Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.530
Filtrar
1.
Cell ; 167(3): 670-683.e10, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27768890

RESUMO

Spotted fever group (SFG) rickettsiae are human pathogens that infect cells in the vasculature. They disseminate through host tissues by a process of cell-to-cell spread that involves protrusion formation, engulfment, and vacuolar escape. Other bacterial pathogens rely on actin-based motility to provide a physical force for spread. Here, we show that SFG species Rickettsia parkeri typically lack actin tails during spread and instead manipulate host intercellular tension and mechanotransduction to promote spread. Using transposon mutagenesis, we identified surface cell antigen 4 (Sca4) as a secreted effector of spread that specifically promotes protrusion engulfment. Sca4 interacts with the cell-adhesion protein vinculin and blocks association with vinculin's binding partner, α-catenin. Using traction and monolayer stress microscopy, we show that Sca4 reduces vinculin-dependent mechanotransduction at cell-cell junctions. Our results suggest that Sca4 relieves intercellular tension to promote protrusion engulfment, which represents a distinctive strategy for manipulating cytoskeletal force generation to enable spread.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Mecanotransdução Celular , Infecções por Rickettsia/metabolismo , Infecções por Rickettsia/microbiologia , Rickettsia/patogenicidade , Vinculina/metabolismo , Actinas/metabolismo , Sequência de Aminoácidos , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Elementos de DNA Transponíveis/genética , Febre/metabolismo , Febre/microbiologia , Humanos , Mutagênese Insercional , Mutação , Rickettsia/metabolismo , alfa Catenina/metabolismo
2.
FASEB J ; 38(1): e23326, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38019196

RESUMO

The identification and recombinant production of functional antigens and/or epitopes of pathogens represent a crucial step for the development of an effective protein-based vaccine. Many vaccine targets are outer membrane proteins anchored into the lipidic bilayer through an extended hydrophobic portion making their recombinant production challenging. Moreover, only the extracellular loops, and not the hydrophobic regions, are naturally exposed to the immune system. In this work, the Domain 3 (D3) from Group B Streptococcus (GBS) pilus 2a backbone protein has been identified and engineered to be used as a scaffold for the display of extracellular loops of two Neisseria gonorrhoeae membrane proteins (PorB.1b and OpaB). A computational structure-based approach has been applied to the design of both the scaffold and the model antigens. Once identified the best D3 engineerable site, several different chimeric D3 displaying PorB.1b and OpaB extracellular loops were produced as soluble proteins. Each molecule has been characterized in terms of solubility, stability, and ability to correctly display the foreign epitope. This antigen dissection strategy allowed the identification of most immunogenic extracellular loops of both PorB.1b and OpaB gonococcal antigens. The crystal structure of chimeric D3 displaying PorB.1b immunodominant loop has been obtained confirming that the engineerization did not alter the predicted native structure of this epitope. Taken together, the reported data suggest that D3 is a novel protein scaffold for epitope insertion and display, and a valid alternative to the production of whole membrane protein antigens. Finally, this work describes a generalized computational structure-based approach for the identification, design, and dissection of epitopes in target antigens through chimeric proteins.


Assuntos
Proteínas de Membrana , Vacinas , Epitopos/genética , Antígenos de Bactérias/genética , Bicamadas Lipídicas
3.
Proc Natl Acad Sci U S A ; 119(11): e2109667119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35275791

RESUMO

SignificanceYersinia pestis, the etiologic agent of plague, has been responsible for high mortality in several epidemics throughout human history. This plague bacillus has been used as a biological weapon during human history and is currently one of the deadliest biological threats. Currently, no licensed plague vaccines are available in the Western world. Since an array of immunogens are enclosed in outer membrane vesicles (OMVs), immune responses elicited by OMVs against a diverse range of antigens may reduce the likelihood of antigen circumvention. Therefore, self-adjuvanting OMVs from a remodeled Yersinia pseudotuberculosis strain as a type of plague vaccine could diversify prophylactic choices and solve current vaccine limitations.


Assuntos
Antígenos de Bactérias , Lipídeo A , Vacina contra a Peste , Peste , Proteínas Citotóxicas Formadoras de Poros , Yersinia pseudotuberculosis , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Dose Letal Mediana , Lipídeo A/genética , Lipídeo A/imunologia , Camundongos , Peste/prevenção & controle , Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/genética , Vacina contra a Peste/imunologia , Plasmídeos/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/imunologia , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/imunologia
4.
Proc Natl Acad Sci U S A ; 119(11): e2122161119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35271388

RESUMO

SignificanceTuberculosis (TB), an ancient disease of humanity, continues to be a major cause of worldwide death. The causative agent of TB, Mycobacterium tuberculosis, and its close pathogenic relative Mycobacterium marinum, initially infect, evade, and exploit macrophages, a major host defense against invading pathogens. Within macrophages, mycobacteria reside within host membrane-bound compartments called phagosomes. Mycobacterium-induced damage of the phagosomal membranes is integral to pathogenesis, and this activity has been attributed to the specialized mycobacterial secretion system ESX-1, and particularly to ESAT-6, its major secreted protein. Here, we show that the integrity of the unstructured ESAT-6 C terminus is required for macrophage phagosomal damage, granuloma formation, and virulence.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Mycobacterium marinum , Mycobacterium tuberculosis , Fagossomos , Tuberculoma , Sistemas de Secreção Tipo VII , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Mycobacterium marinum/metabolismo , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Fagossomos/metabolismo , Fagossomos/microbiologia , Conformação Proteica , Tuberculoma/microbiologia , Sistemas de Secreção Tipo VII/metabolismo , Virulência
5.
Proc Natl Acad Sci U S A ; 119(25): e2202059119, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35714289

RESUMO

The bacterial genus Bartonella comprises numerous emerging pathogens that cause a broad spectrum of disease manifestations in humans. The targets and mechanisms of the anti-Bartonella immune defense are ill-defined and bacterial immune evasion strategies remain elusive. We found that experimentally infected mice resolved Bartonella infection by mounting antibody responses that neutralized the bacteria, preventing their attachment to erythrocytes and suppressing bacteremia independent of complement or Fc receptors. Bartonella-neutralizing antibody responses were rapidly induced and depended on CD40 signaling but not on affinity maturation. We cloned neutralizing monoclonal antibodies (mAbs) and by mass spectrometry identified the bacterial autotransporter CFA (CAMP-like factor autotransporter) as a neutralizing antibody target. Vaccination against CFA suppressed Bartonella bacteremia, validating CFA as a protective antigen. We mapped Bartonella-neutralizing mAb binding to a domain in CFA that we found is hypervariable in both human and mouse pathogenic strains, indicating mutational antibody evasion at the Bartonella subspecies level. These insights into Bartonella immunity and immune evasion provide a conceptual framework for vaccine development, identifying important challenges in this endeavor.


Assuntos
Anticorpos Neutralizantes , Antígenos de Bactérias , Bacteriemia , Infecções por Bartonella , Bartonella , Sistemas de Secreção Tipo V , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Bacteriemia/imunologia , Bacteriemia/microbiologia , Bacteriemia/prevenção & controle , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/uso terapêutico , Bartonella/genética , Bartonella/imunologia , Infecções por Bartonella/imunologia , Infecções por Bartonella/microbiologia , Infecções por Bartonella/prevenção & controle , Clonagem Molecular , Evasão da Resposta Imune , Camundongos , Sistemas de Secreção Tipo V/imunologia , Vacinação
6.
Infect Immun ; 92(1): e0024423, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38099660

RESUMO

Interactions among pathogen genotypes that vary in host specificity may affect overall transmission dynamics in multi-host systems. Borrelia burgdorferi, a bacterium that causes Lyme disease, is typically transmitted among wildlife by Ixodes ticks. Despite the existence of many alleles of B. burgdorferi's sensu stricto outer surface protein C (ospC) gene, most human infections are caused by a small number of ospC alleles ["human infectious alleles" (HIAs)], suggesting variation in host specificity associated with ospC. To characterize the wildlife host association of B. burgdorferi's ospC alleles, we used metagenomics to sequence ospC alleles from 68 infected individuals belonging to eight mammalian species trapped at three sites in suburban New Brunswick, New Jersey (USA). We found that multiple allele ("mixed") infections were common. HIAs were most common in mice (Peromyscus spp.) and only one HIA was detected at a site where mice were rarely captured. ospC allele U was exclusively found in chipmunks (Tamias striatus), and although a significant number of different alleles were observed in chipmunks, including HIAs, allele U never co-occurred with other alleles in mixed infections. Our results suggest that allele U may be excluding other alleles, thereby reducing the capacity of chipmunks to act as reservoirs for HIAs.


Assuntos
Borrelia burgdorferi , Borrelia , Coinfecção , Ixodes , Doença de Lyme , Animais , Humanos , Borrelia burgdorferi/genética , Borrelia/genética , Alelos , Doença de Lyme/microbiologia , Ixodes/genética , Ixodes/microbiologia , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Sciuridae/genética , Especificidade de Hospedeiro
7.
Infect Immun ; 92(5): e0044023, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38591882

RESUMO

Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Vacinas contra Escherichia coli , Escherichia coli Extraintestinal Patogênica , Proteínas Hemolisinas , Animais , Escherichia coli Extraintestinal Patogênica/genética , Escherichia coli Extraintestinal Patogênica/imunologia , Infecções por Escherichia coli/prevenção & controle , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/imunologia , Camundongos , Proteínas Hemolisinas/imunologia , Proteínas Hemolisinas/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Vacinas contra Escherichia coli/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Feminino , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Sistemas de Secreção Tipo V/imunologia , Sistemas de Secreção Tipo V/genética , Modelos Animais de Doenças , Humanos
8.
Infect Immun ; 92(6): e0009024, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38700336

RESUMO

bb0616 of Borrelia burgdorferi, the Lyme disease pathogen, encodes a hypothetical protein of unknown function. In this study, we showed that BB0616 was not surface-exposed or associated with the membrane through localization analyses using proteinase K digestion and cell partitioning assays. The expression of bb0616 was influenced by a reduced pH but not by growth phases, elevated temperatures, or carbon sources during in vitro cultivation. A transcriptional start site for bb0616 was identified by using 5' rapid amplification of cDNA ends, which led to the identification of a functional promoter in the 5' regulatory region upstream of bb0616. By analyzing a bb0616-deficient mutant and its isogenic complemented counterparts, we found that the infectivity potential of the mutant was significantly attenuated. The inactivation of bb0616 displayed no effect on borrelial growth in the medium or resistance to oxidative stress, but the mutant was significantly more susceptible to osmotic stress. In addition, the production of global virulence regulators such as BosR and RpoS as well as virulence-associated outer surface lipoproteins OspC and DbpA was reduced in the mutant. These phenotypes were fully restored when gene mutation was complemented with a wild-type copy of bb0616. Based on these findings, we concluded that the hypothetical protein BB0616 is required for the optimal infectivity of B. burgdorferi, potentially by impacting B. burgdorferi virulence gene expression as well as survival of the spirochete under stressful conditions.


Assuntos
Proteínas de Bactérias , Borrelia burgdorferi , Regulação Bacteriana da Expressão Gênica , Doença de Lyme , Borrelia burgdorferi/genética , Borrelia burgdorferi/patogenicidade , Borrelia burgdorferi/metabolismo , Animais , Camundongos , Doença de Lyme/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Regiões Promotoras Genéticas , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Virulência , Camundongos Endogâmicos C3H , Fator sigma/genética , Fator sigma/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Sítio de Iniciação de Transcrição , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Teste de Complementação Genética , Concentração de Íons de Hidrogênio
9.
PLoS Pathog ; 18(6): e1010628, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35767594

RESUMO

Helicobacter pylori (H. pylori) is a common gastric pathogen that infects approximately half of the world's population. Infection with H. pylori can lead to diverse pathological conditions, including chronic gastritis, peptic ulcer disease, and cancer. The latter is the most severe consequence of H. pylori infection. According to epidemiological studies, gastric infection with H. pylori is the strongest known risk factor for non-cardia gastric cancer (GC), which remains one of the leading causes of cancer-related deaths worldwide. However, it still remains to be poorly understood how host-microbe interactions result in cancer development in the human stomach. Here we focus on the H. pylori bacterial factors that affect the host ubiquitin proteasome system. We investigated E3 ubiquitin ligases SIVA1 and ULF that regulate p14ARF (p19ARF in mice) tumor suppressor. ARF plays a key role in regulation of the oncogenic stress response and is frequently inhibited during GC progression. Expression of ARF, SIVA1 and ULF proteins were investigated in gastroids, H. pylori-infected mice and human gastric tissues. The role of the H. pylori type IV secretion system was assessed using various H. pylori isogenic mutants. Our studies demonstrated that H. pylori infection results in induction of ULF, decrease in SIVA1 protein levels, and subsequent ubiquitination and degradation of p14ARF tumor suppressor. Bacterial CagA protein was found to sequentially bind to SIVA1 and ULF proteins. This process is regulated by CagA protein phosphorylation at the EPIYA motifs. Downregulation of ARF protein leads to inhibition of cellular apoptosis and oncogenic stress response that may promote gastric carcinogenesis.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Apoptose , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Carcinogênese/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Camundongos , Neoplasias Gástricas/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitinas/metabolismo
10.
PLoS Pathog ; 18(5): e1010511, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605029

RESUMO

Hematogenous dissemination is a critical step in the evolution of local infection to systemic disease. The Lyme disease (LD) spirochete, which efficiently disseminates to multiple tissues, has provided a model for this process, in particular for the key early event of pathogen adhesion to the host vasculature. This occurs under shear force mediated by interactions between bacterial adhesins and mammalian cell-surface proteins or extracellular matrix (ECM). Using real-time intravital imaging of the Lyme spirochete in living mice, we previously identified BBK32 as the first LD spirochetal adhesin demonstrated to mediate early vascular adhesion in a living mouse; however, deletion of bbk32 resulted in loss of only about half of the early interactions, suggesting the existence of at least one other adhesin (adhesin-X) that promotes early vascular interactions. VlsE, a surface lipoprotein, was identified long ago by its capacity to undergo rapid antigenic variation, is upregulated in the mammalian host and required for persistent infection in immunocompetent mice. In immunodeficient mice, VlsE shares functional overlap with OspC, a multi-functional protein that displays dermatan sulfate-binding activity and is required for joint invasion and colonization. In this research, using biochemical and genetic approaches as well as intravital imaging, we have identified VlsE as adhesin-X; it is a dermatan sulfate (DS) adhesin that efficiently promotes transient adhesion to the microvasculature under shear force via its DS binding pocket. Intravenous inoculation of mice with a low-passage infectious B. burgdorferi strain lacking both bbk32 and vlsE almost completely eliminated transient microvascular interactions. Comparative analysis of binding parameters of VlsE, BBK32 and OspC provides a possible explanation why these three DS adhesins display different functionality in terms of their ability to promote early microvascular interactions.


Assuntos
Adesinas Bacterianas , Variação Antigênica , Antígenos de Bactérias , Proteínas de Bactérias , Borrelia burgdorferi , Lipoproteínas , Doença de Lyme , Microvasos , Adesinas Bacterianas/genética , Adesinas Bacterianas/imunologia , Animais , Variação Antigênica/genética , Variação Antigênica/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Borrelia burgdorferi/genética , Borrelia burgdorferi/imunologia , Dermatan Sulfato/imunologia , Lipoproteínas/genética , Lipoproteínas/imunologia , Doença de Lyme/genética , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Mamíferos , Camundongos , Microvasos/imunologia , Microvasos/microbiologia , Resistência ao Cisalhamento
11.
Microb Pathog ; 190: 106636, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38556103

RESUMO

Enterotoxigenic Escherichia coli (ETEC) is one of the main causes of diarrhea in children and travelers in low-income regions. The virulence of ETEC is attributed to its heat-labile and heat-stable enterotoxins, as well as its colonization factors (CFs). CFs are essential for ETEC adherence to the intestinal epithelium. However, its invasive capability remains unelucidated. In this study, we demonstrated that the CS6-positive ETEC strain 4266 can invade mammalian epithelial cells. The invasive capability was reduced in the 4266 ΔCS6 mutant but reintroduction of CS6 into this mutant restored the invasiveness. Additionally, the laboratory E. coli strain Top 10, which lacks the invasive capability, was able to invade Caco-2 cells after gaining the CS6-expressing plasmid pCS6. Cytochalasin D inhibited cell invasion in both 4266 and Top10 pCS6 cells, and F-actin accumulation was observed near the bacteria on the cell membrane, indicating that CS6-positive bacteria were internalized via actin polymerization. Other cell signal transduction inhibitors, such as genistein, wortmannin, LY294002, PP1, and Ro 32-0432, inhibited the CS6-mediated invasion of Caco-2 cells. The internalized bacteria of both 4266 and Top10 pCS6 strains were able to survive for up to 48 h, and 4266 cells were able to replicate within Caco-2 cells. Immunofluorescence microscopy revealed that the internalized 4266 cells were present in bacteria-containing vacuoles, which underwent a maturation process indicated by the recruitment of the early endosomal marker EEA-1 and late endosomal marker LAMP-1 throughout the infection process. The autophagy marker LC3 was also observed near these vacuoles, indicating the initiation of LC-3-associated phagocytosis (LAP). However, intracellular bacteria continued to replicate, even after the initiation of LAP. Moreover, intracellular filamentation was observed in 4266 cells at 24 h after infection. Overall, this study shows that CS6, in addition to being a major CF, mediates cell invasion. This demonstrates that once internalized, CS6-positive ETEC is capable of surviving and replicating within host cells. This capability may be a key factor in the extended and recurrent nature of ETEC infections in humans, thus highlighting the critical role of CS6.


Assuntos
Citocalasina D , Escherichia coli Enterotoxigênica , Proteínas de Escherichia coli , Humanos , Células CACO-2 , Escherichia coli Enterotoxigênica/patogenicidade , Escherichia coli Enterotoxigênica/genética , Escherichia coli Enterotoxigênica/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Citocalasina D/farmacologia , Actinas/metabolismo , Células Epiteliais/microbiologia , Aderência Bacteriana , Infecções por Escherichia coli/microbiologia , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/genética , Morfolinas/farmacologia , Transdução de Sinais , Androstadienos/farmacologia , Wortmanina/farmacologia , Endocitose , Cromonas/farmacologia , Plasmídeos/genética
12.
Microb Pathog ; 192: 106685, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38750774

RESUMO

QseC is a membrane sensor kinase that enables bacteria to perceive autoinducers -3, adrenaline, and norepinephrine to initiate downstream gene transcription. In this study, we found that the QseC protein of Glaesserella parasuis can serve as an effective antigen to activate the host's immune response. Therefore, we investigated the immunogenicity and host protective effect of this protein. ELISA and indirect immunofluorescence results showed that QseC protein can induce high titer levels of humoral immunity in mice and regularly generate specific serum antibodies. We used MTS reagents to detect lymphocyte proliferation levels and found that QseC protein can cause splenic lymphocyte proliferation with memory and specificity. Further immunological analysis of the spleen cell supernatant revealed significant upregulation of levels of IL-1ß, IL-4 and IFN-γ in the QseC + adjuvant group. In the mouse challenge experiment, it was found that QseC + adjuvant can provide effective protection. The results of this study demonstrate that QseC protein provides effective protection in a mouse model and has the potential to serve as a candidate antigen for a novel subunit vaccine for further research.


Assuntos
Anticorpos Antibacterianos , Infecções por Haemophilus , Interferon gama , Interleucina-4 , Animais , Camundongos , Interleucina-4/metabolismo , Interleucina-4/imunologia , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Infecções por Haemophilus/imunologia , Infecções por Haemophilus/prevenção & controle , Infecções por Haemophilus/microbiologia , Interferon gama/metabolismo , Histidina Quinase/genética , Histidina Quinase/metabolismo , Histidina Quinase/imunologia , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Imunidade Humoral , Camundongos Endogâmicos BALB C , Baço/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/genética , Proliferação de Células , Feminino , Adjuvantes Imunológicos , Haemophilus parasuis/imunologia , Haemophilus parasuis/genética , Citocinas/metabolismo , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/genética , Modelos Animais de Doenças , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Linfócitos/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Vacinas de Subunidades Antigênicas/genética
13.
Biotechnol Bioeng ; 121(7): 2163-2174, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38595326

RESUMO

Pathogenic bacterial membrane proteins (MPs) are a class of vaccine and antibiotic development targets with widespread clinical application. However, the inherent hydrophobicity of MPs poses a challenge to fold correctly in living cells. Herein, we present a comprehensive method to improve the soluble form of MP antigen by rationally designing multi-epitope chimeric antigen (ChA) and screening two classes of protein-assisting folding element. The study uses a homologous protein antigen as a functional scaffold to generate a ChA possessing four epitopes from transferrin-binding protein A of Glaesserella parasuis. Our engineered strain, which co-expresses P17 tagged-ChA and endogenous chaperones groEL-ES, yields a 0.346 g/L highly soluble ChA with the property of HPS-positive serum reaction. Moreover, the protein titer of ChA reaches 4.27 g/L with >90% soluble proportion in 5-L bioreactor, which is the highest titer reported so far. The results highlight a timely approach to design and improve the soluble expression of MP antigen in industrially viable applications.


Assuntos
Antígenos de Bactérias , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Reatores Biológicos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Escherichia coli/genética , Escherichia coli/metabolismo , Clostridiales/genética , Clostridiales/metabolismo , Solubilidade
14.
Genome ; 67(6): 204-209, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38330385

RESUMO

Mycoplasmopsis bovis is a worldwide economically important pathogen of cattle that can cause or indirectly contribute to bovine respiratory disease. M. bovis is also a primary etiological agent of respiratory disease in bison with high mortality rates. A major challenge in the development of an efficacious M. bovis vaccine is the design of antigens that contain both MHC-1 and MHC-2 T-cell epitopes, and that account for population level diversity within the species. Publicly available genomes and sequence read archive libraries of 381 M. bovis strains isolated from cattle (n = 202) and bison (n = 179) in North America were used to identify a core genome of 575 genes, including 38 that encode either known or predicted secreted or outer membrane proteins. The antigenic potentials of the proteins were characterized by the presence and strength of their T-cell epitopes, and their protein variant diversity at the population-level. The proteins had surprisingly low diversity and varying predictive levels of T-cell antigenicity. These results provide a reference for the selection or design of antigens for vaccine testing against strains infecting North American cattle and bison.


Assuntos
Bison , Animais , Bison/microbiologia , Bovinos , América do Norte , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/genética , Variação Genética , Mycoplasma bovis/genética , Mycoplasma bovis/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Genoma Bacteriano , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Doenças dos Bovinos/microbiologia , Doenças dos Bovinos/imunologia
15.
Eur J Clin Microbiol Infect Dis ; 43(2): 233-241, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38010594

RESUMO

PURPOSE: Streptococcus pyogenes (mostly termed group A Streptococcus - GAS) is the most important bacterial causative of pharyngitis. However, epidemiology of GAS pharyngitis is not widely established. This study describes GAS pharyngitis cases and emm-type distribution in a prospective study covering over 2 years in two Hospital Districts in Finland. METHODS: A prospective, systematic collection of GAS pharyngitis isolates was conducted between March 2018 and December 2020 in two large Hospital Districts in Finland. Patient characteristics (age, gender) were included if available. All GAS isolates collected were emm typed. RESULTS: Altogether 1320 GAS pharyngitis strains were collected, 904 in the Hospital District 1 (HD1) and 416 in Hospital District 2 (HD2). In HD1, age and gender data were available. Females were overrepresented (58% of all cases). In addition, the age and gender distributions were noted to be significantly different (p < 0.0001) with females having a more uniform distribution until age of 40. emm28 was common among the age group of 20-29-year-olds and emm89 in children under 10 years of age, respectively. In HD1, most of the isolates were collected during winter and autumn months. Significant differences by season in the frequency of emm12, emm89, emm75 and group of "others" were observed. CONCLUSION: Age distribution among GAS pharyngitis cases was significantly different between genders (p < 0.0001). In addition, age group specific and seasonal variations in emm GAS types causing the disease were observed. These findings warrant further investigation, especially for understanding population-based spread of GAS even in more detail.


Assuntos
Faringite , Infecções Estreptocócicas , Criança , Humanos , Feminino , Masculino , Adulto Jovem , Adulto , Streptococcus pyogenes , Estudos Prospectivos , Finlândia/epidemiologia , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Infecções Estreptocócicas/epidemiologia , Infecções Estreptocócicas/microbiologia , Proteínas de Transporte/genética , Faringite/epidemiologia , Faringite/microbiologia , Genótipo
16.
Helicobacter ; 29(3): e13104, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38923222

RESUMO

Helicobacter pylori (H. pylori) strain is the most genetically diverse pathogenic bacterium and now alarming serious human health concern ranging from chronic gastritis to gastric cancer and human death all over the world. Currently, the majority of commercially available diagnostic assays for H. pylori is a challenging task due to the heterogeneity of virulence factors in various geographical regions. In this concern, designing of universal multi-epitope immunogenic biomarker targeted for all H. pylori strains would be crucial to successfully immunodiagnosis assay and vaccine development for H. pylori infection. Hence, the present study aimed to explore the potential immunogenic epitopes of PSA D15 and Cag11 proteins of H. pylori, using immunoinformatics web tools in order to design novel immune-reactive multi-epitope antigens for enhanced immunodiagnosis in humans. Through an in silico immunoinformatics approach, high-ranked B-cell, MHC-I, and MHC-II epitopes of PSA D15 and Cag11 proteins were predicted, screened, and selected. Subsequently, a novel multi-epitope PSA D15 and Cag11 antigens were designed by fused the high-ranked B-cell, MHC-I, and MHC-II epitopes and 50S ribosomal protein L7/L12 adjuvant using linkers. The antigenicity, solubility, physicochemical properties, secondary and tertiary structures, 3D model refinement, and validations were carried. Furthermore, the designed multi-epitope antigens were subjected to codon adaptation and in silico cloning, immune response simulation, and molecular docking with receptor molecules. A novel, stable multi-epitope PSA D15 and Cag11 H. pylori antigens were developed and immune simulation of the designed antigens showed desirable levels of immunological response. Molecular docking of designed antigens with immune receptors (B-cell, MHC-I, MHC-II, and TLR-2/4) revealed robust interactions and stable binding affinity to the receptors. The codon optimized and in silico cloned showed that the designed antigens were successfully expressed (CAI value of 0.95 for PSA D15 and 1.0 for Cag11) after inserted into pET-32ba (+) plasmid of the E. coli K12 strain. In conclusion, this study revealed that the designed multi-epitope antigens have a huge immunological potential candidate biomarker and useful in developing immunodiagnostic assays and vaccines for H. pylori infection.


Assuntos
Antígenos de Bactérias , Biologia Computacional , Helicobacter pylori , Helicobacter pylori/imunologia , Helicobacter pylori/genética , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/química , Humanos , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Epitopos/imunologia , Testes Imunológicos/métodos , Simulação de Acoplamento Molecular , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/genética , Imunoinformática
17.
Helicobacter ; 29(3): e13100, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38873839

RESUMO

BACKGROUND: The formation of gallstones is often accompanied by chronic inflammation, and the mechanisms underlying inflammation and stone formation are not fully understood. Our aim is to utilize single-cell transcriptomics, bulk transcriptomics, and microbiome data to explore key pathogenic bacteria that may contribute to chronic inflammation and gallstone formation, as well as their associated mechanisms. METHODS: scRNA-seq data from a gallstone mouse model were extracted from the Gene Expression Omnibus (GEO) database and analyzed using the FindCluster() package for cell clustering analysis. Bulk transcriptomics data from patients with gallstone were also extracted from the GEO database, and intergroup functional differences were assessed using GO and KEGG enrichment analysis. Additionally, 16S rRNA sequencing was performed on gallbladder mucosal samples from asymptomatic patients with gallstone (n = 6) and liver transplant donor gallbladder mucosal samples (n = 6) to identify key bacteria associated with stone formation and chronic inflammation. Animal models were constructed to investigate the mechanisms by which these key pathogenic bacterial genera promote gallstone formation. RESULTS: Analysis of scRNA-seq data from the gallstone mouse model (GSE179524) revealed seven distinct cell clusters, with a significant increase in neutrophil numbers in the gallstone group. Analysis of bulk transcriptomics data from patients with gallstone (GSE202479) identified chronic inflammation in the gallbladder, potentially associated with dysbiosis of the gallbladder microbiota. 16S rRNA sequencing identified Helicobacter pylori as a key bacterium associated with gallbladder chronic inflammation and stone formation. CONCLUSIONS: Dysbiosis of the gallbladder mucosal microbiota is implicated in gallstone disease and leads to chronic inflammation. This study identified H. pylori as a potential key mucosal resident bacterium contributing to gallstone formation and discovered its key pathogenic factor CagA, which causes damage to the gallbladder mucosal barrier. These findings provide important clues for the prevention and treatment of gallstones.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Células Epiteliais , Vesícula Biliar , Cálculos Biliares , Helicobacter pylori , Animais , Cálculos Biliares/microbiologia , Cálculos Biliares/patologia , Células Epiteliais/microbiologia , Camundongos , Humanos , Vesícula Biliar/microbiologia , Vesícula Biliar/patologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Helicobacter pylori/fisiologia , RNA Ribossômico 16S/genética , Modelos Animais de Doenças , Permeabilidade , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Feminino , Masculino , Camundongos Endogâmicos C57BL
18.
Infection ; 52(2): 667-671, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38064158

RESUMO

PURPOSE: Hypertoxigenic Streptococcus pyogenes emm1 lineage M1UK has recently been associated with upsurges of invasive infections and scarlet fever in several countries, but whole-genome sequencing surveillance data of lineages circulating in Germany is lacking. In this study, we investigated recent iGAS isolates from our laboratory at a German tertiary care center for the presence of the M1UK lineage. METHODS: Whole-genome sequencing was employed to characterize a collection of 47 consecutive non-copy isolates recovered from blood cultures (21) and tissue samples (26) in our laboratory between October 2022 and April 2023. RESULTS: M protein gene (emm) typing distinguished 14 different emm types, with emm1 (17) being the dominant type. Single-nucleotide polymorphism (SNP) analysis confirmed the presence of all 27 SNPs characteristic for the M1UK lineage in 14 of 17 emm1 isolates. CONCLUSION: This study has shown for the first time that M1UK is present in Germany and might constitute a driving force in the observed surge of GAS infections. This observation mirrors developments in the UK and other countries and underscores the importance of WGS surveillance to understand the epidemiology of GAS.


Assuntos
Infecções Estreptocócicas , Streptococcus pyogenes , Humanos , Streptococcus pyogenes/genética , Centros de Atenção Terciária , Genótipo , Proteínas de Transporte , Reino Unido , Infecções Estreptocócicas/epidemiologia , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética
19.
Infection ; 52(2): 345-384, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38270780

RESUMO

PURPOSE: This study aims to comprehensively review the multifaceted factors underlying the successful colonization and infection process of Helicobacter pylori (H. pylori), a prominent Gram-negative pathogen in humans. The focus is on elucidating the functions, mechanisms, genetic regulation, and potential cross-interactions of these elements. METHODS: Employing a literature review approach, this study examines the intricate interactions between H. pylori and its host. It delves into virulence factors like VacA, CagA, DupA, Urease, along with phase variable genes, such as babA, babC, hopZ, etc., giving insights about the bacterial perspective of the infection The association of these factors with the infection has also been added in the form of statistical data via Funnel and Forest plots, citing the potential of the virulence and also adding an aspect of geographical biasness to the virulence factors. The biochemical characteristics and clinical relevance of these factors and their effects on host cells are individually examined, both comprehensively and statistically. RESULTS: H. pylori is a Gram-negative, spiral bacterium that successfully colonises the stomach of more than half of the world's population, causing peptic ulcers, gastric cancer, MALT lymphoma, and other gastro-duodenal disorders. The clinical outcomes of H. pylori infection are influenced by a complex interplay between virulence factors and phase variable genes produced by the infecting strain and the host genetic background. A meta-analysis of the prevalence of all the major virulence factors has also been appended. CONCLUSION: This study illuminates the diverse elements contributing to H. pylori's colonization and infection. The interplay between virulence factors, phase variable genes, and host genetics determines the outcome of the infection. Despite biochemical insights into many factors, their comprehensive regulation remains an understudied area. By offering a panoramic view of these factors and their functions, this study enhances understanding of the bacterium's perspective, i.e. H. pylori's journey from infiltration to successful establishment within the host's stomach.


Assuntos
Helicobacter pylori , Úlcera Péptica , Neoplasias Gástricas , Humanos , Virulência/genética , Helicobacter pylori/genética , Úlcera Péptica/microbiologia , Fatores de Virulência/genética , Proteínas de Bactérias/genética , Antígenos de Bactérias/genética
20.
Cell Mol Biol Lett ; 29(1): 70, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741147

RESUMO

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.


Assuntos
Antígenos de Bactérias , Proliferação de Células , Mycobacterium tuberculosis , Linfócitos T , Humanos , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/genética , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Interferon gama/metabolismo , Interferon gama/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/genética , Fator de Necrose Tumoral alfa/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA