Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.511
Filtrar
1.
Front Immunol ; 15: 1390468, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726006

RESUMEN

Introduction: Relapsing fever (RF) remains a neglected human disease that is caused by a number of diverse pathogenic Borrelia (B.) species. Characterized by high cell densities in human blood, relapsing fever spirochetes have developed plentiful strategies to avoid recognition by the host defense mechanisms. In this scenario, spirochetal lipoproteins exhibiting multifunctional binding properties in the interaction with host-derived molecules are known to play a key role in adhesion, fibrinolysis and complement activation. Methods: Binding of CihC/FbpC orthologs to different human proteins and conversion of protein-bound plasminogen to proteolytic active plasmin were examined by ELISA. To analyze the inhibitory capacity of CihC/FbpC orthologs on complement activation, a microtiter-based approach was performed. Finally, AlphaFold predictions were utilized to identified the complement-interacting residues. Results and discussion: Here, we elucidate the binding properties of CihC/FbpC-orthologs from distinct RF spirochetes including B. parkeri, B. hermsii, B. turicatae, and B. recurrentis to human fibronectin, plasminogen, and complement component C1r. All CihC/FbpC-orthologs displayed similar binding properties to fibronectin, plasminogen, and C1r, respectively. Functional studies revealed a dose dependent binding of plasminogen to all borrelial proteins and conversion to active plasmin. The proteolytic activity of plasmin was almost completely abrogated by tranexamic acid, indicating that lysine residues are involved in the interaction with this serine protease. In addition, a strong inactivation capacity toward the classical pathway could be demonstrated for the wild-type CihC/FbpC-orthologs as well as for the C-terminal CihC fragment of B. recurrentis. Pre-incubation of human serum with borrelial molecules except CihC/FbpC variants lacking the C-terminal region protected serum-susceptible Borrelia cells from complement-mediated lysis. Utilizing AlphaFold2 predictions and existing crystal structures, we mapped the putative key residues involved in C1r binding on the CihC/FbpC orthologs attempting to explain the relatively small differences in C1r binding affinity despite the substitutions of key residues. Collectively, our data advance the understanding of the multiple binding properties of structural and functional highly similar molecules of relapsing fever spirochetes proposed to be involved in pathogenesis and virulence.


Asunto(s)
Proteínas Bacterianas , Borrelia , Fibrinólisis , Plasminógeno , Unión Proteica , Fiebre Recurrente , Humanos , Borrelia/inmunología , Borrelia/metabolismo , Fiebre Recurrente/microbiología , Fiebre Recurrente/inmunología , Fiebre Recurrente/metabolismo , Plasminógeno/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Activación de Complemento , Evasión Inmune , Adhesión Bacteriana , Interacciones Huésped-Patógeno/inmunología , Fibronectinas/metabolismo , Fibrinolisina/metabolismo , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo
2.
PLoS One ; 19(5): e0301477, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38768108

RESUMEN

Food allergy is widely recognized as a significant health issue, having escalated into a global epidemic, subsequently giving rise to the development of numerous additional complications. Currently, the sole efficient method to curb the progression of allergy is through the implementation of an elimination diet. The increasing number of newly identified allergens makes it harder to completely remove or avoid them effectively. The immunoreactivity of proteins of bacterial origin remains an unexplored topic. Despite the substantial consumption of microbial proteins in our diets, the immunologic mechanisms they might induce require thorough validation. This stands as the primary objective of this study. The primary objective of this study was to evaluate the effects of bacterial proteins on the intestinal barrier and immune system parameters during hypersensitivity induction in both developing and mature organisms. The secondary objective was to evaluate the role of lipids in the immunoreactivity programming of these bacterial proteins. Notably, in this complex, comprehensively designed in vitro, in vivo, and ex vivo trial, the immunoreactivity of various bacterial proteins will be examined. In summary, the proposed study intends to address the knowledge gaps regarding the effects of Lactobacillus microbial proteins on inflammation, apoptosis, autophagy, and intestinal barrier integrity in a single study.


Asunto(s)
Proteínas Bacterianas , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Lípidos , Leche/microbiología , Leche/inmunología , Ratones , Lactobacillales/metabolismo , Lactobacillales/inmunología , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/microbiología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología
3.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38771879

RESUMEN

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Asunto(s)
Proteínas Bacterianas , Macrófagos , Proteínas de la Membrana , Infecciones Estafilocócicas , Staphylococcus aureus , Sistemas de Secreción Tipo VII , Ubiquitinación , Staphylococcus aureus/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Animales , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Sistemas de Secreción Tipo VII/inmunología , Sistemas de Secreción Tipo VII/genética , Ratones , Evasión Inmune , Interacciones Huésped-Patógeno/inmunología
4.
Proc Natl Acad Sci U S A ; 121(19): e2318003121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38691588

RESUMEN

Peptides presented by HLA-E, a molecule with very limited polymorphism, represent attractive targets for T cell receptor (TCR)-based immunotherapies to circumvent the limitations imposed by the high polymorphism of classical HLA genes in the human population. Here, we describe a TCR-based bispecific molecule that potently and selectively binds HLA-E in complex with a peptide encoded by the inhA gene of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis in humans. We reveal the biophysical and structural bases underpinning the potency and specificity of this molecule and demonstrate its ability to redirect polyclonal T cells to target HLA-E-expressing cells transduced with mycobacterial inhA as well as primary cells infected with virulent Mtb. Additionally, we demonstrate elimination of Mtb-infected cells and reduction of intracellular Mtb growth. Our study suggests an approach to enhance host T cell immunity against Mtb and provides proof of principle for an innovative TCR-based therapeutic strategy overcoming HLA polymorphism and therefore applicable to a broader patient population.


Asunto(s)
Antígenos de Histocompatibilidad Clase I , Mycobacterium tuberculosis , Receptores de Antígenos de Linfocitos T , Linfocitos T , Mycobacterium tuberculosis/inmunología , Humanos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Linfocitos T/inmunología , Antígenos HLA-E , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Tuberculosis/inmunología
5.
Nat Commun ; 15(1): 3954, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729958

RESUMEN

Defense-associated sirtuin 2 (DSR2) systems are widely distributed across prokaryotic genomes, providing robust protection against phage infection. DSR2 recognizes phage tail tube proteins and induces abortive infection by depleting intracellular NAD+, a process that is counteracted by another phage-encoded protein, DSR Anti Defense 1 (DSAD1). Here, we present cryo-EM structures of Bacillus subtilis DSR2 in its apo, Tube-bound, and DSAD1-bound states. DSR2 assembles into an elongated tetramer, with four NADase catalytic modules clustered in the center and the regulatory-sensing modules distributed at four distal corners. Interestingly, monomeric Tube protein, rather than its oligomeric states, docks at each corner of the DSR2 tetramer to form a 4:4 DSR2-Tube assembly, which is essential for DSR2 NADase activity. DSAD1 competes with Tube for binding to DSR2 by occupying an overlapping region, thereby inhibiting DSR2 immunity. Thus, our results provide important insights into the assembly, activation and inhibition of the DSR2 anti-phage defense system.


Asunto(s)
Bacillus subtilis , Proteínas Bacterianas , Bacteriófagos , Microscopía por Crioelectrón , Bacillus subtilis/inmunología , Bacillus subtilis/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Bacteriófagos/genética , Bacteriófagos/inmunología , Evasión Inmune , Sirtuinas/metabolismo , Sirtuinas/genética , Proteínas Virales/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/química , Proteínas Virales/genética , Unión Proteica , Modelos Moleculares , NAD/metabolismo
6.
Biochem Biophys Res Commun ; 717: 150040, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38718566

RESUMEN

Mtb12, a small protein secreted by Mycobacterium tuberculosis, is known to elicit immune responses in individuals infected with the pathogen. It serves as an antigen recognized by the host's immune system. Due to its immunogenic properties and pivotal role in tuberculosis (TB) pathogenesis, Mtb12 is considered a promising candidate for TB diagnosis and vaccine development. However, the structural and functional properties of Mtb12 are largely unexplored, representing a significant gap in our understanding of M. tuberculosis biology. In this study, we present the first structure of Mtb12, which features a unique tertiary configuration consisting of four beta strands and four alpha helices. Structural analysis reveals that Mtb12 has a surface adorned with a negatively charged pocket adjacent to a central cavity. The features of these structural elements and their potential effects on the function of Mtb12 warrant further exploration. These findings offer valuable insights for vaccine design and the development of diagnostic tools.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Mycobacterium tuberculosis , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Modelos Moleculares , Peso Molecular , Secuencia de Aminoácidos , Conformación Proteica , Humanos
7.
Front Immunol ; 15: 1392316, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711516

RESUMEN

Streptococcus pneumoniae remains a significant global threat, with existing vaccines having important limitations such as restricted serotype coverage and high manufacturing costs. Pneumococcal lipoproteins are emerging as promising vaccine candidates due to their surface exposure and conservation across various serotypes. While prior studies have explored their potential in mice, data in a human context and insights into the impact of the lipid moiety remain limited. In the present study, we examined the immunogenicity of two pneumococcal lipoproteins, DacB and MetQ, both in lipidated and non-lipidated versions, by stimulation of primary human immune cells. Immune responses were assessed by the expression of common surface markers for activation and maturation as well as cytokines released into the supernatant. Our findings indicate that in the case of MetQ lipidation was crucial for activation of human antigen-presenting cells such as dendritic cells and macrophages, while non-lipidated DacB demonstrated an intrinsic potential to induce an innate immune response. Nevertheless, immune responses to both proteins were enhanced by lipidation. Interestingly, following stimulation of dendritic cells with DacB, LipDacB and LipMetQ, cytokine levels of IL-6 and IL-23 were significantly increased, which are implicated in triggering potentially important Th17 cell responses. Furthermore, LipDacB and LipMetQ were able to induce proliferation of CD4+ T cells indicating their potential to induce an adaptive immune response. These findings contribute valuable insights into the immunogenic properties of pneumococcal lipoproteins, emphasizing their potential role in vaccine development against pneumococcal infections.


Asunto(s)
Inmunidad Adaptativa , Proteínas Bacterianas , Citocinas , Streptococcus pneumoniae , Humanos , Streptococcus pneumoniae/inmunología , Citocinas/metabolismo , Proteínas Bacterianas/inmunología , Lipoproteínas/inmunología , Lipoproteínas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Vacunas Neumococicas/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/prevención & control , Macrófagos/inmunología , Macrófagos/metabolismo , Células Cultivadas
8.
Front Immunol ; 15: 1372349, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698863

RESUMEN

Pseudomonas aeruginosa (Pa) is an opportunistic bacterial pathogen responsible for severe hospital acquired infections in immunocompromised and elderly individuals. Emergence of increasingly drug resistant strains and the absence of a broad-spectrum prophylactic vaccine against both T3SA+ (type III secretion apparatus) and ExlA+/T3SA- Pa strains worsen the situation in a post-pandemic world. Thus, we formulated a candidate subunit vaccine (called ExlA/L-PaF/BECC/ME) against both Pa types. This bivalent vaccine was generated by combining the C-terminal active moiety of exolysin A (ExlA) produced by non-T3SA Pa strains with our T3SA-based vaccine platform, L-PaF, in an oil-in-water emulsion. The ExlA/L-PaF in ME (MedImmune emulsion) was then mixed with BECC438b, an engineered lipid A analogue and a TLR4 agonist. This formulation was administered intranasally (IN) to young and elderly mice to determine its potency across a diverse age-range. The elderly mice were used to mimic the infection seen in elderly humans, who are more susceptible to serious Pa disease compared to their young adult counterparts. After Pa infection, mice immunized with ExlA/L-PaF/BECC/ME displayed a T cell-mediated adaptive response while PBS-vaccinated mice experienced a rapid onset inflammatory response. Important genes and pathways were observed, which give rise to an anti-Pa immune response. Thus, this vaccine has the potential to protect aged individuals in our population from serious Pa infection.


Asunto(s)
Emulsiones , Infecciones por Pseudomonas , Vacunas contra la Infección por Pseudomonas , Pseudomonas aeruginosa , Vacunas de Subunidad , Animales , Pseudomonas aeruginosa/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Ratones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas/inmunología , Vacunas contra la Infección por Pseudomonas/administración & dosificación , Femenino , Desarrollo de Vacunas , Humanos , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/sangre , Modelos Animales de Enfermedad , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética
9.
Cell Mol Biol Lett ; 29(1): 70, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741147

RESUMEN

BACKGROUND: Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) is a peptide antigen released from the mycobacterial cytoplasm into the supernatant of Mycobacterium tuberculosis (Mtb) attenuated H37Ra strain after autoclaving at 121 °C for 20 min. Mtb-HAg can specifically induce γδ T-cell proliferation in vitro. However, the exact composition of Mtb-HAg and the protein antigens that are responsible for its function are currently unknown. METHODS: Mtb-HAg extracted from the Mtb H37Ra strain was subjected to LC‒MS mass spectrometry. Twelve of the identified protein fractions were recombinantly expressed in Escherichia coli by genetic engineering technology using pET-28a as a plasmid and purified by Ni-NTA agarose resin to stimulate peripheral blood mononuclear cells (PBMCs) from different healthy individuals. The proliferation of γδ T cells and major γδ T-cell subset types as well as the production of TNF-α and IFN-γ were determined by flow cytometry. Their proliferating γδ T cells were isolated and purified using MACS separation columns, and Mtb H37Ra-infected THP-1 was co-cultured with isolated and purified γδ T cells to quantify Mycobacterium viability by counting CFUs. RESULTS: In this study, Mtb-HAg from the attenuated Mtb H37Ra strain was analysed by LC‒MS mass spectrometry, and a total of 564 proteins were identified. Analysis of the identified protein fractions revealed that the major protein components included heat shock proteins and Mtb-specific antigenic proteins. Recombinant expression of 10 of these proteins in by Escherichia coli genetic engineering technology was used to successfully stimulate PBMCs from different healthy individuals, but 2 of the proteins, EsxJ and EsxA, were not expressed. Flow cytometry results showed that, compared with the IL-2 control, HspX, GroEL1, and GroES specifically induced γδ T-cell expansion, with Vγ2δ2 T cells as the main subset, and the secretion of the antimicrobial cytokines TNF-α and IFN-γ. In contrast, HtpG, DnaK, GroEL2, HbhA, Mpt63, EsxB, and EsxN were unable to promote γδ T-cell proliferation and the secretion of TNF-α and IFN-γ. None of the above recombinant proteins were able to induce the secretion of TNF-α and IFN-γ by αß T cells. In addition, TNF-α, IFN-γ-producing γδ T cells inhibit the growth of intracellular Mtb. CONCLUSION: Activated γδ T cells induced by Mtb-HAg components HspX, GroES, GroEL1 to produce TNF-α, IFN-γ modulate macrophages to inhibit intracellular Mtb growth. These data lay the foundation for subsequent studies on the mechanism by which Mtb-HAg induces γδ T-cell proliferation in vitro, as well as the development of preventive and therapeutic vaccines and rapid diagnostic reagents.


Asunto(s)
Antígenos Bacterianos , Proliferación Celular , Mycobacterium tuberculosis , Linfocitos T , Humanos , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Interferón gamma/metabolismo , Interferón gamma/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Factor de Necrosis Tumoral alfa/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología
10.
Sci Rep ; 14(1): 9141, 2024 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-38644371

RESUMEN

Tuberculosis remains a large health threat, despite the availability of the tuberculosis vaccine, BCG. As BCG efficacy gradually decreases from adolescence, BCG-Prime and antigen-booster may be an efficient strategy to confer vaccine efficacy. Mycobacterial DNA-binding protein 1 (MDP1, namely Rv2986c, hupB or HU) is a major Mycobacterium tuberculosis protein that induces vaccine-efficacy by co-administration with CpG DNA. To produce MDP1 for booster-vaccine use, we have created recombinant MDP1 produced in both Escherichia coli (eMDP1) and Mycolicibacterium smegmatis (mMDP1), an avirulent rapid-growing mycobacteria. We tested their immunogenicity by checking interferon (IFN)-gamma production by stimulated peripheral blood cells derived from BCG-vaccinated individuals. Similar to native M. tuberculosis MDP1, we observed that most lysin resides in the C-terminal half of mMDP1 are highly methylated. In contrast, eMDP1 had less post-translational modifications and IFN-gamma stimulation. mMDP1 stimulated the highest amount of IFN-gamma production among the examined native M. tuberculosis proteins including immunodominant MPT32 and Antigen 85 complex. MDP1-mediated IFN-gamma production was more strongly enhanced when combined with a new type of CpG DNA G9.1 than any other tested CpG DNAs. Taken together, these results suggest that the combination of mMDP1 and G9.1 possess high potential use for human booster vaccine against tuberculosis.


Asunto(s)
Vacuna BCG , Proteínas Bacterianas , Proteínas de Unión al ADN , Interferón gamma , Mycobacterium tuberculosis , Procesamiento Proteico-Postraduccional , Humanos , Interferón gamma/metabolismo , Proteínas Bacterianas/inmunología , Vacuna BCG/inmunología , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Mycobacterium tuberculosis/inmunología , Proteínas Recombinantes/inmunología , Oligodesoxirribonucleótidos/farmacología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Islas de CpG , Mycobacterium smegmatis/inmunología , Mycobacterium smegmatis/metabolismo , Escherichia coli/metabolismo , Escherichia coli/genética , Femenino
11.
Nat Microbiol ; 9(5): 1176-1188, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38684911

RESUMEN

Matching donor and recipient blood groups based on red blood cell (RBC) surface ABO glycans and antibodies in plasma is crucial to avoid potentially fatal reactions during transfusions. Enzymatic conversion of RBC glycans to the universal group O is an attractive solution to simplify blood logistics and prevent ABO-mismatched transfusions. The gut symbiont Akkermansia muciniphila can degrade mucin O-glycans including ABO epitopes. Here we biochemically evaluated 23 Akkermansia glycosyl hydrolases and identified exoglycosidase combinations which efficiently transformed both A and B antigens and four of their carbohydrate extensions. Enzymatic removal of canonical and extended ABO antigens on RBCs significantly improved compatibility with group O plasmas, compared to conversion of A or B antigens alone. Finally, structural analyses of two B-converting enzymes identified a previously unknown putative carbohydrate-binding module. This study demonstrates the potential utility of mucin-degrading gut bacteria as valuable sources of enzymes for production of universal blood for transfusions.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Akkermansia , Glicósido Hidrolasas , Sistema del Grupo Sanguíneo ABO/inmunología , Humanos , Glicósido Hidrolasas/metabolismo , Mucinas/metabolismo , Eritrocitos/inmunología , Polisacáridos/metabolismo , Microbioma Gastrointestinal , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología
12.
Immunol Cell Biol ; 102(5): 365-380, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572664

RESUMEN

Staphylococcus aureus is a significant bacterial pathogen in both community and hospital settings, and the escalation of antimicrobial-resistant strains is of immense global concern. Vaccination is an inviting long-term strategy to curb staphylococcal disease, but identification of an effective vaccine has proved to be challenging. Three well-characterized, ubiquitous, secreted immune evasion factors from the staphylococcal superantigen-like (SSL) protein family were selected for the development of a vaccine. Wild-type SSL3, 7 and 11, which inhibit signaling through Toll-like receptor 2, cleavage of complement component 5 and neutrophil function, respectively, were successfully combined into a stable, active fusion protein (PolySSL7311). Vaccination of mice with an attenuated form of the PolySSL7311 protein stimulated significantly elevated specific immunoglobulin G and splenocyte proliferation responses to each component relative to adjuvant-only controls. Vaccination with PolySSL7311, but not a mixture of the individual proteins, led to a > 102 reduction in S. aureus tissue burden compared with controls after peritoneal challenge. Comparable antibody responses were elicited after coadministration of the vaccine in either AddaVax (an analog of MF59) or an Alum-based adjuvant; but only AddaVax conferred a significant reduction in bacterial load, aligning with other studies that suggest both cellular and humoral immune responses are necessary for protective immunity to S. aureus. Anti-sera from mice immunized with PolySSL7311, but not individual proteins, partially neutralized the functional activities of SSL7. This study confirms the importance of these SSLs for the survival of S. aureus in vivo and suggests that PolySSL7311 is a promising vaccine candidate.


Asunto(s)
Proteínas Bacterianas , Infecciones Estafilocócicas , Vacunas Estafilocócicas , Staphylococcus aureus , Superantígenos , Animales , Staphylococcus aureus/inmunología , Vacunas Estafilocócicas/inmunología , Superantígenos/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/prevención & control , Ratones , Proteínas Bacterianas/inmunología , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/sangre , Femenino , Proteínas Recombinantes de Fusión/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Estudios de Factibilidad , Vacunación , Antígenos Bacterianos/inmunología , Ratones Endogámicos BALB C , Adyuvantes Inmunológicos
13.
Clin Immunol ; 263: 110226, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663493

RESUMEN

Antibiotic resistance and the surge of infectious diseases during the pandemic present significant threats to human health. Trained immunity emerges as a promising and innovative approach to address these infections. Synthetic or natural fungal, parasitic and viral components have been reported to induce trained immunity. However, it is not clear whether bacterial virulence proteins can induce protective trained immunity. Our research demonstrates Streptococcus pneumoniae virulence protein PepO, is a highly potent trained immunity inducer for combating broad-spectrum infection. Our findings showcase that rPepO training confers robust protection to mice against various pathogenic infections by enhancing macrophage functionality. rPepO effectively re-programs macrophages, re-configures their epigenetic modifications and bolsters their immunological responses, which is independent of T or B lymphocytes. In vivo and in vitro experiments confirm that trained macrophage-secreted complement C3 activates peritoneal B lymphocyte and enhances its bactericidal capacity. In addition, we provide the first evidence that granulocyte colony-stimulating factor (G-CSF) derived from trained macrophages plays a pivotal role in shaping central-trained immunity. In summation, our research demonstrates the capability of rPepO to induce both peripheral and central trained immunity in mice, underscoring its potential application in broad-spectrum anti-infection therapy. Our research provides a new molecule and some new target options for infectious disease prevention.


Asunto(s)
Macrófagos , Ratones Endogámicos C57BL , Streptococcus pneumoniae , Animales , Streptococcus pneumoniae/inmunología , Ratones , Macrófagos/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/prevención & control , Proteínas Bacterianas/inmunología , Linfocitos B/inmunología , Femenino , Inmunidad Entrenada
14.
Virulence ; 15(1): 2345019, 2024 12.
Artículo en Inglés | MEDLINE | ID: mdl-38656137

RESUMEN

Klebsiella pneumoniae is an important gram-negative bacterium that causes severe respiratory and healthcare-associated infections. Although antibiotic therapy is applied to treat severe infections caused by K. pneumoniae, drug-resistant isolates pose a huge challenge to clinical practices owing to adverse reactions and the mismanagement of antibiotics. Several studies have attempted to develop vaccines against K. pneumoniae, but there are no licensed vaccines available for the control of K. pneumoniae infection. In the current study, we constructed a novel DNA vaccine, pVAX1-YidR, which encodes a highly conserved virulence factor YidR and a recombinant expression plasmid pVAX1-IL-17 encoding Interleukin-17 (IL-17) as a molecular adjuvant. Adaptive immune responses were assessed in immunized mice to compare the immunogenicity of the different vaccine schemes. The results showed that the targeted antigen gene was expressed in HEK293T cells using an immunofluorescence assay. Mice immunized with pVAX1-YidR elicited a high level of antibodies, induced strong cellular immune responses, and protected mice from K. pneumoniae challenge. Notably, co-immunization with pVAX1-YidR and pVAX1-IL-17 significantly augmented host adaptive immune responses and provided better protection against K. pneumoniae infections in vaccinated mice. Our study demonstrates that combined DNA vaccines and molecular adjuvants is a promising strategy to develop efficacious antibacterial vaccines against K. pneumoniae infections.


Asunto(s)
Vacunas Bacterianas , Interleucina-17 , Infecciones por Klebsiella , Klebsiella pneumoniae , Vacunas de ADN , Animales , Femenino , Humanos , Ratones , Inmunidad Adaptativa , Adyuvantes Inmunológicos/administración & dosificación , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/administración & dosificación , Modelos Animales de Enfermedad , Células HEK293 , Inmunidad Celular , Inmunización , Interleucina-17/inmunología , Interleucina-17/genética , Infecciones por Klebsiella/prevención & control , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Klebsiella pneumoniae/genética , Ratones Endogámicos BALB C , Vacunas de ADN/inmunología , Vacunas de ADN/genética , Vacunas de ADN/administración & dosificación , Factores de Virulencia/inmunología , Factores de Virulencia/genética
15.
J Immunol Methods ; 529: 113669, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582259

RESUMEN

Because of their superior properties for certain biological applications small antibody derivatives like fragment of antigen binding (Fab) have found widespread use in basic research and as therapeutics. However, generation of Fab-fragments is still a rather complex matter, reflected by the fact that a variety of methods and purification techniques are necessary for the production of all the different classes of Fab-fragments (kappa/lambda light chains, type of species). Here we demonstrate that Fab-fragments derived from six different antibodies of human or murine origin produced by transient expression in HEK cells can be purified in a single step to a high degree of purity by standard protein G affinity chromatography. This is most likely due to alternative contact sites for protein G located in the CH1 domain of the Fab heavy chain. Our data demonstrate that protein G affinity chromatography as for whole antibodies is a robust method for the purification of tag-less Fab-fragments independent of species, significantly simplifying the process of Fab-fragment purification.


Asunto(s)
Cromatografía de Afinidad , Fragmentos Fab de Inmunoglobulinas , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Cromatografía de Afinidad/métodos , Humanos , Animales , Ratones , Células HEK293 , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/química
16.
Fish Shellfish Immunol ; 149: 109572, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636739

RESUMEN

Streptococcosis outbreaks caused by Streptococcus agalactiae infection in tilapia aquaculture have been consistently reported and associated with high mortality and morbidity leading to significant economic losses. Existing vaccine candidates against Streptococcus spp. are designed for intraperitoneal injections that are not practical and labor-intensive which have prompted farmers to protect aquatic animals with antibiotics, thus encouraging the emergence of multidrug resistant bacteria. In this study, a live recombinant L. lactis vaccine expressing a 1403 bp surface immunogenic protein (SIP) and a 1100 bp truncated SIP (tSIP) gene was developed and evaluated against S. agalactiae infection in tilapia. Both SIP and tSIP sequences were cloned and transformed into L. lactis. The recombinant L.lactis vaccine was orally administered to juvenile tilapia for a month. Detection of SIP-specific serum IgM in vaccinated groups compared to control groups indicated that recombinant proteins expressed from L. lactis could elicit immunogenic reactions in tilapia. Fish immunized with the tSIP vaccine also showed the highest level of protection compared to other test groups, and the mortality rate was significantly reduced compared to both control groups. The relative percentage of survival (RPS) against S. agalactiae for both SIP and tSIP-vaccinated groups was 50 % and 89 %, respectively, at 14 days post-challenge. Significant up-regulation of IgM, IL-1ß, IL-10, TNF-α and IFN-γ were observed at day 34 between the vaccinated and control groups. These results indicated that the recombinant lactococcal tSIP vaccine can elicit both cell-mediated and humoral responses and is recommended as a potential oral vaccine against S. agalactiae infection. Future work will include further in vivo challenge assessments of this vaccine candidate fused with adjuvants to boost immunogenicity levels in tilapia.


Asunto(s)
Cíclidos , Enfermedades de los Peces , Infecciones Estreptocócicas , Streptococcus agalactiae , Animales , Streptococcus agalactiae/inmunología , Infecciones Estreptocócicas/veterinaria , Infecciones Estreptocócicas/prevención & control , Infecciones Estreptocócicas/inmunología , Enfermedades de los Peces/prevención & control , Enfermedades de los Peces/inmunología , Cíclidos/inmunología , Administración Oral , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Estreptocócicas/inmunología , Vacunas Estreptocócicas/administración & dosificación , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Lactococcus lactis/genética , Lactococcus lactis/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética
17.
Cytokine ; 179: 156622, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38648681

RESUMEN

Tuberculosis is caused by Mycobacterium tuberculosis (M tb), which is recognized by macrophages and produces inflammatory cytokines, and chemokines at the site of infection. The present study was proposed to understand the interaction of M tb antigens, cytokines, and chemokines. We have evaluated the chemokine MCP-1 levels and its expression in PBMCs stimulated with M tb antigens Ag85A, ESAT6 and recombinant cytokines rhTNF-α, rhIFN-γ, rhTGF-ß, and rhIL-10 in active pulmonary TB (APTB) patients, household contacts (HHC) at 0 months, 6 months and healthy controls (HC). We have observed low levels of MCP-1 with Ag85A, ESAT6, and rhTNF-α stimulations in APTB 0M compared to HHC and HC (p < 0.0067, p < 0.0001, p < 0.01, p < 0.005, p < 0.0065, p < 0.0001) and significantly increased after treatment with rhTNF-α. The MCP-1 levels with rhIFN-γ were high in APTB, HHC at 0 M and significant between APTB 0 M vs. 6 M, HHC vs. HC, and HHC 0M vs. 6M (p < 0.0352, p < 0.0252, p < 0.00062). The rhTGF-ß, rhIL-10 induced high MCP-1 levels in APTB, HHC compared to HC (p < 0.0414, p < 0.0312, p < 0.004, p < 0.0001) and significantly decreased after treatment with rhIL-10 (p < 0.0001). The MCP-1 expression was low with all the stimulations in APTB 0M when compared to HC and after treatment. Whereas, HHC shown low MCP-1 expression with rhTNF-α, rhIFN-γ and Ag85A and high with rhTGF-ß, rhIL-10 and ESAT6. In conclusion, the study determined the differential expression and production of MCP-1 with M tb antigens and recombinant cytokines. Further, cohort studies are required to study these interaction to identify the high risk individuals, which might help for TB control.


Asunto(s)
Antígenos Bacterianos , Quimiocina CCL2 , Citocinas , Mycobacterium tuberculosis , Proteínas Recombinantes , Humanos , Antígenos Bacterianos/inmunología , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Masculino , Mycobacterium tuberculosis/inmunología , Femenino , Proteínas Recombinantes/inmunología , Adulto , Citocinas/metabolismo , Proteínas Bacterianas/inmunología , Persona de Mediana Edad , Interferón gamma/inmunología , Interferón gamma/metabolismo , Tuberculosis Pulmonar/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-10/metabolismo , Interleucina-10/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Tuberculosis/inmunología , Factor de Crecimiento Transformador beta/inmunología
18.
Mem Inst Oswaldo Cruz ; 119: e230040, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38655925

RESUMEN

BACKGROUND: The availability of genes and protein sequences for parasites has provided valuable information for drug target identification and vaccine development. One such parasite is Bartonella quintana, a Gram-negative, intracellular pathogen that causes bartonellosis in mammalian hosts. OBJECTIVE: Despite progress in understanding its pathogenesis, limited knowledge exists about the virulence factors and regulatory mechanisms specific to B. quintana. METHODS AND FINDINGS: To explore these aspects, we have adopted a subtractive proteomics approach to analyse the proteome of B. quintana. By subtractive proteins between the host and parasite proteome, a set of proteins that are likely unique to the parasite but absent in the host were identified. This analysis revealed that out of the 1197 protein sequences of the parasite, 660 proteins are non-homologous to the human host. Further analysis using the Database of Essential Genes predicted 159 essential proteins, with 28 of these being unique to the pathogen and predicted as potential putative targets. Subcellular localisation of the predicted targets revealed 13 cytoplasmic, eight membranes, one periplasmic, and multiple location proteins. The three-dimensional structure and B cell epitopes of the six membrane antigenic protein were predicted. Four B cell epitopes in KdtA and mraY proteins, three in lpxB and BQ09550, whereas the ftsl and yidC proteins were located with eleven and six B cell epitopes, respectively. MAINS CONCLUSIONS: This insight prioritises such proteins as novel putative targets for further investigations on their potential as drug and vaccine candidates.


Asunto(s)
Vacunas Bacterianas , Bartonella quintana , Proteómica , Bartonella quintana/inmunología , Bartonella quintana/genética , Vacunas Bacterianas/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Humanos , Simulación por Computador , Factores de Virulencia/inmunología , Factores de Virulencia/genética , Proteoma
19.
Front Immunol ; 15: 1330796, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665909

RESUMEN

Introduction: There is no useful method to discriminate between latent tuberculosis infection (LTBI) and active pulmonary tuberculosis (PTB). This study aimed to investigate the potential of cytokine profiles to discriminate between LTBI and active PTB using whole-blood stimulation with Mycobacterium tuberculosis (MTB) antigens, including latency-associated antigens. Materials and methods: Patients with active PTB, household contacts of active PTB patients and community exposure subjects were recruited in Manila, the Philippines. Peripheral blood was collected from the participants and used for whole-blood stimulation (WBS) with either the early secretory antigenic target and the 10-kDa culture filtrate protein (ESAT-6/CFP-10), Rv3879c or latency-associated MTB antigens, including mycobacterial DNA-binding protein 1 (MDP-1), α-crystallin (Acr) and heparin-binding hemagglutinin (HBHA). Multiple cytokine concentrations were analyzed using the Bio-Plex™ multiplex cytokine assay. Results: A total of 78 participants consisting of 15 active PTB patients, 48 household contacts and 15 community exposure subjects were eligible. The MDP-1-specific IFN-γ level in the active PTB group was significantly lower than that in the household contact group (p < 0.001) and the community exposure group (p < 0.001). The Acr-specific TNF-α and IL-10 levels in the active PTB group were significantly higher than those in the household contact (TNF-α; p = 0.001, IL-10; p = 0.001) and community exposure (TNF-α; p < 0.001, IL-10; p = 0.01) groups. However, there was no significant difference in the ESAT-6/CFP-10-specific IFN-γ levels among the groups. Conclusion: The patterns of cytokine profiles induced by latency-associated MTB antigens using WBS have the potential to discriminate between LTBI and active PTB. In particular, combinations of IFN-γ and MDP-1, TNF-α and Acr, and IL-10 and Acr are promising. This study provides the first demonstration of the utility of MDP-1-specific cytokine responses in WBS.


Asunto(s)
Antígenos Bacterianos , Citocinas , Tuberculosis Latente , Mycobacterium tuberculosis , Tuberculosis Pulmonar , Humanos , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/sangre , Masculino , Tuberculosis Latente/diagnóstico , Tuberculosis Latente/inmunología , Tuberculosis Latente/sangre , Tuberculosis Latente/microbiología , Femenino , Mycobacterium tuberculosis/inmunología , Filipinas , Adulto , Citocinas/sangre , Persona de Mediana Edad , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/sangre , Tuberculosis Pulmonar/diagnóstico , Tuberculosis Pulmonar/microbiología , Adulto Joven , Proteínas Bacterianas/inmunología
20.
Biomolecules ; 14(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38672487

RESUMEN

Tuberculosis (TB) is the leading global cause of death f rom an infectious bacterial agent. Therefore, limiting its epidemic spread is a pressing global health priority. The chaperone-like protein HtpG of M. tuberculosis (Mtb) is a large dimeric and multi-domain protein with a key role in Mtb pathogenesis and promising antigenic properties. This dual role, likely associated with the ability of Heat Shock proteins to act both intra- and extra-cellularly, makes HtpG highly exploitable both for drug and vaccine development. This review aims to gather the latest updates in HtpG structure and biological function, with HtpG operating in conjunction with a large number of chaperone molecules of Mtb. Altogether, these molecules help Mtb recovery after exposure to host-like stress by assisting the whole path of protein folding rescue, from the solubilisation of aggregated proteins to their refolding. Also, we highlight the role of structural biology in the development of safer and more effective subunit antigens. The larger availability of structural information on Mtb antigens and a better understanding of the host immune response to TB infection will aid the acceleration of TB vaccine development.


Asunto(s)
Antígenos Bacterianos , Proteínas Bacterianas , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Factores de Virulencia , Mycobacterium tuberculosis/inmunología , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/química , Factores de Virulencia/inmunología , Factores de Virulencia/química , Humanos , Vacunas contra la Tuberculosis/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/química , Tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis/microbiología , Animales , Chaperonas Moleculares/inmunología , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA