; hence, we investigated the potential of Sm-specific Tregs (Sm-Tregs) to suppress disease. Here we identify a HLA-DR15 restricted immunodominant Sm T cell epitope using biophysical affinity binding assays, then identify high-affinity Sm-specific T cell receptors (TCRs) using high-throughput single-cell sequencing. Using lentiviral vectors, we transduce our lead Sm-specific TCR into Tregs derived from patients with SLE who are anti-Sm and HLA-DR15 positive. Compared with polyclonal mock-transduced Tregs, Sm-Tregs potently suppress Sm-specific pro-inflammatory responses in vitro and suppress disease progression in a humanized mouse model of lupus nephritis. These results show that Sm-Tregs are a promising therapy for SLE.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Camundongos , Animais , Humanos , Linfócitos T Reguladores , Autoantígenos/metabolismo
2.
Methods Mol Biol ; 2691: 351-369, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37355557

RESUMO

Epitope-specific immunotherapies have enabled the targeted treatment of a variety of diseases, ranging from cancer, infection, and autoimmune disorders. For CD8+ T cell-based therapies, the precise identification of immunogenic peptides presented by human leukocyte antigen (HLA) class I is essential which can be achieved by immunopeptidomics. Here, using lentivirus-mediated transduction and cell sorting approaches, we present a method to engineer a cell line that does not express its native HLA but instead expresses an HLA of interest (in this instance HLA-A*02:01). This technique can be used to elucidate the immunopeptidome of cell lines expressing different HLAs.


Assuntos
Antígenos de Neoplasias , Antígenos de Histocompatibilidade Classe I , Humanos , Antígenos de Histocompatibilidade Classe I/genética , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe II , Linhagem Celular Tumoral , Epitopos de Linfócito T , Apresentação de Antígeno
3.
Front Immunol ; 13: 821595, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154139

RESUMO

Heterologous immunity, when the memory T cell response elicited by one pathogen recognizes another pathogen, has been offered as a contributing factor for the high variability in coronavirus disease 2019 (COVID-19) severity outcomes. Here we demonstrate that sensitization with bacterial peptides can induce heterologous immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) derived peptides and that vaccination with the SARS-CoV-2 spike protein can induce heterologous immunity to bacterial peptides. Using in silico prediction methods, we identified 6 bacterial peptides with sequence homology to either the spike protein or non-structural protein 3 (NSP3) of SARS-CoV-2. Notwithstanding the effects of bystander activation, in vitro co-cultures showed that all individuals tested (n=18) developed heterologous immunity to SARS-CoV-2 peptides when sensitized with the identified bacterial peptides. T cell recall responses measured included cytokine production (IFN-γ, TNF, IL-2), activation (CD69) and proliferation (CellTrace). As an extension of the principle of heterologous immunity between bacterial pathogens and COVID-19, we tracked donor responses before and after SARS-CoV-2 vaccination and measured the cross-reactive T cell responses to bacterial peptides with similar sequence homology to the spike protein. We found that SARS-CoV-2 vaccination could induce heterologous immunity to bacterial peptides. These findings provide a mechanism for heterologous T cell immunity between common bacterial pathogens and SARS-CoV-2, which may explain the high variance in COVID-19 outcomes from asymptomatic to severe. We also demonstrate proof-of-concept that SARS-CoV-2 vaccination can induce heterologous immunity to pathogenic bacteria derived peptides.


Assuntos
Infecções Bacterianas/imunologia , COVID-19/imunologia , Imunidade Heteróloga/imunologia , SARS-CoV-2/imunologia , Linfócitos T/imunologia , Adulto , Vacinas contra COVID-19/imunologia , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Imunidade Celular/imunologia , Masculino , Glicoproteína da Espícula de Coronavírus/imunologia
4.
Virulence ; 13(1): 225-240, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35094646

RESUMO

Streptococcus pyogenes, a leading human pathogen, is responsible for a wide range of diseases, including skin and soft tissue infections and severe invasive diseases. S. pyogenes produces a large arsenal of virulence factors, including several immune evasion factors. We have identified an open reading frame (spy0136) in the S. pyogenes SF370 genome encoding a protein of unknown function. Using recombinant Spy0136 in a pull-down assay with human plasma and ELISA, we have identified four complement proteins (C1r, C1s, C3, and C5) as binding partners. Treatment of the complement proteins with PNGase F abrogated binding to C1s, C3, and C5, indicating glycan-dependent interactions. rSpy0136 inhibited complement-mediated hemolysis and interfered with all three complement pathways in a Wieslab complement assay. Furthermore, rSpy0136 inhibited deposition of the C3b opsonin and the membrane attack complex (MAC) on the surface of S. pyogenes. We therefore named the previously unknown protein 'complement evasion factor' (CEF).An S. pyogenes Δspy0136/cef deletion mutant showed decreased virulence in an in-vitro whole blood killing assay and a Galleria mellonella (wax moth) infection model. Furthermore, an L. lactis spy0136/cef gain-of-function mutant showed increased survival during growth in whole human blood. Analysis of serum samples from patients with invasive S. pyogenes revealed Spy0136/CEF sero-conversion indicating expression during disease. In summary, we have identified a novel S. pyogenes immune evasion factor that binds to several complement proteins to interfere with complement function. This is the first example of a S. pyogenes virulence factor binding to several different target proteins via glycan-dependent interactions.


Assuntos
Infecções Estreptocócicas , Streptococcus pyogenes , Proteínas de Bactérias/metabolismo , Proteínas do Sistema Complemento , Humanos , Evasão da Resposta Imune , Streptococcus pyogenes/genética , Fatores de Virulência/metabolismo
5.
Nat Commun ; 12(1): 5110, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433824

RESUMO

HLA-DQ8, a genetic risk factor in type I diabetes (T1D), presents hybrid insulin peptides (HIPs) to autoreactive CD4+ T cells. The abundance of spliced peptides binding to HLA-DQ8 and how they are subsequently recognised by the autoreactive T cell repertoire is unknown. Here we report, the HIP (GQVELGGGNAVEVLK), derived from splicing of insulin and islet amyloid polypeptides, generates a preferred peptide-binding motif for HLA-DQ8. HLA-DQ8-HIP tetramer+ T cells from the peripheral blood of a T1D patient are characterised by repeated TRBV5 usage, which matches the TCR bias of CD4+ T cells reactive to the HIP peptide isolated from the pancreatic islets of a patient with T1D. The crystal structure of three TRBV5+ TCR-HLA-DQ8-HIP complexes shows that the TRBV5-encoded TCR ß-chain forms a common landing pad on the HLA-DQ8 molecule. The N- and C-termini of the HIP is recognised predominantly by the TCR α-chain and TCR ß-chain, respectively, in all three TCR ternary complexes. Accordingly, TRBV5 + TCR recognition of HIP peptides might occur via a 'polarised' mechanism, whereby each chain within the αßTCR heterodimer recognises distinct origins of the spliced peptide presented by HLA-DQ8.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Antígenos HLA-DQ/metabolismo , Insulina/metabolismo , Peptídeos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Sequência de Aminoácidos , Linfócitos T CD4-Positivos/química , Linfócitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Antígenos HLA-DQ/química , Antígenos HLA-DQ/genética , Humanos , Insulina/química , Insulina/genética , Peptídeos/química , Ligação Proteica , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética
6.
Sci Immunol ; 6(58)2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863750

RESUMO

Individuals expressing HLA-DR4 bearing the shared susceptibility epitope (SE) have an increased risk of developing rheumatoid arthritis (RA). Posttranslational modification of self-proteins via citrullination leads to the formation of neoantigens that can be presented by HLA-DR4 SE allomorphs. However, in T cell-mediated autoimmunity, the interplay between the HLA molecule, posttranslationally modified epitope(s), and the responding T cell repertoire remains unclear. In HLA-DR4 transgenic mice, we show that immunization with a Fibß-74cit69-81 peptide led to a population of HLA-DR4Fibß-74cit69-81 tetramer+ T cells that exhibited biased T cell receptor (TCR) ß chain usage, which was attributable to selective clonal expansion from the preimmune repertoire. Crystal structures of pre- and postimmune TCRs showed that the SE of HLA-DR4 represented a main TCR contact zone. Immunization with a double citrullinated epitope (Fibß-72,74cit69-81) altered the responding HLA-DR4 tetramer+ T cell repertoire, which was due to the P2-citrulline residue interacting with the TCR itself. We show that the SE of HLA-DR4 has dual functionality, namely, presentation and a direct TCR recognition determinant. Analogous biased TCR ß chain usage toward the Fibß-74cit69-81 peptide was observed in healthy HLA-DR4+ individuals and patients with HLA-DR4+ RA, thereby suggesting a link to human RA.


Assuntos
Artrite Reumatoide/imunologia , Epitopos de Linfócito T/metabolismo , Antígeno HLA-DR4/metabolismo , Linfócitos T/imunologia , Adulto , Idoso de 80 Anos ou mais , Alelos , Animais , Artrite Reumatoide/sangue , Autoantígenos/imunologia , Autoantígenos/metabolismo , Citrulinação/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Antígeno HLA-DR4/genética , Antígeno HLA-DR4/imunologia , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Cadeias HLA-DRB1/metabolismo , Humanos , Masculino , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(6): 3063-3073, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31974305

RESUMO

The highly homologous human leukocyte antigen (HLA)-DQ2 molecules, HLA-DQ2.5 and HLA-DQ2.2, are implicated in the pathogenesis of celiac disease (CeD) by presenting gluten peptides to CD4+ T cells. However, while HLA-DQ2.5 is strongly associated with disease, HLA-DQ2.2 is not, and the molecular basis underpinning this differential disease association is unresolved. We here provide structural evidence for how the single polymorphic residue (HLA-DQ2.5-Tyr22α and HLA-DQ2.2-Phe22α) accounts for HLA-DQ2.2 additionally requiring gluten epitopes possessing a serine at the P3 position of the peptide. In marked contrast to the biased T cell receptor (TCR) usage associated with HLA-DQ2.5-mediated CeD, we demonstrate with extensive single-cell sequencing that a diverse TCR repertoire enables recognition of the immunodominant HLA-DQ2.2-glut-L1 epitope. The crystal structure of two CeD patient-derived TCR in complex with HLA-DQ2.2 and DQ2.2-glut-L1 (PFSEQEQPV) revealed a docking strategy, and associated interatomic contacts, which was notably distinct from the structures of the TCR:HLA-DQ2.5:gliadin epitope complexes. Accordingly, while the molecular surfaces of the antigen-binding clefts of HLA-DQ2.5 and HLA-DQ2.2 are very similar, differences in the nature of the peptides presented translates to differences in responding T cell repertoires and the nature of engagement of the respective antigen-presenting molecules, which ultimately is associated with differing disease penetrance.


Assuntos
Doença Celíaca , Antígenos HLA-DQ , Receptores de Antígenos de Linfócitos T , Linfócitos T CD4-Positivos/química , Linfócitos T CD4-Positivos/metabolismo , Doença Celíaca/genética , Doença Celíaca/imunologia , Doença Celíaca/metabolismo , Linhagem Celular , Cristalografia por Raios X , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/metabolismo , Glutens/química , Glutens/imunologia , Glutens/metabolismo , Antígenos HLA-DQ/química , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo
8.
JCI Insight ; 4(18)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31487265

RESUMO

Autoimmune diseases resulting from MHC class II-restricted autoantigen-specific T cell immunity include the systemic inflammatory autoimmune conditions rheumatoid arthritis and vasculitis. While currently treated with broad-acting immunosuppressive drugs, a preferable strategy is to regulate antigen-specific effector T cells (Teffs) to restore tolerance by exploiting DC antigen presentation. We targeted draining lymph node (dLN) phagocytic DCs using liposomes encapsulating 1α,25-dihydroxyvitamin D3 (calcitriol) and antigenic peptide to elucidate mechanisms of tolerance used by DCs and responding T cells under resting and immunized conditions. PD-L1 expression was upregulated in dLNs of immunized relative to naive mice. Subcutaneous administration of liposomes encapsulating OVA323-339 and calcitriol targeted dLN PD-L1hi DCs of immunized mice and reduced their MHC class II expression. OVA323-339/calcitriol liposomes suppressed expansion, differentiation, and function of Teffs and induced Foxp3+ and IL-10+ peripheral Tregs in an antigen-specific manner, which was dependent on PD-L1. Peptide/calcitriol liposomes modulated CD40 expression by human DCs and promoted Treg induction in vitro. Liposomes encapsulating calcitriol and disease-associated peptides suppressed the severity of rheumatoid arthritis and Goodpasture's vasculitis models with suppression of antigen-specific memory T cell differentiation and function. Accordingly, peptide/calcitriol liposomes leverage DC PD-L1 for antigen-specific T cell regulation and induce antigen-specific tolerance in inflammatory autoimmune diseases.


Assuntos
Doença Antimembrana Basal Glomerular/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Calcitriol/administração & dosagem , Células Dendríticas/imunologia , Epitopos Imunodominantes/administração & dosagem , Transferência Adotiva , Animais , Doença Antimembrana Basal Glomerular/diagnóstico , Doença Antimembrana Basal Glomerular/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/imunologia , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Células CHO , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Cricetulus , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/transplante , Modelos Animais de Doenças , Feminino , Antígenos HLA-DR/genética , Antígenos HLA-DR/imunologia , Antígenos HLA-DR/metabolismo , Humanos , Tolerância Imunológica/efeitos dos fármacos , Epitopos Imunodominantes/imunologia , Memória Imunológica/efeitos dos fármacos , Injeções Subcutâneas , Lipossomos , Linfonodos/citologia , Camundongos , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Índice de Gravidade de Doença , Linfócitos T/imunologia , Linfócitos T/metabolismo
10.
J Biol Chem ; 293(9): 3236-3251, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29317506

RESUMO

The HLA-DRB1 locus is strongly associated with rheumatoid arthritis (RA) susceptibility, whereupon citrullinated self-peptides bind to HLA-DR molecules bearing the shared epitope (SE) amino acid motif. However, the differing propensity for citrullinated/non-citrullinated self-peptides to bind given HLA-DR allomorphs remains unclear. Here, we used a fluorescence polarization assay to determine a hierarchy of binding affinities of 34 self-peptides implicated in RA against three HLA-DRB1 allomorphs (HLA-DRB1*04:01/*04:04/*04:05) each possessing the SE motif. For all three HLA-DRB1 allomorphs, we observed a strong correlation between binding affinity and citrullination at P4 of the bound peptide ligand. A differing hierarchy of peptide-binding affinities across the three HLA-DRB1 allomorphs was attributable to the ß-chain polymorphisms that resided outside the SE motif and were consistent with sequences of naturally presented peptide ligands. Structural determination of eight HLA-DR4-self-epitope complexes revealed strict conformational convergence of the P4-Cit and surrounding HLA ß-chain residues. Polymorphic residues that form part of the P1 and P9 pockets of the HLA-DR molecules provided a structural basis for the preferential binding of the citrullinated self-peptides to the HLA-DR4 allomorphs. Collectively, we provide a molecular basis for the interplay between citrullination of self-antigens and HLA polymorphisms that shape peptide-HLA-DR4 binding affinities in RA.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Citrulinação , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/metabolismo , Peptídeos/metabolismo , Polimorfismo Genético , Sequência de Aminoácidos , Artrite Reumatoide/imunologia , Autoantígenos/química , Autoantígenos/metabolismo , Citrulina/metabolismo , Cadeias HLA-DRB1/química , Humanos , Modelos Moleculares , Peptídeos/química , Ligação Proteica , Conformação Proteica em Folha beta , Especificidade por Substrato
11.
PLoS Pathog ; 13(9): e1006549, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28880913

RESUMO

Staphylococcus aureus is an opportunistic pathogen that produces many virulence factors. Two major families of which are the staphylococcal superantigens (SAgs) and the Staphylococcal Superantigen-Like (SSL) exoproteins. The former are immunomodulatory toxins that induce a Vß-specific activation of T cells, while the latter are immune evasion molecules that interfere with a wide range of innate immune defences. The superantigenic properties of Staphylococcal enterotoxin-like X (SElX) have recently been established. We now reveal that SElX also possesses functional characteristics of the SSLs. A region of SElX displays high homology to the sialyl-lactosamine (sLacNac)-specific binding site present in a sub-family of SSLs. By analysing the interaction of SElX with sLacNac-containing glycans we show that SElX has an equivalent specificity and host cell binding range to the SSLs. Mutation of key amino acids in this conserved region affects the ability of SElX to bind to cells of myeloid origin and significantly reduces its ability to protect S. aureus from destruction in a whole blood killing (WBK) assay. Like the SSLs, SElX is up-regulated early during infection and is under the control of the S. aureus exotoxin expression (Sae) two component gene regulatory system. Additionally, the structure of SElX in complex with the sLacNac-containing tetrasaccharide sialyl Lewis X (sLeX) reveals that SElX is a unique single-domain SAg. In summary, SElX is an 'SSL-like' SAg.


Assuntos
Enterotoxinas/metabolismo , Exotoxinas/metabolismo , Evasão da Resposta Imune/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Fatores de Virulência/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Infecções Estafilocócicas/imunologia , Superantígenos/genética , Fatores de Virulência/genética
12.
Ann Rheum Dis ; 76(11): 1915-1923, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801345

RESUMO

OBJECTIVE: The pathogenetic mechanisms by which HLA-DRB1 alleles are associated with anticitrullinated peptide antibody (ACPA)-positive rheumatoid arthritis (RA) are incompletely understood. RA high-risk HLA-DRB1 alleles are known to share a common motif, the 'shared susceptibility epitope (SE)'. Here, the electropositive P4 pocket of HLA-DRB1 accommodates self-peptide residues containing citrulline but not arginine. HLA-DRB1 His/Phe13ß stratifies with ACPA-positive RA, while His13ßSer polymorphisms stratify with ACPA-negative RA and RA protection. Indigenous North American (INA) populations have high risk of early-onset ACPA-positive RA, whereby HLA-DRB1*04:04 and HLA-DRB1*14:02 are implicated as risk factors for RA in INA. However, HLA-DRB1*14:02 has a His13ßSer polymorphism. Therefore, we aimed to verify this association and determine its molecular mechanism. METHODS: HLA genotype was compared in 344 INA patients with RA and 352 controls. Structures of HLA-DRB1*1402-class II loaded with vimentin-64Arg59-71, vimentin-64Cit59-71 and fibrinogen ß-74Cit69-81 were solved using X-ray crystallography. Vimentin-64Cit59-71-specific and vimentin59-71-specific CD4+ T cells were characterised by flow cytometry using peptide-histocompatibility leukocyte antigen (pHLA) tetramers. After sorting of antigen-specific T cells, TCRα and ß-chains were analysed using multiplex, nested PCR and sequencing. RESULTS: ACPA+ RA in INA was independently associated with HLA-DRB1*14:02. Consequent to the His13ßSer polymorphism and altered P4 pocket of HLA-DRB1*14:02, both citrulline and arginine were accommodated in opposite orientations. Oligoclonal autoreactive CD4+ effector T cells reactive with both citrulline and arginine forms of vimentin59-71 were observed in patients with HLA-DRB1*14:02+ RA and at-risk ACPA- first-degree relatives. HLA-DRB1*14:02-vimentin59-71-specific and HLA-DRB1*14:02-vimentin-64Cit59-71-specific CD4+ memory T cells were phenotypically distinct populations. CONCLUSION: HLA-DRB1*14:02 broadens the capacity for citrullinated and native self-peptide presentation and T cell expansion, increasing risk of ACPA+ RA.


Assuntos
Nativos do Alasca/genética , Artrite Reumatoide/etnologia , Artrite Reumatoide/genética , Predisposição Genética para Doença/etnologia , Cadeias HLA-DRB1/genética , Índios Norte-Americanos/genética , Alaska/etnologia , Alelos , Arginina/genética , Arginina/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Linfócitos T CD4-Positivos/imunologia , Canadá/etnologia , Estudos de Casos e Controles , Citrulina/genética , Citrulina/imunologia , Feminino , Citometria de Fluxo , Genótipo , Humanos , Masculino , Peptídeos Cíclicos/imunologia , Polimorfismo Genético , Fatores de Risco , Vimentina/genética
13.
IUCrJ ; 3(Pt 1): 10-9, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26870377

RESUMO

Bacterial type I signal peptidases (SPases) are membrane-anchored serine proteases that process the signal peptides of proteins exported via the Sec and Tat secretion systems. Despite their crucial importance for bacterial virulence and their attractiveness as drug targets, only one such enzyme, LepB from Escherichia coli, has been structurally characterized, and the transient nature of peptide binding has stymied attempts to directly visualize SPase-substrate complexes. Here, the crystal structure of SpsB, the type I signal peptidase from the Gram-positive pathogen Staphylococcus aureus, is reported, and a peptide-tethering strategy that exploits the use of carrier-driven crystallization is described. This enabled the determination of the crystal structures of three SpsB-peptide complexes, both with cleavable substrates and with an inhibitory peptide. SpsB-peptide interactions in these complexes are almost exclusively limited to the canonical signal-peptide motif Ala-X-Ala, for which clear specificity pockets are found. Minimal contacts are made outside this core, with the variable side chains of the peptides accommodated in shallow grooves or exposed faces. These results illustrate how high fidelity is retained despite broad sequence diversity, in a process that is vital for cell survival.

14.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 1): 61-5, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25615971

RESUMO

Staphylococcus aureus infections are becoming increasingly difficult to treat as they rapidly develop resistance to existing antibiotics. Bacterial type I signal peptidases are membrane-associated, cell-surface serine proteases with a unique catalytic mechanism that differs from that of eukaryotic endoplasmic reticulum signal peptidases. They are thus potential antimicrobial targets. S. aureus has a catalytically active type I signal peptidase, SpsB, that is essential for cell viability. To elucidate its structure, the spsB gene from S. aureus Newman strain was cloned and overexpressed in Escherichia coli. After exploring many different protein-modification constructs, SpsB was expressed as a fusion protein with maltose-binding protein and crystallized by hanging-drop vapour diffusion. The crystals belonged to the monoclinic space group P2(1) and diffracted to 2.05 Šresolution. The crystal structure of SpsB is anticipated to provide structural insight into Gram-positive signal peptidases and to aid in the development of antibacterial agents that target type I signal peptidases.


Assuntos
Proteínas de Bactérias/química , Proteínas de Membrana/química , Serina Endopeptidases/química , Staphylococcus aureus/enzimologia , Substituição de Aminoácidos , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/isolamento & purificação , Cristalização , Cristalografia por Raios X , Proteínas Ligantes de Maltose/biossíntese , Proteínas Ligantes de Maltose/química , Proteínas Ligantes de Maltose/isolamento & purificação , Proteínas de Membrana/biossíntese , Proteínas de Membrana/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Serina Endopeptidases/biossíntese , Serina Endopeptidases/isolamento & purificação
15.
Nephrology (Carlton) ; 17(1): 11-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22050577

RESUMO

Confirmation of kidney transplant rejection still requires a histological diagnosis on renal allograft biopsy. Research continues for new non-invasive means for early diagnosis and treatment of kidney allograft rejection. Examination of the urine in renal transplant recipients provides a logical and readily accessible non-invasive window on allograft function, reflecting the function of the kidney in its transplanted environment. Renal tubular epithelial cells (TEC) respond dynamically to the surrounding microenvironment and play an important role in allograft survival. Proteins released from TEC into the urine potentially serve as biomarkers for the early diagnosis of graft dysfunction and rejection. Activated proximal TEC express human leucocyte antigens and co-stimulatory molecules, transiently transforming into non-professional antigen-presenting cells that augment renal allograft rejection. Chemokines and chemoattractants expressed on proximal tubules may also facilitate the migration of alloreactive lymphocytes to local site of injury and stimulate cytokine release from infiltrating lymphocytes. Proximal TEC are also potential targets for circulating alloreactive antibodies and complement leading to cell damage. Changes in cell state during development, regeneration or immune response require a rapid modulation of both surface protein expression and secretion, altering the repertoire of proteins secreted or expressed at the TEC plasma membrane. Due to the proximity of TEC to the tubular lumen, these proteins are passed into the urine. In this regard, TEC possess a unique anatomic location within the transplanted organ and are therefore ideal indicators of graft function. Hence, measurement of the changes of TEC-derived molecules in the urine, in response to different challenges or modification, may predict graft outcome.


Assuntos
Biomarcadores/urina , Fatores Quimiotáticos/urina , Rejeição de Enxerto , Sobrevivência de Enxerto/imunologia , Transplante de Rim/efeitos adversos , Proteínas de Membrana/urina , Movimento Celular/imunologia , Fatores Quimiotáticos/imunologia , Fatores Quimiotáticos/metabolismo , Diagnóstico Precoce , Células Epiteliais/imunologia , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/urina , Humanos , Imunidade Celular , Transplante de Rim/imunologia , Túbulos Renais Proximais/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Valor Preditivo dos Testes
16.
Transplantation ; 89(9): 1071-8, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20098353

RESUMO

BACKGROUND.: Urine is a potentially rich source of biomarkers for monitoring kidney dysfunction. In this study, we have investigated the potential of soluble human leukocyte antigen (sHLA)-DR in the urine for noninvasive monitoring of renal transplant patients. METHODS.: Urinary soluble HLA-DR levels were measured by sandwich enzyme-linked immunosorbent assay in 103 patients with renal diseases or after renal transplantation. sHLA-DR in urine was characterized by Western blotting and mass spectrometry. RESULTS.: Acute graft rejection was associated with a significantly elevated level of urinary sHLA-DR (P<0.0001), compared with recipients with stable graft function or healthy individuals. A receiver operating characteristic curve analysis showed the area under the curve to be 0.88 (P<0.001). At a selected threshold, the sensitivity was 80% and specificity was 98% for detection of acute renal transplant rejection. sHLA-DR was not exosomally associated and was of lower molecular weight compared with the HLA-DR expressed as heterodimer on the plasma membrane of antigen-presenting cells. CONCLUSIONS.: sHLA-DR excreted into urine is a promising indicator of renal transplant rejection.


Assuntos
Biomarcadores/urina , Rejeição de Enxerto/urina , Antígenos HLA-DR/urina , Transplante de Rim/efeitos adversos , Doença Aguda , Adulto , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/patologia , Antígenos HLA-DR/sangue , Humanos , Inflamação/sangue , Inflamação/imunologia , Nefropatias/classificação , Nefropatias/cirurgia , Transplante de Rim/patologia , Pessoa de Meia-Idade , Solubilidade
17.
Hybridoma (Larchmt) ; 28(6): 423-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20025501

RESUMO

We report the generation of a monoclonal antibody (MAb) that reacts to the N-terminus of the denatured HLA-DRalpha chain. The 1C4.6 MAb was raised against a peptide corresponding to amino acid residues 10 to 32 of a highly conserved region within the alpha1 domain of HLA-DR. This region partially overlaps with the epitope recognized by the conformationally dependent L243 MAb. In Western blot analysis, MAb 1C4.6 reacted with denatured HLA-DRalpha chains, but failed to bind the HLA-DRbeta chain expressed individually by transfectant cells, confirming that it recognizes an epitope on the alpha-chain of HLA-DR. In addition, this antibody was found to be isotype specific to HLA-DRalpha, as it did not cross-react to HLA class II proteins HLA-DP and-HLA-DQ. The 1C4.6 MAb is a valuable addition to existing reagents used to probe the structure and function of MHC class II molecules. This anti-HLA-DRalpha1 domain MAb may prove valuable for studies of HLA class II heterodimer assembly, structure, and function, as well as for studies into the release of soluble MHC class II.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Antígenos HLA-DR/imunologia , Hibridomas/imunologia , Sequência de Aminoácidos , Animais , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Epitopos/genética , Epitopos/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular
18.
Microbiology (Reading) ; 153(Pt 10): 3218-3227, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17906121

RESUMO

Unlike the colicins, microcins and related peptide antibiotics, little is known about antibiotic proteins (M(r)>10,000) from Gram-positive bacteria, since only few examples have been described to date. In this study we used heterologous expression of recombinant proteins to access the 17 kDa antibiotic protein SA-M57 from Streptococcus pyogenes, along with two proteins of unknown function identified in publicly available databases: EF1097 from Enterococcus faecalis and YpkK from Corynebacterium jeikeium. Here we show that all three are antibiotic proteins with different spectra of antimicrobial activity that kill sensitive bacteria at nanomolar concentrations. In silico structure predictions indicate that although the three proteins share little sequence similarity, they may be composed of conserved secondary structural elements: a relatively unstructured, acidic N-terminal portion and a basic C-terminal portion characterized by two helical elements separated by a loop structure and stabilized by an essential disulphide. Expression of individual segments as well as protein chimaeras revealed that, at least in the case of YpkK, the C-terminal portion is responsible for the killing action of the protein, whereas the role of the N-terminal portion remains unclear. Both scnM57 and ef1097 appear to be widely distributed in Strep. pyogenes and Ent. faecalis (respectively), whereas ypkK is found only rarely amongst clinical isolates of C. jeikeium. Finally, we determined that the proteins kill sensitive bacteria without lysis, a feature that distinguishes them from known murolytic proteins.


Assuntos
Antibacterianos/farmacologia , Bacteriocinas/genética , Bacteriocinas/farmacologia , Corynebacterium/genética , Enterococcus faecalis/genética , Streptococcus pyogenes/genética , Sequência de Aminoácidos , Antibacterianos/química , Bacteriocinas/química , Expressão Gênica , Bactérias Gram-Positivas/efeitos dos fármacos , Viabilidade Microbiana , Dados de Sequência Molecular , Peso Molecular , Estrutura Secundária de Proteína
19.
Appl Environ Microbiol ; 71(11): 7613-7, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16269816

RESUMO

Streptococcus mutans UA159, the genome sequence reference strain, exhibits nonlantibiotic mutacin activity. In this study, bioinformatic and mutational analyses were employed to demonstrate that the antimicrobial repertoire of strain UA159 includes mutacin IV (specified by the nlm locus) and a newly identified bacteriocin, mutacin V (encoded by SMU.1914c).


Assuntos
Proteínas de Bactérias/genética , Bacteriocinas/biossíntese , Genoma Bacteriano , Streptococcus mutans/genética , Proteínas de Bactérias/metabolismo , Biologia Computacional , Testes de Sensibilidade Microbiana , Mutagênese , Padrões de Referência , Streptococcus mitis/efeitos dos fármacos , Streptococcus mutans/classificação , Streptococcus mutans/metabolismo , Streptococcus oralis/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...