Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38474124

RESUMEN

Enteropathogenic Escherichia coli (EPEC) produce a capsule of polysaccharides identical to those composing the O-antigen polysaccharide of its LPS (lipopolysaccharide) molecules. In light of this, the impact of O26 polysaccharides on the immune evasion mechanisms of capsulated O26 EPEC compared to non-capsulated enterohemorrhagic Escherichia coli (EHEC) was investigated. Our findings reveal that there was no significant difference between the levels in EPEC and EHEC of rhamnose (2.8:2.5), a molecule considered to be a PAMP (Pathogen Associated Molecular Patterns). However, the levels of glucose (10:1.69), heptose (3.6:0.89) and N-acetylglucosamine (4.5:2.10), were significantly higher in EPEC than EHEC, respectively. It was also observed that the presence of a capsule in EPEC inhibited the deposition of C3b on the bacterial surface and protected the pathogen against lysis by the complement system. In addition, the presence of a capsule also protected EPEC against phagocytosis by macrophages. However, the immune evasion provided by the capsule was overcome in the presence of anti-O26 polysaccharide antibodies, and additionally, these antibodies were able to inhibit O26 EPEC adhesion to human epithelial cells. Finally, the results indicate that O26 polysaccharides can generate an effective humoral immune response, making them promising antigens for the development of a vaccine against capsulated O26 E. coli.


Asunto(s)
Escherichia coli Enterohemorrágica , Escherichia coli Enteropatógena , Infecciones por Escherichia coli , Proteínas de Escherichia coli , Humanos , Evasión Inmune , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/farmacología , Lipopolisacáridos/farmacología , Desarrollo de Vacunas
2.
Int J Mol Sci ; 23(16)2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-36012772

RESUMEN

The authors wish to make the following corrections to this paper [...].

3.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639117

RESUMEN

Leptospirosis is a neglected infectious disease caused by pathogenic species of the genus Leptospira. The acute disease is well-described, and, although it resembles other tropical diseases, it can be diagnosed through the use of serological and molecular methods. While the chronic renal disease, carrier state, and kidney fibrosis due to Leptospira infection in humans have been the subject of discussion by researchers, the mechanisms involved in these processes are still overlooked, and relatively little is known about the establishment and maintenance of the chronic status underlying this infectious disease. In this review, we highlight recent findings regarding the cellular communication pathways involved in the renal fibrotic process, as well as the relationship between renal fibrosis due to leptospirosis and CKD/CKDu.


Asunto(s)
Fibrosis/epidemiología , Enfermedades Renales/epidemiología , Leptospira/fisiología , Leptospirosis/complicaciones , Animales , Fibrosis/microbiología , Humanos , Enfermedades Renales/microbiología , Leptospirosis/microbiología
4.
J Virol ; 90(21): 9570-9581, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27512066

RESUMEN

Dengue virus (DENV) infects millions of people worldwide and is a major public health problem. DENV nonstructural protein 1 (NS1) is a conserved glycoprotein that associates with membranes and is also secreted into the plasma in DENV-infected patients. The present study describes a novel mechanism by which NS1 inhibits the terminal complement pathway. We first identified the terminal complement regulator vitronectin (VN) as a novel DENV2 NS1 binding partner by using a yeast two-hybrid system. This interaction was further assessed by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR) assay. The NS1-VN complex was also detected in plasmas from DENV-infected patients, suggesting that this interaction occurs during DENV infection. We also demonstrated that the DENV2 NS1 protein, either by itself or by interacting with VN, hinders the formation of the membrane attack complex (MAC) and C9 polymerization. Finally, we showed that DENV2, West Nile virus (WNV), and Zika virus (ZIKV) NS1 proteins produced in mammalian cells inhibited C9 polymerization. Taken together, our results points to a role for NS1 as a terminal pathway inhibitor of the complement system. IMPORTANCE: Dengue is the most important arthropod-borne viral disease nowadays and is caused by dengue virus (DENV). The flavivirus NS1 glycoprotein has been characterized functionally as a complement evasion protein that can attenuate the activation of the classical, lectin, and alternative pathways. The present study describes a novel mechanism by which DENV NS1 inhibits the terminal complement pathway. We identified the terminal complement regulator vitronectin (VN) as a novel DENV NS1 binding partner, and the NS1-VN complex was detected in plasmas from DENV-infected patients, suggesting that this interaction occurs during DENV infection. We also demonstrated that the NS1-VN complex inhibited membrane attack complex (MAC) formation, thus interfering with the complement terminal pathway. Interestingly, NS1 itself also inhibited MAC activity, suggesting a direct role of this protein in the inhibition process. Our findings imply a role for NS1 as a terminal pathway inhibitor of the complement system.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Virus del Dengue/metabolismo , Dengue/metabolismo , Dengue/virología , Vitronectina/metabolismo , Línea Celular Tumoral , Flavivirus/metabolismo , Humanos , Unión Proteica/fisiología , Técnicas del Sistema de Dos Híbridos , Proteínas no Estructurales Virales/metabolismo , Virus del Nilo Occidental/metabolismo , Virus Zika/metabolismo , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología
5.
Infect Immun ; 83(3): 888-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534939

RESUMEN

Upon infection, pathogenic Leptospira species bind several complement regulators in order to overcome host innate immunity. We previously characterized a 20-kDa leptospiral surface protein which interacts with C4b binding protein (C4BP): leptospiral complement regulator-acquiring protein A (LcpA). Here we show that LcpA also interacts with human factor H (FH), which remains functionally active once bound to the protein. Antibodies directed against short consensus repeat 20 (SCR20) inhibited binding of FH to LcpA by approximately 90%, thus confirming that this particular domain is involved in the interaction. We have also shown for the first time that leptospires bind human vitronectin and that the interaction is mediated by LcpA. Coincubation with heparin blocked LcpA-vitronectin interaction in a dose-dependent manner, strongly suggesting that binding may occur through the heparin binding domains of vitronectin. LcpA also bound to the terminal pathway component C9 and inhibited Zn(2+)-induced polymerization and membrane attack complex (MAC) formation. Competitive binding assays indicated that LcpA interacts with C4BP, FH, and vitronectin through distinct sites. Taken together, our findings indicate that LcpA may play a role in leptospiral immune evasion.


Asunto(s)
Proteínas Bacterianas/química , Leptospira interrogans/química , Leptospira/química , Fragmentos de Péptidos/química , Vitronectina/química , Anticuerpos Monoclonales/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/inmunología , Sitios de Unión , Unión Competitiva , Activación de Complemento , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/inmunología , Complemento C9/química , Complemento C9/inmunología , Factor H de Complemento/química , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/química , Heparina/química , Humanos , Evasión Inmune , Leptospira/inmunología , Leptospira/patogenicidad , Leptospira interrogans/inmunología , Leptospira interrogans/patogenicidad , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Unión Proteica , Vitronectina/inmunología , Zinc/química
6.
J Infect Dis ; 209(6): 876-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24163418

RESUMEN

Leptospirosis is an infectious disease of public health importance. To successfully colonize the host, pathogens have evolved multiple strategies to escape the complement system. Here we demonstrate that the culture supernatant of pathogenic but not saprophytic Leptospira inhibit the three complement pathways. We showed that the proteolytic activity in the supernatants of pathogenic strains targets the central complement molecule C3 and specific proteins from each pathway, such as factor B, C2, and C4b. The proteases cleaved α and ß chains of C3 and work in synergy with host regulators to inactivate C3b. Proteolytic activity was inhibited by 1,10-phenanthroline, suggesting the participation of metalloproteases. A recombinant leptospiral metalloprotease from the thermolysin family cleaved C3 in serum and could be one of the proteases responsible for the supernatant activity. We conclude that pathogenic leptospiral proteases can deactivate immune effector molecules and represent potential targets to the development of new therapies in leptospirosis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Complemento C3/metabolismo , Leptospira/inmunología , Leptospirosis/microbiología , Péptido Hidrolasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Vía Clásica del Complemento , Humanos , Evasión Inmune , Leptospira/química , Leptospira/enzimología , Leptospira/patogenicidad , Leptospirosis/inmunología , Modelos Biológicos , Péptido Hidrolasas/inmunología , Péptido Hidrolasas/aislamiento & purificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Termolisina/química , Termolisina/metabolismo
7.
J Biol Chem ; 287(7): 4826-34, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22147698

RESUMEN

LipL32 is the most abundant outer membrane protein from pathogenic Leptospira and has been shown to bind extracellular matrix (ECM) proteins as well as Ca(2+). Recent crystal structures have been obtained for the protein in the apo- and Ca(2+)-bound forms. In this work, we produced three LipL32 mutants (D163-168A, Q67A, and S247A) and evaluated their ability to interact with Ca(2+) and with ECM glycoproteins and human plasminogen. The D163-168A mutant modifies aspartate residues involved in Ca(2+) binding, whereas the other two modify residues in a cavity on the other side of the protein structure. Loss of calcium binding in the D163-D168A mutant was confirmed using intrinsic tryptophan fluorescence, circular dichroism, and thermal denaturation whereas the Q67A and S247A mutants presented the same Ca(2+) affinity as the wild-type protein. We then evaluated if Ca(2+) binding to LipL32 would be crucial for its interaction with collagen type IV and plasma proteins fibronectin and plasminogen. Surprisingly, the wild-type protein and all three mutants, including the D163-168A variant, bound to these ECM proteins with very similar affinities, both in the presence and absence of Ca(2+) ions. In conclusion, calcium binding to LipL32 may be important to stabilize the protein, but is not necessary to mediate interaction with host extracellular matrix proteins.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Calcio/metabolismo , Colágeno Tipo IV/metabolismo , Fibronectinas/metabolismo , Leptospira/metabolismo , Lipoproteínas/metabolismo , Plasminógeno/metabolismo , Sustitución de Aminoácidos , Antígenos Bacterianos/química , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Cationes Bivalentes , Colágeno Tipo IV/química , Colágeno Tipo IV/genética , Fibronectinas/química , Fibronectinas/genética , Humanos , Leptospira/química , Leptospira/genética , Lipoproteínas/química , Lipoproteínas/genética , Mutación Missense , Plasminógeno/química , Plasminógeno/genética , Unión Proteica , Estabilidad Proteica
8.
J Infect Dis ; 205(6): 995-1004, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22291192

RESUMEN

Leptospira, the causative agent of leptospirosis, interacts with several host molecules, including extracellular matrix components, coagulation cascade proteins, and human complement regulators. Here we demonstrate that acquisition of factor H (FH) on the Leptospira surface is crucial for bacterial survival in the serum and that these spirochetes, besides interacting with FH, FH related-1, and C4b binding protein (C4BP), also acquire FH like-1 from human serum. We also demonstrate that binding to these complement regulators is mediated by leptospiral immunoglobulin-like (Lig) proteins, previously shown to interact with fibronectin, laminin, collagen, elastin, tropoelastin, and fibrinogen. Factor H binds to Lig proteins via short consensus repeat domains 5 and 20. Competition assays suggest that FH and C4BP have distinct binding sites on Lig proteins. Moreover, FH and C4BP bound to immobilized Ligs display cofactor activity, mediating C3b and C4b degradation by factor I. In conclusion, Lig proteins are multifunctional molecules, contributing to leptospiral adhesion and immune evasion.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Inactivadoras del Complemento C3b/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Leptospira/patogenicidad , Leptospirosis/inmunología , Adhesión Bacteriana , Proteínas Bacterianas/genética , Sitios de Unión , Clonación Molecular , Complemento C3b/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Evasión Inmune , Inmunoglobulinas/química , Leptospira/genética , Leptospira/metabolismo , Leptospirosis/microbiología , Plásmidos
9.
Front Cell Infect Microbiol ; 13: 1148383, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37868347

RESUMEN

Lipids are a big family of molecules with a vast number of functions in the cell membranes, within the cytoplasm, and extracellularly. Lipid droplets (LDs) are the most common storage organelles and are present in almost every tissue type in the body. They also have structural functions serving as building blocks of cellular membranes and may be precursors of other molecules such as hormones, and lipoproteins, and as messengers in signal transduction. Fatty acids (FAs), such as sterol esters and triacylglycerols, are stored in LDs and are used in ß-oxidation as fuel for tricarboxylic acid cycle (TCA) and adenosine triphosphate (ATP) generation. FA uptake and entrance in the cytoplasm are mediated by membrane receptors. After a cytoplasmic round of α- and ß-oxidation, FAs are guided into the mitochondrial matrix by the L-carnitine shuttle system, where they are fully metabolized, and enter the TCA cycle. Pathogen infections may lead to impaired lipid metabolism, usage of membrane phospholipids, and LD accumulation in the cytoplasm of infected cells. Otherwise, bacterial pathogens may use lipid metabolism as a carbon source, thus altering the reactions and leading to cellular and organelles malfunctioning. This review aims to describe cellular lipid metabolism and alterations that occur upon infections.


Asunto(s)
Ácidos Grasos , Metabolismo de los Lípidos , Ácidos Grasos/metabolismo , Fosfolípidos , Triglicéridos , Biología
10.
Vaccines (Basel) ; 11(2)2023 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-36851237

RESUMEN

Sphingomyelin is a major constituent of eukaryotic cell membranes, and if degraded by bacteria sphingomyelinases may contribute to the pathogenesis of infection. Among Leptospira spp., there are five sphingomyelinases exclusively expressed by pathogenic leptospires, in which Sph2 is expressed during natural infections, cytotoxic, and implicated in the leptospirosis hemorrhagic complications. Considering this and the lack of information about associations between Sph2 and leptospirosis severity, we use a combination of immunoinformatics approaches to identify its B-cell epitopes, evaluate their reactivity against samples from leptospirosis patients, and investigate the role of antibodies anti-Sph2 in protection against severe leptospirosis. Two B-cell epitopes, Sph2(176-191) and Sph2(446-459), were predicted in Sph2 from L. interrogans serovar Lai, presenting different levels of identity when compared with other pathogenic leptospires. These epitopes were recognized by about 40% of studied patients with a prevalence of IgG antibodies against both Sph2(176-191) and Sph2(446-459). Remarkably, just individuals with low reactivity to Sph2(176-191) presented clinical complications, while high responders had only mild symptoms. Therefore, we identified two B-cell linear epitopes, recognized by antibodies of patients with leptospirosis, that could be further explored in the development of multi-epitope vaccines against leptospirosis.

11.
Pathogens ; 11(2)2022 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-35215183

RESUMEN

Bacterial acquisition of metals from a host is an essential attribute to facilitate survival and colonization within an infected organism. Staphylococcus aureus, a bacterial pathogen of medical importance, has evolved its strategies to acquire multiple metals, including iron, manganese, and zinc. Other important strategies for the colonization and infection of the host have been reported for staphylococci and include the expression of adhesins on the bacterial surface, as well as the acquisition of host plasminogen and complement regulatory proteins. Here we assess the ability of the zinc transport protein AdcA from Staphylococcus aureus, first characterized elsewhere as a zinc-binding protein of the ABC (ATP-binding cassette) transporters, to bind to host molecules. Like other staphylococcus ion-scavenging proteins, such as MntC, a manganese-binding protein, AdcA interacts with human plasminogen. Once activated, plasmin bound to AdcA cleaves fibrinogen and vitronectin. In addition, AdcA interacts with the human negative complement regulator factor H (FH). Plasminogen and FH have been shown to bind to distinct sites on the AdcA C-terminal portion. In conclusion, our in vitro data pave the way for future studies addressing the relevance of AdcA interactions with host molecules in vivo.

12.
Front Microbiol ; 13: 1051698, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36519163

RESUMEN

Pathogenic species of Leptospira are etiologic agents of leptospirosis, an emerging zoonotic disease of worldwide extent and endemic in tropical regions. The growing number of identified leptospiral species sheds light to their genetic diversity and unique virulence mechanisms, many of them still remain unknown. Toxins and adhesins are important virulence factors in several pathogens, constituting promising antigens for the development of vaccines with cross-protection and long-lasting effect against leptospirosis. For this aim, we used the shotgun phage display technique to unravel new proteins with adhesive properties. A shotgun library was constructed using fragmented genomic DNA from Leptospira interrogans serovar Copenhageni strain Fiocruz L1-130 and pG8SAET phagemid vector. Selection of phages bearing new possible cell-binding antigens was performed against VERO cells, using BRASIL biopanning methodology. Analysis of selected clones revealed the hypothetical protein LIC10778, a potentially exposed virulence factor that belongs to the virulence-modifying (VM) protein family (PF07598), composed of 13 members in the leptospiral strain Fiocruz L1-130. Prediction of LIC10778 tertiary structure indicates that the protein contains a cellular-binding domain (N-terminal portion) and an unknown domain of no assigned activity (C-terminal portion). The predicted N-terminal domain shared structural similarities with the cell-binding and internalization domain of toxins like Ricin and Abrin, as well as to the Community-Acquired Respiratory Distress Syndrome (CARDS) toxin in Mycoplasma pneumoniae. Interestingly, recombinant portions of the N-terminal region of LIC10778 protein showed binding to laminin, collagens I and IV, vitronectin, and plasma and cell fibronectins using overlay blotting technique, especially regarding the binding site identified by phage display. These data validate our preliminary phage display biopanning and support the predicted three-dimensional models of LIC10778 protein and other members of PF07598 protein family, confirming the identification of the N-terminal cell-binding domains that are similar to ricin-like toxins. Moreover, fluorescent fused proteins also confirmed that N-terminal region of LIC10778 is capable of binding to VERO and A549 cell lines, further highlighting its virulence role during host-pathogen interaction in leptospirosis probably mediated by its C-terminal domain. Indeed, recent results in the literature confirmed this assumption by demonstrating the cytotoxicity of a closely related PF07598 member.

13.
Pathogens ; 11(8)2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-36015015

RESUMEN

The serogroup O55 of E. coli is composed of strains whose mechanisms of virulence are different from each other. Since the O55 polysaccharides are present in all E. coli O55 strains, and so are the polymers that compose the capsule of O55 atypical enteropathogenic E. coli (aEPEC), it was investigated whether anti-O55 antibodies were able to help the innate immune system to eliminate capsulated aEPEC and Shiga toxin-producing E. coli (STEC) belonging to the serogroup O55. The results demonstrate that the capsule of EPEC was able to inhibit the deposition of C3b on the bacterial surface and, as a consequence, their lysis by the alternative pathway of the complement system. However, in the presence of antibodies, the ability of the complement to lyse these pathogens was restored. It was also observed that macrophages were able to ingest EPEC and STEC, but they were only able to kill the ingested pathogens in the presence of antibodies. Anti-O55 antibodies were also able to inhibit aEPEC and STEC O55 adherence to human epithelial cells. In summary, the results demonstrated that the O55 polysaccharides have the potential to induce an effective humoral immune response against STEC and EPEC, indicating that they are good antigen targets to be used in vaccine formulations against these pathogens.

14.
Front Cell Infect Microbiol ; 12: 966370, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36081769

RESUMEN

Extracellular proteolytic enzymes are produced by a variety of pathogenic microorganisms, and contribute to host colonization by modulating virulence. Here, we present a first characterization of leptolysin, a Leptospira metalloprotease of the pappalysin family identified in a previous exoproteomic study. Comparative molecular analysis of leptolysin with two other pappalysins from prokaryotes, ulilysin and mirolysin, reveals similarities regarding calcium, zinc, and arginine -binding sites conservation within the catalytic domain, but also discloses peculiarities. Variations observed in the primary and tertiary structures may reflect differences in primary specificities. Purified recombinant leptolysin of L. interrogans was obtained as a ~50 kDa protein. The protease exhibited maximal activity at pH 8.0 and 37°C, and hydrolytic activity was observed in the presence of different salts with maximum efficiency in NaCl. Substrate specificity was assessed using a small number of FRET peptides, and showed a marked preference for arginine residues at the P1 position. L. interrogans leptolysin proteolytic activity on proteinaceous substrates such as proteoglycans and plasma fibronectin was also evaluated. All proteins tested were efficiently degraded over time, confirming the protease´s broad-spectrum activity in vitro. In addition, leptolysin induced morphological alterations on HK-2 cells, which may be partially attributed to extracellular matrix (ECM) degradation. Hemorrhagic foci were observed in the dorsal skin of mice intradermally injected with leptolysin, as a plausible consequence of ECM disarray and vascular endothelium glycocalyx damage. Assuming that leptospiral proteases play an important role in all stages of the infectious process, characterizing their functional properties, substrates and mechanisms of action is of great importance for therapeutic purposes.


Asunto(s)
Leptospira , Metaloproteasas , Animales , Arginina/metabolismo , Leptospira/química , Leptospira/metabolismo , Leptospirosis , Metaloproteasas/metabolismo , Metaloproteasas/farmacología , Ratones , Péptido Hidrolasas/metabolismo
15.
FEBS Lett ; 594(16): 2633-2644, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32153015

RESUMEN

Leptospires are highly invasive spirochetes equipped with efficient strategies for dissemination in the host. The Leptospira genus currently comprises 64 species divided into two major clades: the saprophytes composed of nonpathogenic, free-living organisms, and the pathogens encompassing all the species that cause mild or severe infections in humans and animals. While saprophytes are highly susceptible to the lytic action of the complement system, pathogenic (virulent) strains have evolved virulence strategies that allow efficient colonization of a variety of hosts and target organs, including mechanisms to circumvent hosts' innate and acquired immune responses. Pathogenic Leptospira avoid complement-mediated killing by recruiting host complement regulatory proteins and by targeting complement proteins using own and host-expressed proteases. This review outlines the role of complement in eradicating saprophytic Leptospira and the stratagems adopted by pathogenic Leptospira to maneuver the host complement system for their benefit.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Evasión Inmune , Leptospira , Leptospirosis/inmunología , Animales , Humanos , Leptospira/inmunología , Leptospira/patogenicidad , Leptospirosis/patología
16.
Methods Mol Biol ; 2134: 187-198, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32632870

RESUMEN

Like many other pathogens of medical importance, pathogenic Leptospira employ diverse strategies to circumvent Complement System activation. Under physiological conditions, this central humoral arm of innate immunity is tightly controlled by negative Complement regulatory proteins. However, upon infection, pathogenic microorganisms interfere with normal Complement host defense mechanisms by recruiting or mimicking Complement regulators and by secreting endogenous proteases or acquiring host's proteases that inactivate key Complement components. In this chapter, we describe in detail some of the most frequently used assays to evaluate Leptospira Complement resistance.


Asunto(s)
Bioensayo/métodos , Proteínas del Sistema Complemento/inmunología , Activación de Complemento/inmunología , Humanos , Inmunidad Humoral/inmunología , Inmunidad Innata/inmunología , Leptospira/inmunología , Leptospirosis/inmunología , Leptospirosis/microbiología , Unión Proteica/inmunología
17.
Microbes Infect ; 22(10): 550-557, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32730816

RESUMEN

The Complement System (CS) plays an important role in the immune response against leptospirosis and can be activated by the Alternative and Lectin Pathways (Innate Immunity) and by the Classical Pathway (Acquired Immunity). Here we analyzed a broad range of nonpathogenic and pathogenic Leptospira strains considering their interaction with each CS pathway. We determined bacterial survival rate and CS protein deposition in the presence of purified proteins, specific component depleted sera and NHS treated with the chelating agents EDTA (inhibits all three activation pathways) or EGTA (inhibits the Classical and Lectin Pathways). We suggest that the Lectin and the Alternative Pathways have an important role to eliminate saprophytic leptospires since i) approximately 50% survival of both saprophytic strains was observed in the presence of MBL-deficient serum; ii) approximately 50% survival of Leptospira biflexa Patoc I was observed in the presence of NHS - EGTA and iii) C1q-depleted serum caused significant bacterial lysis. In all serovars investigated the deposition of C5-C9 proteins on saprophytic Leptospira strains was more pronounced when compared to pathogenic species confirming previous studies in the literature. No difference on C3 deposition was observed between nonpathogenic and pathogenic strains. In conclusion, Leptospira strains interact to different degrees with CS proteins, especially those necessary to form MAC, indicating that some strains and specific ligands could favor the binding of certain CS proteins.


Asunto(s)
Activación de Complemento , Leptospira/inmunología , Leptospirosis/inmunología , Proteínas del Sistema Complemento/inmunología , Humanos , Evasión Inmune , Leptospira/patogenicidad , Viabilidad Microbiana/inmunología
18.
Front Immunol ; 11: 572562, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240263

RESUMEN

Properdin (P) is a positive regulatory protein that stabilizes the C3 convertase and C5 convertase of the complement alternative pathway (AP). Several studies have suggested that properdin can bind directly to the surface of certain pathogens regardless of the presence of C3bBb. Saprophytic Leptospira are susceptible to complement-mediated killing, but the interaction of properdin with Leptospira spp. has not been evaluated so far. In this work, we demonstrate that properdin present in normal human serum, purified properdin, as well as properdin oligomers P2, P3, and P4, interact with Leptospira. Properdin can bind directly to the bacterial surface even in the absence of C3b. In line with our previous findings, AP activation was shown to be important for killing non-pathogenic L. biflexa, and properdin plays a key role in this process since this microorganism survives in P-depleted human serum and the addition of purified properdin to P-depleted human serum decreases the number of viable leptospires. A panel of pathogenic L.interrogans recombinant proteins was used to identify putative properdin targets. Lsa30, an outer membrane protein from L. interrogans, binds to unfractionated properdin and to a lesser extent to P2-P4 properdin oligomers. In conclusion, properdin plays an important role in limiting bacterial proliferation of non-pathogenic Leptospira species. Once bound to the leptospiral surface, this positive complement regulatory protein of the AP contributes to the formation of the C3 convertase on the leptospire surface even in the absence of prior addition of C3b.


Asunto(s)
Complemento C3b/metabolismo , Factor B del Complemento/metabolismo , Leptospira interrogans/fisiología , Leptospira/fisiología , Leptospirosis/metabolismo , Properdina/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Procesos de Crecimiento Celular , Vía Alternativa del Complemento , Citotoxicidad Inmunológica , Humanos , Leptospira/patogenicidad , Leptospira interrogans/patogenicidad , Leptospirosis/inmunología , Properdina/inmunología , Unión Proteica , Virulencia
19.
Infect Immun ; 76(6): 2642-50, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18391007

RESUMEN

LipL32 is the major leptospiral outer membrane lipoprotein expressed during infection and is the immunodominant antigen recognized during the humoral immune response to leptospirosis in humans. In this study, we investigated novel aspects of LipL32. In order to define the immunodominant domains(s) of the molecule, subfragments corresponding to the N-terminal, intermediate, and C-terminal portions of the LipL32 gene were cloned and the proteins were expressed and purified by metal affinity chromatography. Our immunoblot results indicate that the C-terminal and intermediate domains of LipL32 are recognized by sera of patients with laboratory-confirmed leptospirosis. An immunoglobulin M response was detected exclusively against the LipL32 C-terminal fragment in both the acute and convalescent phases of illness. We also evaluated the capacity of LipL32 to interact with extracellular matrix (ECM) components. Dose-dependent, specific binding of LipL32 to collagen type IV and plasma fibronectin was observed, and the binding capacity could be attributed to the C-terminal portion of this molecule. Both heparin and gelatin could inhibit LipL32 binding to fibronectin in a concentration-dependent manner, indicating that the 30-kDa heparin-binding and 45-kDa gelatin-binding domains of fibronectin are involved in this interaction. Taken together, our results provide evidence that the LipL32 C terminus is recognized early in the course of infection and is the domain responsible for mediating interaction with ECM proteins.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Colágeno Tipo IV/metabolismo , Fibronectinas/metabolismo , Epítopos Inmunodominantes/inmunología , Leptospira/inmunología , Lipoproteínas/inmunología , Lipoproteínas/metabolismo , Animales , Especificidad de Anticuerpos , Proteínas de la Membrana Bacteriana Externa/genética , Clonación Molecular , Colágeno Tipo IV/química , Femenino , Fibronectinas/química , Gelatina/farmacología , Heparina/farmacología , Humanos , Sueros Inmunes/inmunología , Epítopos Inmunodominantes/biosíntesis , Epítopos Inmunodominantes/genética , Leptospira/genética , Leptospira/metabolismo , Leptospirosis/sangre , Leptospirosis/inmunología , Lipoproteínas/genética , Ratones , Ratones Endogámicos BALB C , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología
20.
Biotechnol Rep (Amst) ; 19: e00266, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29992100

RESUMEN

Enzymes from the thermolysin family are crucial factors in the pathogenesis of several diseases caused by bacteria and are potential targets for therapeutic interventions. Thermolysin encoded by the gene LIC13322 of the causative agent of leptospirosis, Leptospira interrogans, was shown to cleave proteins from the Complement System. However, the production of this recombinant protein using traditional refolding processes with high levels of denaturing reagents for thermolysin inclusion bodies (TL-IBs) solubilization results in poor recovery and low proteolytic activity probably due to improper refolding of the protein. Based on the assumption that leptospiral proteases play a crucial role during infection, the aim of this work was to obtain a functional recombinant thermolysin for future studies on the role of these metalloproteases on leptospiral infection. The association of high hydrostatic pressure (HHP) and alkaline pH was utilized for thermolysin refolding. Incubation of a suspension of TL-IBs at HHP and a pH of 11.0 is non-denaturing but effective for thermolysin solubilization. Soluble protein does not reaggregate by dialysis to pH 8.0. A volumetric yield of 46 mg thermolysin/L of bacterial culture and a yield of near 100% in relation to the total thermolysin present in TL-IBs were obtained. SEC-purified thermolysin suffers fragmentation, likely due to autoproteolysis and presents proteolytic activity against complement C3 α-chain, possibly by a generation of a C3b-like molecule. The proteolytic activity of thermolysin against C3 was time and dose-dependent. The experience gained in this study shall help to establish efficient HHP-based processes for refolding of bioactive proteins from IBs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA