Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.058
Filtrar
1.
Nat Commun ; 15(1): 8663, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375351

RESUMEN

Listeria monocytogenes (LM) possesses the ability to breach multiple barriers and elicit intricate immune responses. However, there remains a lack of explicit understanding regarding how LM evades innate immune surveillance within the body. Here, we utilized liver intravital imaging to elucidate the dynamic process of LM during infection in the liver. We discovered that LM can rapidly escape from Kupffer cells (KCs) through listeriolysin O (LLO) and proliferate within hepatocytes. Upon LM exposure to the hepatic sinusoids, neutrophils rapidly aggregate at the site of infection. Subsequently, LM can induce type I interferon (IFN-I) production primarily in the spleen, which acts systemically on neutrophils to hamper their swarming by deactivating the ERK pathway, thus evading neutrophil-mediated eradication. Furthermore, our findings suggest that virus-induced IFN-I suppresses neutrophil swarming, and COVID-19 patients exhibit impaired neutrophil aggregation function. In conclusion, our findings provide compelling evidence demonstrating that intracellular bacteria represented by LM can hijack host defense mechanisms against viral infections to evade immune surveillance. Additionally, impaired neutrophil swarming caused by IFN-I is one of the significant factors contributing to the increased susceptibility to bacterial infections following viral infections.


Asunto(s)
COVID-19 , Interferón Tipo I , Macrófagos del Hígado , Listeria monocytogenes , Listeriosis , Neutrófilos , Animales , Femenino , Humanos , Masculino , Ratones , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , COVID-19/inmunología , COVID-19/virología , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Hepatocitos/virología , Hepatocitos/inmunología , Evasión Inmune , Inmunidad Innata , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Macrófagos del Hígado/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Listeriosis/microbiología , Hígado/inmunología , Hígado/virología , Hígado/microbiología , Sistema de Señalización de MAP Quinasas/inmunología , Ratones Endogámicos C57BL , Neutrófilos/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Bazo/inmunología
2.
Nat Commun ; 15(1): 8702, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379345

RESUMEN

Staphylococcus aureus remains a leading global cause of bacterial infection-associated mortality and has eluded prior vaccine development efforts. S. aureus α-toxin (Hla) is an essential virulence factor in disease, impairing the T cell response to infection. The anti-Hla antibody response is a correlate of human protective immunity. Here we observe that this response is limited early in human life and design a vaccine strategy to elicit immune protection against Hla in a neonatal mice. By targeted disruption of the interaction of Hla with its receptor ADAM10, we identify a vaccine antigen (HlaH35L/R66C/E70C, HlaHRE) that elicits an ~100-fold increase in the neutralizing anti-Hla response. Immunization with HlaHRE enhances the T follicular helper (TFH) cell response to S. aureus infection, correlating with the magnitude of the neutralizing anti-toxin response and disease protection. Furthermore, maternal HlaHRE immunization confers protection to offspring. Together, these findings illuminate a path for S. aureus vaccine development at the maternal-infant interface.


Asunto(s)
Proteína ADAM10 , Animales Recién Nacidos , Toxinas Bacterianas , Proteínas Hemolisinas , Infecciones Estafilocócicas , Vacunas Estafilocócicas , Staphylococcus aureus , Vacunación , Animales , Staphylococcus aureus/inmunología , Proteína ADAM10/metabolismo , Proteína ADAM10/inmunología , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/metabolismo , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/inmunología , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Ratones , Humanos , Vacunas Estafilocócicas/inmunología , Vacunas Estafilocócicas/administración & dosificación , Femenino , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/inmunología , Ratones Endogámicos C57BL , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antibacterianos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo
3.
Science ; 386(6717): 69-75, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39361752

RESUMEN

Clostridioides difficile infection (CDI) is an urgent public health threat with limited preventative options. In this work, we developed a messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccine targeting C. difficile toxins and virulence factors. This multivalent vaccine elicited robust and long-lived systemic and mucosal antigen-specific humoral and cellular immune responses across animal models, independent of changes to the intestinal microbiota. Vaccination protected mice from lethal CDI in both primary and recurrent infection models, and inclusion of non-toxin cellular and spore antigens improved decolonization of toxigenic C. difficile from the gastrointestinal tract. Our studies demonstrate mRNA-LNP vaccine technology as a promising platform for the development of novel C. difficile therapeutics with potential for limiting acute disease and promoting bacterial decolonization.


Asunto(s)
Toxinas Bacterianas , Vacunas Bacterianas , Clostridioides difficile , Infecciones por Clostridium , Nanopartículas , Vacunas Combinadas , Vacunas de ARNm , Animales , Femenino , Ratones , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/genética , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Clostridioides difficile/inmunología , Clostridioides difficile/genética , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/inmunología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Inmunidad Celular , Inmunidad Humoral , Liposomas , Ratones Endogámicos C57BL , Vacunas de ARNm/administración & dosificación , Vacunas de ARNm/inmunología , ARN Mensajero/genética , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología
4.
Dis Aquat Organ ; 160: 7-12, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39387475

RESUMEN

Acute hepatopancreatic necrosis disease (AHPND) is a devastating shrimp disease caused by a binary toxin, PirAB, produced by Vibrio parahaemolyticus and other closely related bacteria. To address AHPND, over 300 unique single-domain antibodies (also known as nanobodies) derived from the VHH domains of Lama glama heavy-chain-only antibodies were raised against either PirA or PirB and characterized. Nanobodies were shortlisted based on their affinities for either PirA or PirB, their relative stability in intestinal fluids, and their ability to reduce PirAB-induced death in brine shrimp Artemia salina. From these data, a subset of nanobodies was tested for their ability to reduce AHPND in whiteleg shrimp Penaeus vannamei, and nanobodies targeting either PirA or PirB provided significant disease protection to whiteleg shrimp. These results show that nanobodies can be a new option for shrimp farmers to reduce or eliminate the impact of AHPND on their operations.


Asunto(s)
Penaeidae , Anticuerpos de Dominio Único , Vibrio parahaemolyticus , Animales , Penaeidae/inmunología , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Toxinas Bacterianas/inmunología , Hepatopáncreas
5.
Gut Microbes ; 16(1): 2399215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39284098

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) are a major cause of diarrheal illness in humans and animals, induced by enterotoxins produced by these pathogens. Despite the crucial role of neutrophils in combatting bacterial infections, our understanding of how enterotoxins impact neutrophil function is limited. To address this knowledge gap, we used heat-labile enterotoxin (LT) and heat-stable enterotoxin a (STa) to investigate their impact on the effector functions of neutrophils. Our study reveals that pSTa does not exert any discernible effect on the function of neutrophils. In contrast, LT altered the migration and phagocytosis of neutrophils and induced the production of inflammatory factors via activation of cAMP/PKA and ERK1/2 signaling. LT also attenuated the release of neutrophil extracellular traps by neutrophils via the PKA signaling pathway. Our findings provide novel insights into the impact of LT on neutrophil function, shedding light on the underlying mechanisms that govern its immunoregulatory effects. This might help ETEC in subverting the immune system and establishing infection.


Asunto(s)
Toxinas Bacterianas , Proteínas Quinasas Dependientes de AMP Cíclico , AMP Cíclico , Escherichia coli Enterotoxigénica , Enterotoxinas , Infecciones por Escherichia coli , Proteínas de Escherichia coli , Neutrófilos , Fagocitosis , Enterotoxinas/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Humanos , AMP Cíclico/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Trampas Extracelulares/metabolismo , Trampas Extracelulares/inmunología , Transducción de Señal
6.
Virulence ; 15(1): 2399798, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39229975

RESUMEN

Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs) with Methicillin-Resistant S. aureus (MRSA) strains being a major contributor in both community and hospital settings. S. aureus relies on metabolic diversity and a large repertoire of virulence factors to cause disease. This includes α-hemolysin (Hla), an integral player in tissue damage found in various models, including SSTIs. Previously, we identified a role for the Spx adapter protein, YjbH, in the regulation of several virulence factors and as an inhibitor of pathogenesis in a sepsis model. In this study, we found that YjbH is critical for tissue damage during SSTI, and its absence leads to decreased proinflammatory chemokines and cytokines in the skin. We identified no contribution of YjbI, encoded on the same transcript as YjbH. Using a combination of reporters and quantitative hemolysis assays, we demonstrated that YjbH impacts Hla expression and activity both in vitro and in vivo. Additionally, expression of Hla from a non-native promoter reversed the tissue damage phenotype of the ΔyjbIH mutant. Lastly, we identified reduced Agr activity as the likely cause for reduced Hla production in the ΔyjbH mutant. This work continues to define the importance of YjbH in the pathogenesis of S. aureus infection as well as identify a new pathway important for Hla production.


Asunto(s)
Proteínas Bacterianas , Toxinas Bacterianas , Regulación Bacteriana de la Expresión Génica , Proteínas Hemolisinas , Staphylococcus aureus , Transactivadores , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Staphylococcus aureus/patogenicidad , Staphylococcus aureus/inmunología , Staphylococcus aureus/genética , Ratones , Animales , Transactivadores/genética , Transactivadores/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/inmunología , Piel/microbiología , Piel/patología , Piel/inmunología , Factores de Virulencia/genética , Humanos , Infecciones de los Tejidos Blandos/microbiología , Infecciones de los Tejidos Blandos/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Citocinas/metabolismo , Citocinas/inmunología , Citocinas/genética
7.
Immunohorizons ; 8(9): 606-621, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39240270

RESUMEN

The human body harbors a substantial population of bacteria, which may outnumber host cells. Thus, there are multiple interactions between both cell types. Given the common presence of Staphylococcus aureus in the human body and the role of Th17 cells in controlling this pathogen on mucous membranes, we sought to investigate the effect of α-hemolysin, which is produced by this bacterium, on differentiating Th17 cells. RNA sequencing analysis revealed that α-hemolysin influences the expression of signature genes for Th17 cells as well as genes involved in epigenetic regulation. We observed alterations in various histone marks and genome methylation levels via whole-genome bisulfite sequencing. Our findings underscore how bacterial proteins can significantly influence the transcriptome, epigenome, and phenotype of human Th17 cells, highlighting the intricate and complex nature of the interaction between immune cells and the microbiota.


Asunto(s)
Toxinas Bacterianas , Epigénesis Genética , Proteínas Hemolisinas , Staphylococcus aureus , Células Th17 , Staphylococcus aureus/inmunología , Staphylococcus aureus/genética , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/metabolismo , Humanos , Células Th17/inmunología , Células Th17/metabolismo , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/genética , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Metilación de ADN , Diferenciación Celular , Transcriptoma
8.
Antimicrob Agents Chemother ; 68(10): e0069424, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39269189

RESUMEN

Treatment of Pseudomonas aeruginosa infection is challenging due to its intrinsic and acquired antibiotic resistance. As the number of current therapeutic options for P. aeruginosa infections is limited, developing novel treatments against the pathogen is an urgent clinical priority. The suppression of virulence of P. aeruginosa could be a new therapeutic option, and the type III secretion system (T3SS), which enables the bacteria to translocate various kinds of toxins into host cells and inhibits cellular functions, is considered as one possible target. In this report, we examined T3SS inhibition by COT-143/INFEX702, a humanized monoclonal antibody against PcrV, T3SS component, and present the crystal structure of the antibody-PcrV complex. COT-143 inhibited T3SS-dependent cytotoxicity and protected mice from the mortality caused by P. aeruginosa infection. The inhibition of cytotoxicity coincided with inhibition of translocon formation in a host cell membrane, which is necessary for T3SS intoxication. COT-143 protected murine neutrophils and facilitated phagocytosis of P. aeruginosa. These results suggest that COT-143 facilitates P. aeruginosa clearance by protecting neutrophil via inhibition of T3SS-dependent toxin translocation. This is the first report to show that an anti-PcrV antibody directly interferes with translocon formation to inhibit intoxication of host cells.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Sistemas de Secreción Tipo III , Animales , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Ratones , Sistemas de Secreción Tipo III/antagonistas & inhibidores , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/inmunología , Humanos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Anticuerpos Monoclonales/farmacología , Fagocitosis/efectos de los fármacos , Anticuerpos Monoclonales Humanizados/farmacología , Femenino , Ratones Endogámicos BALB C , Antígenos Bacterianos , Proteínas Citotóxicas Formadoras de Poros
9.
Anaerobe ; 89: 102896, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39127403

RESUMEN

INTRODUCTION: Clostridioides difficile is the main cause of antibiotic-associated diarrhea in humans and is a major enteropathogen in several animal species. In newborn piglets, colonic lesions caused by C. difficile A and B toxins (TcdA and TcdB, respectively) cause diarrhea and significant production losses. OBJECTIVE: The present study aimed to develop two recombinant vaccines from immunogenic C-terminal fragments of TcdA and TcdB and evaluate the immune response in rabbits and in breeding sows. Two vaccines were produced: bivalent (rAB), consisting of recombinant fragments of TcdA and TcdB, and chimeric (rQAB), corresponding to the synthesis of the same fragments in a single protein. Groups of rabbits were inoculated with 10 or 50 µg of proteins adjuvanted with aluminum or 0.85 % sterile saline in a final volume of 1 mL/dose. Anti-TcdA and anti-TcdB IgG antibodies were detected in rabbits and sows immunized with both rAB and rQAB vaccines by ELISA. The vaccinated sows were inoculated intramuscularly with 20 µg/dose using a prime-boost approach. RESULTS: Different antibody titers (p ≤ 0.05) were observed among the vaccinated groups of sows (rAB and rQAB) and control. Additionally, newborn piglets from vaccinated sows were also positive for anti-TcdA and anti-TcdB IgGs, in contrast to control piglets (p ≤ 0.05). Immunization of sows with the rQAB vaccine conferred higher anti-TcdA and anti-TcdB responses in piglets, suggesting the superiority of this compound over rAB. CONCLUSION: The synthesized recombinant proteins were capable of inducing antibody titers against C. difficile toxins A and B in sows, and were passively transferred to piglets through colostrum.


Asunto(s)
Animales Recién Nacidos , Anticuerpos Antibacterianos , Toxinas Bacterianas , Vacunas Bacterianas , Clostridioides difficile , Infecciones por Clostridium , Enfermedades de los Porcinos , Vacunas Sintéticas , Animales , Femenino , Porcinos , Conejos , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/veterinaria , Infecciones por Clostridium/inmunología , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Embarazo , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Clostridioides difficile/inmunología , Clostridioides difficile/genética , Anticuerpos Antibacterianos/sangre , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/genética , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Enterotoxinas/inmunología , Enterotoxinas/genética
10.
Korean J Intern Med ; 39(4): 659-667, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38986495

RESUMEN

BACKGROUND/AIMS: Sensitization to staphylococcal superantigens (SAgs) could contribute to asthma severity. However, its relevance with eosinophilic phenotype has not yet been clarified. This study aimed to investigate associations between serum specific IgE levels to SAg and eosinophilic airway inflammation in adult asthmatics. METHODS: The serum specific IgE levels to 3 SAgs, including staphylococcal enterotoxin A (SEA) and B (SEB), and toxic shock syndrome toxin-1 (TSST-1) were measured by ImmunoCAP in 230 adult asthmatic patients and 50 healthy controls (HCs). Clinical characteristics and laboratory parameters, including serum total/free IgE, and 2 eosinophil-activation markers, eosinophil cationic protein (ECP), and eosinophil-derived neurotoxin (EDN), were analyzed according to blood eosinophil counts (BEC; 150 cells/µL) and serum specific IgE levels to 3 SAgs (0.35 kU/L). RESULTS: Asthmatic patients showed higher serum specific IgE levels to 3 SAgs than HCs (p < 0.05 for all). The serum total/clinfree IgE levels were significantly higher in asthmatics with positive IgE responses to 3 SAgs than those without (p < 0.05 for all). There were no significant differences in clinical parameters including age, asthma severity, comorbidities, or smoking according to IgE responses to 3 SAgs. Patients with positive IgE responses to SEB (not to SEA/TSST-1) had higher serum specific IgE levels to house dust mites and ECP/EDN as well as higher BEC with positive correlations between serum SEB-specific IgE levels and BEC/ECP/EDN (p < 0.05 for all). CONCLUSION: These findings suggest that serum SEB-specific IgE levels could contribute to eosinophil activation as well as IgE production in adult asthma.


Asunto(s)
Asma , Enterotoxinas , Eosinófilos , Inmunoglobulina E , Fenotipo , Superantígenos , Humanos , Enterotoxinas/inmunología , Inmunoglobulina E/sangre , Masculino , Asma/inmunología , Asma/sangre , Asma/diagnóstico , Femenino , Persona de Mediana Edad , Adulto , Eosinófilos/inmunología , Estudios de Casos y Controles , Superantígenos/inmunología , Superantígenos/sangre , Biomarcadores/sangre , Anciano , Eosinofilia/inmunología , Eosinofilia/sangre , Eosinofilia/diagnóstico , Proteína Catiónica del Eosinófilo/sangre , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/sangre , Neurotoxina Derivada del Eosinófilo/sangre
11.
Infect Immun ; 92(8): e0030024, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39057915

RESUMEN

The cGAS/STING sensor system drives innate immune responses to intracellular microbial double-stranded DNA (dsDNA) and bacterial cyclic nucleotide second messengers (e.g., c-di-AMP). STING-dependent cell-intrinsic responses can increase resistance to microbial infection and speed pathogen clearance. Correspondingly, STING activation and signaling are known to be targeted for suppression by effectors from several bacterial pathogens. Whether STING responses are also positively regulated through sensing of specific bacterial effectors is less clear. We find that STING activation through dsDNA, by its canonical ligand 2'-3' cGAMP, or the small molecule DMXAA is potentiated following intracellular delivery of the AB5 toxin family member pertussis toxin from Bordetella pertussis or the B subunit of cholera toxin from Vibrio cholerae. Entry of pertussis toxin or cholera toxin B into mouse macrophages triggers markers of endoplasmic reticulum (ER) stress and enhances ligand-dependent STING responses at the level of STING receptor activation in a manner that is independent of toxin enzymatic activity. Our results provide an example in which STING responses integrate information about the presence of relevant ER-transiting bacterial toxins into the innate inflammatory response and may help to explain the in vivo adjuvant effects of catalytically inactive toxins.


Asunto(s)
Toxinas Bacterianas , Estrés del Retículo Endoplásmico , Inmunidad Innata , Proteínas de la Membrana , Animales , Ratones , Estrés del Retículo Endoplásmico/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/inmunología , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Transducción de Señal , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Humanos
12.
Mol Immunol ; 173: 10-19, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39004021

RESUMEN

Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a ß-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.


Asunto(s)
Adyuvantes Inmunológicos , Enterotoxinas , Proteínas de Escherichia coli , Animales , Femenino , Humanos , Ratones , Antígenos Virales/inmunología , Antígenos Virales/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/genética , Enterotoxinas/inmunología , Enterotoxinas/genética , Escherichia coli/inmunología , Proteínas de Escherichia coli/inmunología , Proteínas de Escherichia coli/genética , Inmunidad Mucosa/inmunología , Inmunoglobulina G/inmunología , Ratones Endogámicos BALB C , Mutación , Rotavirus/inmunología , Infecciones por Rotavirus/inmunología , Vacunas contra Rotavirus/inmunología , Vacunas contra Rotavirus/administración & dosificación
13.
Cell Host Microbe ; 32(6): 794-803, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870897

RESUMEN

Most bacteria live in communities, often with closely related strains and species with whom they must compete for space and resources. Consequently, bacteria have acquired or evolved mechanisms to antagonize competitors through the production of antibacterial toxins. Similar to bacterial systems that combat phage infection and mechanisms to thwart antibiotics, bacteria have also acquired and evolved features to protect themselves from antibacterial toxins. Just as there is a large body of research identifying and characterizing antibacterial proteins and toxin delivery systems, studies of bacterial mechanisms to resist and survive assault from competitors' weapons have also expanded tremendously. Emerging data are beginning to reveal protective processes and mechanisms that are as diverse as the toxins themselves. Protection against antibacterial toxins can be acquired by horizontal gene transfer, receptor or target alteration, induction of protective functions, physical barriers, and other diverse processes. Here, we review recent studies in this rapidly expanding field.


Asunto(s)
Bacterias , Toxinas Bacterianas , Bacterias/inmunología , Bacterias/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/inmunología , Transferencia de Gen Horizontal , Humanos , Viabilidad Microbiana , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
14.
Toxins (Basel) ; 16(6)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38922136

RESUMEN

Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of the infection are toxins produced by C. difficile, with toxin A (TcdA) and toxin B (TcdB) as the main virulence factors. Additionally, some strains produce a third toxin known as C. difficile transferase (CDT). Toxins damage the colonic epithelium, initiating a cascade of cellular events that lead to inflammation, fluid secretion, and further tissue damage within the colon. Mechanistically, the toxins bind to cell surface receptors, internalize, and then inactivate GTPase proteins, disrupting the organization of the cytoskeleton and affecting various Rho-dependent cellular processes. This results in a loss of epithelial barrier functions and the induction of cell death. The third toxin, CDT, however, functions as a binary actin-ADP-ribosylating toxin, causing actin depolymerization and inducing the formation of microtubule-based protrusions. In this review, we summarize our current understanding of the interaction between C. difficile toxins and host cells, elucidating the functional consequences of their actions. Furthermore, we will outline how this knowledge forms the basis for developing innovative, toxin-based strategies for treating and preventing CDI.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Interacciones Microbiota-Huesped , Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Orden Génico , Inflamación/patología , Humanos , Animales
15.
Nat Commun ; 15(1): 5467, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937463

RESUMEN

The genome of Mycobacterium tuberculosis encodes for a large repertoire of toxin-antitoxin systems. In the present study, MenT3 and MenT4 toxins belonging to MenAT subfamily of TA systems have been functionally characterized. We demonstrate that ectopic expression of these toxins inhibits bacterial growth and this is rescued upon co-expression of their cognate antitoxins. Here, we show that simultaneous deletion of menT3 and menT4 results in enhanced susceptibility of M. tuberculosis upon exposure to oxidative stress and attenuated growth in guinea pigs and mice. We observed reduced expression of transcripts encoding for proteins that are essential or required for intracellular growth in mid-log phase cultures of ΔmenT4ΔT3 compared to parental strain. Further, the transcript levels of proteins involved in efficient bacterial clearance were increased in lung tissues of ΔmenT4ΔT3 infected mice relative to parental strain infected mice. We show that immunization of mice and guinea pigs with ΔmenT4ΔT3 confers significant protection against M. tuberculosis infection. Remarkably, immunization of mice with ΔmenT4ΔT3 results in increased antigen-specific TH1 bias and activated memory T cell response. We conclude that MenT3 and MenT4 are important for M. tuberculosis pathogenicity and strains lacking menT3 and menT4 have the potential to be explored further as vaccine candidates.


Asunto(s)
Proteínas Bacterianas , Mycobacterium tuberculosis , Tuberculosis , Animales , Cobayas , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Ratones , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Tuberculosis/microbiología , Femenino , Pulmón/microbiología , Pulmón/patología , Pulmón/inmunología , Eliminación de Gen , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Ratones Endogámicos C57BL , Vacunas contra la Tuberculosis/inmunología , Estrés Oxidativo , Virulencia/genética
16.
ACS Sens ; 9(7): 3697-3706, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38934367

RESUMEN

Overactive or dysregulated cytokine expression is a hallmark of many acute and chronic inflammatory diseases. This is true for acute or chronic infections, neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, cancer, and others. Cytokines such as interleukin-6 (IL-6) are known therapeutic targets and biomarkers for such inflammatory diseases. Platforms for cytokine detection are, therefore, desirable tools for both research and clinical applications. Single-walled carbon nanotubes (SWCNT) are versatile nanomaterials with near-infrared fluorescence that can serve as transducers for optical sensors. When functionalized with an analyte-specific recognition element, SWCNT emission may become sensitive and selective toward the desired target. SWCNT-aptamer sensors are easily assembled, inexpensive, and biocompatible. In this work, we introduced a nanosensor design based on SWCNT and a DNA aptamer specific to IL-6. We first evaluated several SWCNT-aptamer constructs based on this simple direct complexation method, wherein the aptamer both solubilizes the SWCNT and confers sensitivity to IL-6. The sensor limit of detection, 105 ng/mL, lies in the relevant range for pathological IL-6 levels. Upon investigation of sensor kinetics, we found rapid response within seconds of antigen addition which continued over the course of 3 h. We found that this sensor construct is stable and the aptamer is not displaced from the nanotube surface during IL-6 detection. Finally, we investigated the ability of this sensor construct to detect macrophage activation caused by bacterial lipopolysaccharides (LPS) in an in vitro model of disease, finding rapid and sensitive detection of macrophage-expressed IL-6. We are confident that further development of this sensor will have novel implications for diagnosis of acute and chronic inflammatory diseases, in addition to contributing to the understanding of the role of cytokines in these diseases.


Asunto(s)
Aptámeros de Nucleótidos , Técnicas Biosensibles , Interleucina-6 , Activación de Macrófagos , Nanotubos de Carbono , Aptámeros de Nucleótidos/química , Interleucina-6/análisis , Técnicas Biosensibles/métodos , Nanotubos de Carbono/química , Toxinas Bacterianas/inmunología , Humanos , Ratones , Animales , Citocinas/análisis , Citocinas/metabolismo , Células RAW 264.7 , Límite de Detección
17.
Protein Sci ; 33(7): e5035, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38923049

RESUMEN

Single-domain antibodies (sdAbs), such as VHHs, are increasingly being developed for gastrointestinal (GI) applications against pathogens to strengthen gut health. However, what constitutes a suitable developability profile for applying these proteins in a gastrointestinal setting remains poorly explored. Here, we describe an in vitro methodology for the identification of sdAb derivatives, more specifically divalent VHH constructs, that display extraordinary developability properties for oral delivery and functionality in the GI environment. We showcase this by developing a heterodivalent VHH construct that cross-inhibits the toxic activity of the glycosyltransferase domains (GTDs) from three different toxinotypes of cytotoxin B (TcdB) from lineages of Clostridium difficile. We show that the VHH construct possesses high stability and binding activity under gastric conditions, in the presence of bile salts, and at high temperatures. We suggest that the incorporation of early developability assessment could significantly aid in the efficient discovery of VHHs and related constructs fit for oral delivery and GI applications.


Asunto(s)
Proteínas Bacterianas , Toxinas Bacterianas , Clostridioides difficile , Anticuerpos de Dominio Único , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Clostridioides difficile/inmunología , Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Humanos , Tracto Gastrointestinal/metabolismo
18.
Cell Rep ; 43(5): 114245, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38761377

RESUMEN

Recurrent Clostridioides difficile infection (CDI) results in significant morbidity and mortality. We previously established that CDI in mice does not protect against reinfection and is associated with poor pathogen-specific B cell memory (Bmem), recapitulating our observations with human Bmem. Here, we demonstrate that the secreted toxin TcdB2 is responsible for subversion of Bmem responses. TcdB2 from an endemic C. difficile strain delayed immunoglobulin G (IgG) class switch following vaccination, attenuated IgG recall to a vaccine booster, and prevented germinal center formation. The mechanism of TcdB2 action included increased B cell CXCR4 expression and responsiveness to its ligand CXCL12, accounting for altered cell migration and a failure of germinal center-dependent Bmem. These results were reproduced in a C. difficile infection model, and a US Food and Drug Administration (FDA)-approved CXCR4-blocking drug rescued germinal center formation. We therefore provide mechanistic insights into C. difficile-associated pathogenesis and illuminate a target for clinical intervention to limit recurrent disease.


Asunto(s)
Proteínas Bacterianas , Toxinas Bacterianas , Clostridioides difficile , Centro Germinal , Receptores CXCR4 , Animales , Ratones , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Quimiocina CXCL12/metabolismo , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/microbiología , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Memoria Inmunológica , Ratones Endogámicos C57BL , Receptores CXCR4/metabolismo , Receptores CXCR4/inmunología
19.
Microb Pathog ; 192: 106691, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38759933

RESUMEN

Necrotic enteritis (NE) is a potentially fatal poultry disease that causes enormous economic losses in the poultry industry worldwide. The study aimed to evaluate the effects of dietary organic yeast-derived selenium (Se) on immune protection against experimental necrotic enteritis (NE) in commercial broilers. Chickens were fed basal diets supplemented with different Se levels (0.25, 0.50, and 1.00 Se mg/kg). To induce NE, Clostridium perfringens (C. perfringens) was orally administered at 14 days of age post hatch. The results showed that birds fed 0.25 Se mg/kg exhibited significantly increased body weight gain compared with the non-supplemented/infected birds. There were no significant differences in gut lesions between the Se-supplemented groups and the non-supplemented group. The antibody levels against α-toxin and NetB toxin increased with the increase between 0.25 Se mg/kg and 0.50 Se mg/kg. In the jejunal scrapings and spleen, the Se-supplementation groups up-regulated the transcripts for pro-inflammatory cytokines IL-1ß, IL-6, IL-8, iNOS, and LITAF and avian ß-defensin 6, 8, and 13 (AvBD6, 8 and 13). In conclusion, supplementation with organic yeast-derived Se alleviates the negative consequences and provides beneficial protection against experimental NE.


Asunto(s)
Alimentación Animal , Pollos , Infecciones por Clostridium , Clostridium perfringens , Citocinas , Suplementos Dietéticos , Enteritis , Enfermedades de las Aves de Corral , Selenio , Animales , Enteritis/prevención & control , Enteritis/veterinaria , Enteritis/inmunología , Enteritis/microbiología , Selenio/farmacología , Selenio/administración & dosificación , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Clostridium perfringens/inmunología , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/veterinaria , Infecciones por Clostridium/inmunología , Citocinas/metabolismo , Toxinas Bacterianas/inmunología , Necrosis , beta-Defensinas/metabolismo , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Yeyuno/microbiología , Yeyuno/patología , Bazo/inmunología , Levaduras , Óxido Nítrico Sintasa de Tipo II/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Interleucina-1beta/metabolismo , Anticuerpos Antibacterianos/sangre
20.
Anaerobe ; 88: 102859, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38701911

RESUMEN

Clostridioides difficile infection causes pathology that ranges in severity from diarrhea to pseudomembranous colitis. Toxin A and Toxin B are the two primary virulence factors secreted by C. difficile that drive disease severity. The toxins damage intestinal epithelial cells leading to a loss of barrier integrity and induction of a proinflammatory host response. Monoclonal antibodies (mAbs) that neutralize Toxin A and Toxin B, actoxumab and bezlotoxumab, respectively, significantly reduce disease severity in a murine model of C. difficile infection. However, the impact of toxin neutralization on the induction and quality of the innate immune response following infection is unknown. The goal of this study was to define the quality of the host innate immune response in the context of anti-toxin mAbs therapy. At day 2 post-infection, C. difficile-infected, mAbs-treated mice had significantly less disease compared to isotype-treated mice despite remaining colonized with C. difficile. C. difficile-infected mAbs-treated mice still exhibited marked neutrophil infiltration and induction of a subset of proinflammatory cytokines within the intestinal lamina propria following infection that is comparable to isotype-treated mice. Furthermore, both mAbs and isotype-treated mice had an increase in IL-22-producing ILCs in the intestine following infection. MAbs-treated mice exhibited increased infiltration of eosinophils in the intestinal lamina propria, which has been previously reported to promote a protective host response following C. difficile infection. These findings show that activation of host protective mechanisms remain intact in the context of monoclonal antibody-mediated toxin neutralization.


Asunto(s)
Anticuerpos Monoclonales , Toxinas Bacterianas , Clostridioides difficile , Infecciones por Clostridium , Inmunidad Innata , Animales , Toxinas Bacterianas/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/microbiología , Ratones , Enterotoxinas/inmunología , Modelos Animales de Enfermedad , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/inmunología , Femenino , Citocinas/metabolismo , Anticuerpos ampliamente neutralizantes/inmunología , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA