Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(7): e106103, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33522633

RESUMEN

Streptococcus agalactiae, also known as group B Streptococcus (GBS), is the major cause of neonatal sepsis in humans. A critical step to infection is adhesion of bacteria to epithelial surfaces. GBS adhesins have been identified to bind extracellular matrix components and cellular receptors. However, several putative adhesins have no host binding partner characterised. We report here that surface-expressed ß protein of GBS binds to human CEACAM1 and CEACAM5 receptors. A crystal structure of the complex showed that an IgSF domain in ß represents a novel Ig-fold subtype called IgI3, in which unique features allow binding to CEACAM1. Bioinformatic assessment revealed that this newly identified IgI3 fold is not exclusively present in GBS but is predicted to be present in adhesins from other clinically important human pathogens. In agreement with this prediction, we found that CEACAM1 binds to an IgI3 domain found in an adhesin from a different streptococcal species. Overall, our results indicate that the IgI3 fold could provide a broadly applied mechanism for bacteria to target CEACAMs.


Asunto(s)
Adhesinas Bacterianas/química , Antígenos CD/química , Antígeno Carcinoembrionario/química , Moléculas de Adhesión Celular/química , Adhesinas Bacterianas/metabolismo , Animales , Antígenos CD/metabolismo , Sitios de Unión , Células CHO , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Cricetinae , Cricetulus , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Células HeLa , Humanos , Unión Proteica , Streptococcus agalactiae/metabolismo
2.
Mol Microbiol ; 112(4): 1116-1130, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31290194

RESUMEN

Inhibition of cell division is critical for viability under DNA-damaging conditions. DNA damage induces the SOS response that in bacteria inhibits cell division while repairs are being made. In coccoids, such as the human pathogen, Staphylococcus aureus, this process remains poorly studied. Here, we identify SosA as the staphylococcal SOS-induced cell division inhibitor. Overproduction of SosA inhibits cell division, while sosA inactivation sensitizes cells to genotoxic stress. SosA is a small, predicted membrane protein with an extracellular C-terminal domain in which point mutation of residues that are conserved in staphylococci and major truncations abolished the inhibitory activity. In contrast, a minor truncation led to SosA accumulation and a strong cell division inhibitory activity, phenotypically similar to expression of wild-type SosA in a CtpA membrane protease mutant. This suggests that the extracellular C-terminus of SosA is required both for cell division inhibition and for turnover of the protein. Microscopy analysis revealed that SosA halts cell division and synchronizes the cell population at a point where division proteins such as FtsZ and EzrA are localized at midcell, and the septum formation is initiated but unable to progress to closure. Thus, our findings show that SosA is central in cell division regulation in staphylococci.


Asunto(s)
División Celular/genética , División Celular/fisiología , Respuesta SOS en Genética/fisiología , Proteínas Bacterianas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Daño del ADN/genética , Daño del ADN/fisiología , Proteínas de la Membrana/metabolismo , Respuesta SOS en Genética/genética , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
3.
J Immunol ; 193(1): 317-26, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24850720

RESUMEN

IgA nephropathy (IgAN) is characterized by mesangial cell proliferation and extracellular matrix expansion associated with immune deposits consisting of galactose-deficient polymeric IgA1 and C3. We have previously shown that IgA-binding regions of streptococcal M proteins colocalize with IgA in mesangial immune deposits in patients with IgAN. In the present study, the IgA-binding M4 protein from group A Streptococcus was found to bind to galactose-deficient polymeric IgA1 with higher affinity than to other forms of IgA1, as shown by surface plasmon resonance and solid-phase immunoassay. The M4 protein was demonstrated to bind to mesangial cells not via the IgA-binding region but rather via the C-terminal region, as demonstrated by flow cytometry. IgA1 enhanced binding of M4 to mesangial cells, but not vice versa. Costimulation of human mesangial cells with M4 and galactose-deficient polymeric IgA1 resulted in a significant increase in IL-6 secretion compared with each stimulant alone. Galactose-deficient polymeric IgA1 alone, but not M4, induced C3 secretion from the cells, and costimulation enhanced this effect. Additionally, costimulation enhanced mesangial cell proliferation compared with each stimulant alone. These results indicate that IgA-binding M4 protein binds preferentially to galactose-deficient polymeric IgA1 and that these proteins together induce excessive proinflammatory responses and proliferation of human mesangial cells. Thus, tissue deposition of streptococcal IgA-binding M proteins may contribute to the pathogenesis of IgAN.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Complemento C3/inmunología , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/inmunología , Interleucina-6/inmunología , Células Mesangiales/inmunología , Streptococcus/inmunología , Adolescente , Femenino , Glomerulonefritis por IGA/patología , Humanos , Masculino , Células Mesangiales/patología , Persona de Mediana Edad
4.
PLoS Pathog ; 9(4): e1003323, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637608

RESUMEN

Many pathogens express a surface protein that binds the human complement regulator factor H (FH), as first described for Streptococcus pyogenes and the antiphagocytic M6 protein. It is commonly assumed that FH recruited to an M protein enhances virulence by protecting the bacteria against complement deposition and phagocytosis, but the role of FH-binding in S. pyogenes pathogenesis has remained unclear and controversial. Here, we studied seven purified M proteins for ability to bind FH and found that FH binds to the M5, M6 and M18 proteins but not the M1, M3, M4 and M22 proteins. Extensive immunochemical analysis indicated that FH binds solely to the hypervariable region (HVR) of an M protein, suggesting that selection has favored the ability of certain HVRs to bind FH. These FH-binding HVRs could be studied as isolated polypeptides that retain ability to bind FH, implying that an FH-binding HVR represents a distinct ligand-binding domain. The isolated HVRs specifically interacted with FH among all human serum proteins, interacted with the same region in FH and showed species specificity, but exhibited little or no antigenic cross-reactivity. Although these findings suggested that FH recruited to an M protein promotes virulence, studies in transgenic mice did not demonstrate a role for bound FH during acute infection. Moreover, phagocytosis tests indicated that ability to bind FH is neither sufficient nor necessary for S. pyogenes to resist killing in whole human blood. While these data shed new light on the HVR of M proteins, they suggest that FH-binding may affect S. pyogenes virulence by mechanisms not assessed in currently used model systems.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Animales , Sitios de Unión , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis , Unión Proteica , Estructura Terciaria de Proteína , Especificidad de la Especie , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo
5.
Nat Commun ; 14(1): 2275, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37080973

RESUMEN

Life-threatening bacterial infections in women after childbirth, known as puerperal sepsis, resulted in classical epidemics and remain a global health problem. While outbreaks of puerperal sepsis have been ascribed to Streptococcus pyogenes, little is known about disease mechanisms. Here, we show that the bacterial R28 protein, which is epidemiologically associated with outbreaks of puerperal sepsis, specifically targets the human receptor CEACAM1. This interaction triggers events that would favor the development of puerperal sepsis, including adhesion to cervical cells, suppression of epithelial wound repair and subversion of innate immune responses. High-resolution structural analysis showed that an R28 domain with IgI3-like fold binds to the N-terminal domain of CEACAM1. Together, these findings demonstrate that a single adhesin-receptor interaction can drive the pathogenesis of bacterial sepsis and provide molecular insights into the pathogenesis of one of the most important infectious diseases in medical history.


Asunto(s)
Infección Puerperal , Sepsis , Infecciones Estreptocócicas , Femenino , Humanos , Embarazo , Adhesinas Bacterianas/genética , Proteínas Bacterianas/genética , Infección Puerperal/epidemiología , Infección Puerperal/microbiología , Sepsis/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes
6.
Nature ; 442(7105): 943-6, 2006 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-16929299

RESUMEN

All living cells require specific mechanisms that target proteins to the cell surface. In eukaryotes, the first part of this process involves recognition in the endoplasmic reticulum of amino-terminal signal sequences and translocation through Sec translocons, whereas subsequent targeting to different surface locations is promoted by internal sorting signals. In bacteria, N-terminal signal sequences promote translocation across the cytoplasmic membrane, which surrounds the entire cell, but some proteins are nevertheless secreted in one part of the cell by poorly understood mechanisms. Here we analyse localized secretion in the Gram-positive pathogen Streptococcus pyogenes, and show that the signal sequences of two surface proteins, M protein and protein F (PrtF), direct secretion to different subcellular regions. The signal sequence of M protein promotes secretion at the division septum, whereas that of PrtF preferentially promotes secretion at the old pole. Our work therefore shows that a signal sequence may contain information that directs the secretion of a protein to one subcellular region, in addition to its classical role in promoting secretion. This finding identifies a new level of complexity in protein translocation and emphasizes the potential of bacterial systems for the analysis of fundamental cell-biological problems.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Señales de Clasificación de Proteína/fisiología , Streptococcus pyogenes/citología , Streptococcus pyogenes/metabolismo , Adhesinas Bacterianas/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Portadoras/genética , Señales de Clasificación de Proteína/genética , Estructura Terciaria de Proteína , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/genética
7.
Adv Exp Med Biol ; 946: 87-112, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21948364

RESUMEN

A common site in the constant region (Fc) of immunoglobulins is recognized by host receptors and is a frequent target of proteins expressed by pathogens. This site is located at the junction of two constant domains in the antibody heavy chains and produces a large shallow cavity formed by loops of the CH2 and CH3 domains in IgG and IgA (CH3 and CH4 domains in IgM). Crystal structures have been determined for complexes of IgG-Fc and IgA-Fc with a structurally diverse set of host, pathogen and in vitro selected ligands. While pathogen proteins may directly block interactions with the immunoglobulins thereby evading host immunity, it is likely that the same pathogen molecules also interact with other host factors to carry out their primary biological function. Herein we review the structural and functional aspects of host and pathogen molecular recognition of the common site on the Fc of immunoglobulins. We also propose that some pathogen proteins may promote virulence by affecting the bridging between innate and adaptive immunity.


Asunto(s)
Anticuerpos Antibacterianos/química , Anticuerpos Antibacterianos/inmunología , Infecciones Bacterianas/inmunología , Sitios de Unión de Anticuerpos/inmunología , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/inmunología , Bacterias/inmunología , Humanos , Estructura Terciaria de Proteína , Relación Estructura-Actividad
8.
Am J Pathol ; 176(2): 608-18, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20056836

RESUMEN

IgA nephropathy (IgAN) and Henoch-Schönlein purpura (HSP) are diseases characterized by IgA deposits in the kidney and/or skin. Both may arise after upper respiratory tract infections, but the pathogenic mechanisms governing these diseases remain unclear. Patients with IgAN (n = 16) and HSP (n = 17) were included in this study aimed at examining whether IgA-binding M proteins of group A streptococci could be involved. As M proteins vary in sequence, the study focused on the IgA-binding-region (IgA-BR) of three different M proteins: M4, M22, and M60. Renal tissue from IgAN and HSP patients and skin from HSP patients were examined for deposits of streptococcal IgA-BR by immunohistochemistry and electron microscopy using specific antibodies, and a skin sample from a HSP patient was examined by mass spectrometry. IgA-BR deposits were detected in 10/16 IgAN kidneys and 7/13 HSP kidneys. Electron microscopy demonstrated deposits of IgA-BRs in the mesangial matrix and glomerular basement membrane, which colocalized with IgA. Skin samples exhibited IgA-BR deposits in 4/5 biopsies, a result confirmed by mass spectrometry in one patient. IgA-BR deposits were not detected in normal kidney and skin samples. Taken together, these results demonstrate IgA-BR from streptococcal M proteins in patient tissues. IgA-BR, would on gaining access to the circulation, encounter circulatory IgA and form a complex with IgA-Fc that could deposit in tissues and contribute to the pathogenesis of IgAN and HSP.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Glomerulonefritis por IGA/metabolismo , Vasculitis por IgA/metabolismo , Inmunoglobulina A/metabolismo , Precipitinas/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/inmunología , Biopsia , Proteínas Portadoras/química , Proteínas Portadoras/inmunología , Niño , Preescolar , Femenino , Glomerulonefritis por IGA/patología , Humanos , Vasculitis por IgA/patología , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Masculino , Microscopía Electrónica , Datos de Secuencia Molecular , Precipitinas/ultraestructura , Unión Proteica , Piel/metabolismo , Piel/patología , Piel/ultraestructura , Adulto Joven
9.
J Exp Med ; 198(7): 1057-68, 2003 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-14517274

RESUMEN

The M protein of Streptococcus pyogenes is a major bacterial virulence factor that confers resistance to phagocytosis. To analyze how M protein allows evasion of phagocytosis, we used the M22 protein, which has features typical of many M proteins and has two well-characterized regions binding human plasma proteins: the hypervariable NH2-terminal region binds C4b-binding protein (C4BP), which inhibits the classical pathway of complement activation; and an adjacent semivariable region binds IgA-Fc. Characterization of chromosomal S. pyogenes mutants demonstrated that each of the ligand-binding regions contributed to phagocytosis resistance, which could be fully explained as cooperation between the two regions. Deposition of complement on S. pyogenes occurred almost exclusively via the classical pathway, even under nonimmune conditions, but was down-regulated by bacteria-bound C4BP, providing an explanation for the ability of bound C4BP to inhibit phagocytosis. Different opsonizing antisera shared the ability to block binding of both C4BP and IgA, suggesting that the two regions in M22 play important roles also under immune conditions, as targets for protective antibodies. These data indicate that M22 and similar M proteins confer resistance to phagocytosis through ability to bind two components of the human immune system.


Asunto(s)
Antígenos Bacterianos , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Portadoras/fisiología , Proteínas Inactivadoras de Complemento/metabolismo , Glicoproteínas , Inmunoglobulina A/metabolismo , Fagocitosis , Streptococcus pyogenes/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/química , Sitios de Unión , Proteínas Portadoras/química , Vía Clásica del Complemento , Humanos , Datos de Secuencia Molecular , Conejos
10.
Eur J Immunol ; 39(4): 1147-56, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19266484

RESUMEN

Here we unravel the structural features of human IgM and IgA that govern their interaction with the human Fcalpha/mu receptor (hFcalpha/muR). Ligand polymerization status was crucial for the interaction, because hFcalpha/muR binding did not occur with monomeric Ab of either class. hFcalpha/muR bound IgM with an affinity in the nanomolar range, whereas the affinity for dimeric IgA (dIgA) was tenfold lower. Panels of mutant IgM and dIgA were used to identify regions critical for hFcalpha/muR binding. IgM binding required contributions from both Cmu3 and Cmu4 Fc domains, whereas for dIgA, an exposed loop in the Calpha3 domain was crucial. This loop, comprising residues Pro440-Phe443, lies at the Fc domain interface and has been implicated in the binding of host receptors FcalphaRI and polymeric Ig receptor (pIgR), as well as IgA-binding proteins produced by certain pathogenic bacteria. Substitutions within the Pro440-Phe443 loop resulted in loss of hFcalpha/muR binding. Furthermore, secretory component (SC, the extracellular portion of pIgR) and bacterial IgA-binding proteins were shown to inhibit the dIgA-hFcalpha/muR interaction. Therefore, we have identified a motif in the IgA-Fc inter-domain region critical for hFcalpha/muR interaction, and highlighted the multi-functional nature of a key site for protein-protein interaction at the IgA Fc domain interface.


Asunto(s)
Afinidad de Anticuerpos , Inmunoglobulina A/química , Inmunoglobulina M/química , Receptores Fc/inmunología , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Afinidad de Anticuerpos/genética , Afinidad de Anticuerpos/inmunología , Células COS , Chlorocebus aethiops , Humanos , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Proteínas Mutantes/inmunología , Mutación , Dominios y Motivos de Interacción de Proteínas/inmunología , Multimerización de Proteína , Estructura Terciaria de Proteína , Receptores Fc/genética
11.
Nephrol Dial Transplant ; 25(10): 3434-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20558662

RESUMEN

BACKGROUND: IgA nephropathy (IgAN), the most common glomerulonephritis worldwide, is characterized by mesangial deposits containing predominantly IgA. IgAN commonly occurs or exacerbates after upper respiratory tract infections such as streptococcal pharyngitis. Certain group A streptococci express M proteins with IgA-binding regions (IgA-BRs). We have previously shown that these IgA-BRs co-localize with mesangial IgA in IgAN. METHODS: Blood samples from patients with IgAN (n = 21) and age-matched controls (n = 83) were assayed by ELISA to detect an IgG antibody response to the IgA-BRs of the M4, M22 and M60 proteins. Antibodies were assayed for each IgA-BR separately and the results were combined. RESULTS: Antibody levels to the IgA-BRs were significantly higher in IgAN patients than controls (P = 0.016), particularly in patients with recent streptococcal infection (P = 0.008). Conclusions. The results suggest that children with IgAN had a previous infection with a streptococcal strain expressing an IgA-binding M protein.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/metabolismo , Adolescente , Adulto , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Niño , Preescolar , Femenino , Humanos , Masculino
12.
Microbiol Mol Biol Rev ; 85(1)2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33239435

RESUMEN

Vaccines work primarily by eliciting antibodies, even when recovery from natural infection depends on cellular immunity. Large efforts have therefore been made to identify microbial antigens that elicit protective antibodies, but these endeavors have encountered major difficulties, as witnessed by the lack of vaccines against many pathogens. This review summarizes accumulating evidence that subdominant protein regions, i.e., surface-exposed regions that elicit relatively weak antibody responses, are of particular interest for vaccine development. This concept may seem counterintuitive, but subdominance may represent an immune evasion mechanism, implying that the corresponding region potentially is a key target for protective immunity. Following a presentation of the concepts of immunodominance and subdominance, the review will present work on subdominant regions in several major human pathogens: the protozoan Plasmodium falciparum, two species of pathogenic streptococci, and the dengue and influenza viruses. Later sections are devoted to the molecular basis of subdominance, its potential role in immune evasion, and general implications for vaccine development. Special emphasis will be placed on the fact that a whole surface-exposed protein domain can be subdominant, as demonstrated for all of the pathogens described here. Overall, the available data indicate that subdominant protein regions are of much interest for vaccine development, not least in bacterial and protozoal systems, for which antibody subdominance remains largely unexplored.


Asunto(s)
Formación de Anticuerpos/inmunología , Epítopos Inmunodominantes/inmunología , Proteínas de la Membrana/inmunología , Vacunas/inmunología , Virus del Dengue/inmunología , Humanos , Evasión Inmune/inmunología , Orthomyxoviridae/inmunología , Plasmodium falciparum/inmunología , Dominios Proteicos/inmunología , Streptococcus agalactiae/inmunología , Streptococcus pyogenes/inmunología , Vacunación
13.
J Mol Biol ; 369(1): 69-78, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17418236

RESUMEN

Glycosylation defects occur in several human diseases. In IgA nephropathy, IgA1 contains O-glycans that are galactose-deficient and consist mostly of core 1 alpha2,6 sialylated N-acetylgalactosamine, a configuration suspected to prevent beta1,3 galactosylation. We confirmed the same aberrancy in IgA1 secreted by the human DAKIKI B cell line. Biochemical assays indicated CMP-NeuAc:GalNAc-IgA1 alpha2,6-sialyltransferase activity in this cell line. However, a candidate enzyme, ST6-GalNAcI, was not transcribed in DAKIKI cells, B cells isolated from blood, or Epstein-Barr virus (EBV)-immortalized IgA1-producing cells from the blood of IgAN patients and healthy controls. Instead, ST6-GalNAcII transcription was detected at a high level. Expression of the ST6-GalNAcII gene and activity of the CMP-NeuAc:GalNAc-IgA1 alpha2,6-sialyltransferase were higher in IgA1-producing cell lines from IgAN patients than in such cells from healthy controls. These data are the first evidence that human cells that lack ST6-GalNAcI can sialylate core 1 GalNAc-Ser/Thr.


Asunto(s)
Inmunoglobulina A/biosíntesis , Sialiltransferasas/metabolismo , Línea Celular , Línea Celular Transformada , Ensayo de Inmunoadsorción Enzimática , Regulación Enzimológica de la Expresión Génica , Glicosilación , Células HT29 , Herpesvirus Humano 4/metabolismo , Humanos , Lectinas/metabolismo , Leucocitos Mononucleares/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Reversa/genética , Sialiltransferasas/genética , beta-D-Galactósido alfa 2-6-Sialiltransferasa
14.
PLoS Pathog ; 2(5): e47, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16733543

RESUMEN

Many pathogenic microorganisms evade host immunity through extensive sequence variability in a protein region targeted by protective antibodies. In spite of the sequence variability, a variable region commonly retains an important ligand-binding function, reflected in the presence of a highly conserved sequence motif. Here, we analyze the limits of sequence divergence in a ligand-binding region by characterizing the hypervariable region (HVR) of Streptococcus pyogenes M protein. Our studies were focused on HVRs that bind the human complement regulator C4b-binding protein (C4BP), a ligand that confers phagocytosis resistance. A previous comparison of C4BP-binding HVRs identified residue identities that could be part of a binding motif, but the extended analysis reported here shows that no residue identities remain when additional C4BP-binding HVRs are included. Characterization of the HVR in the M22 protein indicated that two relatively conserved Leu residues are essential for C4BP binding, but these residues are probably core residues in a coiled-coil, implying that they do not directly contribute to binding. In contrast, substitution of either of two relatively conserved Glu residues, predicted to be solvent-exposed, had no effect on C4BP binding, although each of these changes had a major effect on the antigenic properties of the HVR. Together, these findings show that HVRs of M proteins have an extraordinary capacity for sequence divergence and antigenic variability while retaining a specific ligand-binding function.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Streptococcus pyogenes/metabolismo , Secuencia de Aminoácidos , Antígenos/biosíntesis , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteína de Unión al Complemento C4b/metabolismo , Regiones Determinantes de Complementariedad , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína
16.
J Immunol Methods ; 297(1-2): 83-95, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15777933

RESUMEN

Many Gram-positive bacteria express surface proteins that bind human plasma proteins. These bacterial proteins, and derivatives of them, are of interest for analysis of bacterial pathogenesis and as immunochemical tools. Well-characterized examples include the IgG-binding reagents staphylococcal protein A and streptococcal protein G, and the recently described streptococcal IgA-binding peptide Sap. Here, we show that a peptide derived from the streptococcal M22 protein can be used for single-step affinity purification of the human complement regulator C4b-binding protein (C4BP). Binding of C4BP was strongly enhanced by dimerization of the peptide via a C-terminal cysteine residue not present in the intact M22 protein. The purified C4BP had the expected binding characteristics, and acted as a cofactor for factor I in the degradation of C4b. Passage of serum through a peptide column under non-saturating conditions resulted in binding of >99.5% of serum C4BP, implying that such a column can be used to deplete serum of C4BP. These data indicate that the C4BP-binding peptide is a versatile tool that can be used for simple and rapid purification of biologically active human C4BP or for removal of C4BP from serum.


Asunto(s)
Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Cromatografía de Afinidad/métodos , Antígenos de Histocompatibilidad/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Bovinos , Proteína de Unión al Complemento C4b , Dimerización , Antígenos de Histocompatibilidad/sangre , Antígenos de Histocompatibilidad/química , Humanos , Ratones , Datos de Secuencia Molecular , Péptidos/química , Conejos
17.
Microbiologyopen ; 4(5): 774-89, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26175306

RESUMEN

The M protein of Streptococcus pyogenes, a major bacterial virulence factor, has an amino-terminal hypervariable region (HVR) that is a target for type-specific protective antibodies. Intriguingly, the HVR elicits a weak antibody response, indicating that it escapes host immunity by two mechanisms, sequence variability and weak immunogenicity. However, the properties influencing the immunogenicity of regions in an M protein remain poorly understood. Here, we studied the antibody response to different regions of the classical M1 and M5 proteins, in which not only the HVR but also the adjacent fibrinogen-binding B repeat region exhibits extensive sequence divergence. Analysis of antisera from S. pyogenes-infected patients, infected mice, and immunized mice showed that both the HVR and the B repeat region elicited weak antibody responses, while the conserved carboxy-terminal part was immunodominant. Thus, we identified a correlation between sequence variability and weak immunogenicity for M protein regions. A potential explanation for the weak immunogenicity was provided by the demonstration that protease digestion selectively eliminated the HVR-B part from whole M protein-expressing bacteria. These data support a coherent model, in which the entire variable HVR-B part evades antibody attack, not only by sequence variability but also by weak immunogenicity resulting from protease attack.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Variación Genética , Streptococcus pyogenes/genética , Streptococcus pyogenes/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Humanos , Evasión Inmune , Ratones , Infecciones Estreptocócicas/inmunología
18.
PLoS One ; 8(11): e81303, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278416

RESUMEN

Recent studies indicate that defective activity of complement factor H (FH) is associated with several human diseases, suggesting that pure FH may be used for therapy. Here, we describe a simple method to isolate human FH, based on the specific interaction between FH and the hypervariable region (HVR) of certain Streptococcus pyogenes M proteins. Special interest was focused on the FH polymorphism Y402H, which is associated with the common eye disease age-related macular degeneration (AMD) and has also been implicated in the binding to M protein. Using a fusion protein containing two copies of the M5-HVR, we found that the Y402 and H402 variants of FH could be efficiently purified by single-step affinity chromatography from human serum containing the corresponding protein. Different M proteins vary in their binding properties, and the M6 and M5 proteins, but not the M18 protein, showed selective binding of the FH Y402 variant. Accordingly, chromatography on a fusion protein derived from the M6-HVR allowed enrichment of the Y402 protein from serum containing both variants. Thus, the exquisite binding specificity of a bacterial protein can be exploited to develop a simple and robust procedure to purify FH and to enrich for the FH variant that protects against AMD.


Asunto(s)
Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Proteínas Portadoras/química , Cromatografía de Afinidad , Péptidos/química , Sustitución de Aminoácidos , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Factor H de Complemento/química , Factor H de Complemento/genética , Factor H de Complemento/aislamiento & purificación , Factor H de Complemento/metabolismo , Variación Genética , Humanos , Péptidos/metabolismo , Unión Proteica
20.
Cell Host Microbe ; 10(2): 147-57, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-21843871

RESUMEN

Sequence variation of antigenic proteins allows pathogens to evade antibody attack. The variable protein commonly includes a hypervariable region (HVR), which represents a key target for antibodies and is therefore predicted to be immunodominant. To understand the mechanism(s) of antibody evasion, we analyzed the clinically important HVR-containing M proteins of the human pathogen Streptococcus pyogenes. Antibodies elicited by M proteins were directed almost exclusively against the C-terminal part and not against the N-terminal HVR. Similar results were obtained for mice and humans with invasive S. pyogenes infection. Nevertheless, only anti-HVR antibodies protected efficiently against infection, as shown by passive immunizations. The HVR fused to an unrelated protein elicited no antibodies, implying that it is inherently weakly immunogenic. These data indicate that the M protein HVR evades antibody attack not only through antigenic variation but also by weak immunogenicity, a paradoxical observation that may apply to other HVR-containing proteins.


Asunto(s)
Variación Antigénica , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/metabolismo , Evasión Inmune , Streptococcus pyogenes/inmunología , Secuencia de Aminoácidos , Animales , Formación de Anticuerpos , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/inmunología , Humanos , Sueros Inmunes/administración & dosificación , Sueros Inmunes/inmunología , Inmunización Pasiva , Inmunización Secundaria , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fagocitosis , Conejos , Proteínas Recombinantes de Fusión/inmunología , Pruebas Serológicas/métodos , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/crecimiento & desarrollo , Streptococcus pyogenes/patogenicidad , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA