Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.569
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34001624

ABSTRACT

Anatomical positioning of memory lymphocytes within barrier tissues accelerates secondary immune responses and is thought to be essential for protection at mucosal surfaces. However, it remains unclear whether resident memory in the female reproductive tract (FRT) is required for Chlamydial immunity. Here, we describe efficient generation of tissue-resident memory CD4 T cells and memory lymphocyte clusters within the FRT after vaginal infection with Chlamydia Despite robust establishment of localized memory lymphocytes within the FRT, naïve mice surgically joined to immune mice, or mice with only circulating immunity following intranasal immunization, were fully capable of resisting Chlamydia infection via the vaginal route. Blocking the rapid mobilization of circulating memory CD4 T cells to the FRT inhibited this protective response. These data demonstrate that secondary protection in the FRT can occur in the complete absence of tissue-resident immune cells. The ability to confer robust protection to barrier tissues via circulating immune memory provides an unexpected opportunity for vaccine development against infections of the FRT.


Subject(s)
Antibodies, Bacterial/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Genitalia, Female/immunology , Immunization/methods , Administration, Intranasal , Administration, Intravaginal , Animals , Antigens, Bacterial/administration & dosage , Bacterial Vaccines/administration & dosage , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/microbiology , Cell Movement/drug effects , Cell Movement/immunology , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia muridarum/drug effects , Chlamydia muridarum/growth & development , Chlamydia muridarum/pathogenicity , Female , Genitalia, Female/drug effects , Genitalia, Female/microbiology , Immunity, Mucosal/drug effects , Immunologic Memory/drug effects , Mice , Parabiosis/methods
2.
Semin Immunol ; 39: 111-118, 2018 10.
Article in English | MEDLINE | ID: mdl-29950273

ABSTRACT

Leprosy is still a considerable health threat in pockets of several low and middle income countries worldwide where intense transmission is witnessed, and often results in irreversible disabilities and deformities due to delayed- or misdiagnosis. Early detection of leprosy represents a substantial hurdle in present-day leprosy health care. The dearth of timely diagnosis has, however, particularly severe consequences in the case of inflammatory episodes, designated leprosy reactions, which represent the major cause of leprosy-associated irreversible neuropathy. There is currently no accurate, routine diagnostic test to reliably detect leprosy reactions, or to predict which patients will develop these immunological exacerbations. Identification of host biomarkers for leprosy reactions, particularly if correlating with early onset prior to development of clinical symptoms, will allow timely interventions that contribute to decreased morbidity. Development of a point-of-care (POC) test based on such correlates would be a definite game changer in leprosy health care. In this review, proteomic-, transcriptomic and metabolomic research strategies aiming at identification of host biomarker-based correlates of leprosy reactions are discussed, next to external factors associated with occurrence of these episodes. The vast diversity in research strategies combined with the variability in patient- and control cohorts argues for harmonisation of biomarker discovery studies with geographically overarching study sites. This will improve identification of specific correlates associated with risk of these damaging inflammatory episodes in leprosy and subsequent application to rapid field tests.


Subject(s)
Antibodies, Bacterial/analysis , Endpoint Determination/methods , Leprosy/diagnosis , Mycobacterium leprae/immunology , Transcriptome/immunology , Antibodies, Bacterial/biosynthesis , Biomarkers/metabolism , CD30 Ligand/genetics , CD30 Ligand/immunology , Cation Transport Proteins/genetics , Cation Transport Proteins/immunology , Delayed Diagnosis , Disease Progression , Humans , Leprosy/immunology , Leprosy/microbiology , Leprosy/pathology , Metabolome/immunology , Mycobacterium leprae/isolation & purification , Mycobacterium leprae/pathogenicity , Point-of-Care Testing , Systems Biology/methods , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
3.
Eur J Immunol ; 50(5): 676-684, 2020 05.
Article in English | MEDLINE | ID: mdl-32026472

ABSTRACT

The obligate intracellular bacterium Chlamydia trachomatis causes the most prevalent bacterial sexually transmitted infection worldwide. CD4 T cells play a central role in the protective immunity against Chlamydia female reproductive tract (FRT) infection, while B cells are thought to be dispensable for resolution of primary Chlamydia infection in mouse models. We recently reported an unexpected requirement of B cells in local Chlamydia-specific CD4 T-cell priming and bacterial containment within the FRT. Here, we sought to tackle the precise effector function of B cells during Chlamydia primary infection. Using mixed bone marrow chimeras that lack B-cell-dependent Ag presentation (MHCIIB-/- ) or devoid of circulating antibodies (AID-/- × µS-/- ), we show that Chlamydia-specific CD4 T-cell expansion does not rely on Ag presentation by B cells. Importantly, we demonstrate that antibody, but not B-cell-dependent Ag presentation, is required for preventing systemic bacterial dissemination following Chlamydia FRT infection.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocytes/immunology , Bacteremia/immunology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Animals , Antigen Presentation , B-Lymphocytes/microbiology , Bacteremia/microbiology , Bacteremia/pathology , Bone Marrow Cells/microbiology , CD4-Positive T-Lymphocytes/microbiology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/pathogenicity , Disease Models, Animal , Female , Immunity, Humoral , Immunoglobulin Isotypes , Mice , Transplantation Chimera , Vagina/immunology , Vagina/microbiology , Whole-Body Irradiation
4.
Arch Microbiol ; 203(5): 2719-2725, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33606039

ABSTRACT

Heat shock proteins are molecular chaperones that are immunogens as well as potent inducers of an antigen-specific immunological response. In this study, we aimed to evaluate if co-immunization of Brucella rOmp22 and rDnaK proteins had boosted immunogenic activity as compared to rOmp22 immunization alone in mice. For this, gene-encoding DnaK of B. abortus was cloned, expressed in E. coli and purified using Ni-NTA agarose. Immuno-modulatory effect of rDnaK protein was evaluated in mice when co-immunized with Brucella rOmp22. Four groups of mice (n = 6 per group) were used in the study. The control group was immunized with rOmp22 alone, while rOmp22 emulsified with conventional adjuvants (Freund's complete and incomplete adjuvants) and rOmp22 mixed with rDnaK were injected to group I and group II in mice, respectively. Group III mice were immunized with rDnaK alone. IgG class switching (IgG1 and IgG2a) response to immunization was assessed by enzyme-linked immunosorbent assay and expression of IL-4 and IL-12 mRNA was assessed by real-time PCR to evaluate the immune response in mice. The ratio of IgG1-IgG2a was less than 1 in mice co-immunized with rOmp22 and rDnaK, indicating that the immune response was directed towards CMI arm in this group of mice. Moreover, IL-12 mRNA expression was also up-regulated to a greater extent in mice co-immunized with rOmp22 and rDnaK as compared to those immunized with rOmp22 along with the conventional adjuvants, or rOmp22 alone. Our data suggest that rDnaK could be responsible for modulating the immune response, specifically the CMI response.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Brucella abortus/immunology , HSP70 Heat-Shock Proteins/immunology , Immunoglobulin Class Switching/immunology , Animals , Antibodies, Bacterial/biosynthesis , Brucella abortus/genetics , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Immunization , Immunoglobulin G/immunology , Interleukin-12 Subunit p35/genetics , Interleukin-4/genetics , Male , Mice , Recombinant Proteins/genetics
5.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32393506

ABSTRACT

Bacillus anthracis is the causative agent of anthrax disease, presents with high mortality, and has been at the center of bioweapon efforts. The only currently U.S. FDA-approved vaccine to prevent anthrax in humans is anthrax vaccine adsorbed (AVA), which is protective in several animal models and induces neutralizing antibodies against protective antigen (PA), the cell-binding component of anthrax toxin. However, AVA requires a five-course regimen to induce immunity, along with an annual booster, and is composed of undefined culture supernatants from a PA-secreting strain. In addition, it appears to be ineffective against strains that lack anthrax toxin. Here, we investigated a vaccine formulation consisting of recombinant proteins from a surface-localized heme transport system containing near-iron transporter (NEAT) domains and its efficacy as a vaccine for anthrax disease. The cocktail of five NEAT domains was protective against a lethal challenge of inhaled bacillus spores at 3 and 28 weeks after vaccination. The reduction of the formulation to three NEATs (IsdX1, IsdX2, and Bslk) was as effective as a five-NEAT domain cocktail. The adjuvant alum, approved for use in humans, was as protective as Freund's Adjuvant, and protective vaccination correlated with increased anti-NEAT antibody reactivity and reduced bacterial levels in organs. Finally, the passive transfer of anti-NEAT antisera reduced mortality and disease severity, suggesting the protective component is comprised of antibodies. Collectively, these results provide evidence that a vaccine based upon recombinant NEAT proteins should be considered in the development of a next-generation anthrax vaccine.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antibodies, Bacterial/biosynthesis , Antibodies, Neutralizing/biosynthesis , Antigens, Bacterial/immunology , Bacillus anthracis/drug effects , Administration, Inhalation , Alum Compounds/administration & dosage , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/genetics , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Carrier Proteins/immunology , Complement C5/deficiency , Female , Freund's Adjuvant/administration & dosage , Humans , Immunogenicity, Vaccine , Mice, Knockout , Survival Analysis , Vaccination/methods
6.
Med Microbiol Immunol ; 209(2): 125-137, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31811379

ABSTRACT

The most promising means of controlling anthrax, a lethal zoonotic disease during the early infection stages, entail restricting the resilient infectious form, i.e., the spores from proliferating to replicating bacilli in the host. The extractible antigen (EA1), a major S-layer protein present on the vegetative cells and spores of Bacillus anthracis, is highly immunogenic and protects mice against lethal challenge upon immunization. In the present study, mice were immunized with r-EA1C, the C terminal crystallization domain of EA1, to generate a neutralizing monoclonal antibody EA752-862, that was evaluated for its anti-spore and anti-bacterial properties. The monoclonal antibody EA752-862 had a minimum inhibitory concentration of 0.08 mg/ml, was bactericidal at a concentration of 0.1 mg/ml and resulted in 100% survival of mice against challenge with B. anthracis vegetative cells. Bacterial cell lysis as observed by scanning electron microscopy and nucleic acid leakage assay could be attributed as a possible mechanism for the bactericidal property. The association of mAb EA752-862 with spores inhibits their subsequent germination to vegetative cells in vitro, enhances phagocytosis of the spores and killing of the vegetative cells within the macrophage, and subsequently resulted in 90% survival of mice upon B. anthracis Ames spore challenge. Therefore, owing to its anti-spore and bactericidal properties, the present study demonstrates mAb EA752-862 as an efficient neutralizing antibody that hinders the establishment of early infection before massive multiplication and toxin release takes place.


Subject(s)
Anthrax/prevention & control , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Bacillus anthracis/immunology , Spores, Bacterial/immunology , Animals , Anthrax/immunology , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/isolation & purification , Antibodies, Bacterial/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/pharmacology , Antigens, Bacterial/immunology , Bacillus anthracis/drug effects , Binding Sites , Female , Immunization , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Phagocytosis/drug effects , Phagocytosis/immunology , Spores, Bacterial/drug effects
7.
Bull Exp Biol Med ; 168(4): 485-487, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32146631

ABSTRACT

Immunogenic and protective activity of recombinant pneumolysin was studied in experiments on male BALB/c mice. The mice were immunized intraperitoneally with recombinant pneumolysin sorbed on Al(OH)3 (200 µg per mouse). In 2 weeks after immunization, the isotypes of antibodies to recombinant pneumolysin in the serum of immunized mice were determined by ELISA. The animals were infected with Streptococcus pneumoniae serotype 3. Immunization with recombinant pneumolysin induced the production of anti-pneumolysin antibodies, mainly of IgG1 subisotype. On day 21 after intraperitoneal infection with S. pneumoniae serotype 3 in a dose of 106 microbial cells, the survival rate of animals immunized with recombinant pneumolysin in a dose of 25 µg/mouse was 67% vs. 0% in the control (p<0.001). Recombinant pneumolysin could be considered as a promising protective antigen for inclusion in the serotype-independent vaccine against S. pneumoniae.


Subject(s)
Antibodies, Bacterial/biosynthesis , Immunoglobulin G/biosynthesis , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/immunology , Streptolysins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/biosynthesis , Immunization/methods , Immunogenicity, Vaccine , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Pneumococcal Infections/immunology , Pneumococcal Infections/mortality , Pneumococcal Infections/pathology , Pneumococcal Vaccines/biosynthesis , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity , Streptolysins/biosynthesis , Survival Analysis
8.
Bull Exp Biol Med ; 169(4): 474-477, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910375

ABSTRACT

The biosurfactant monoacyltrehalose fraction isolated from Rhodococcus ruber IEGM 231 actinobacterium suppresses antibody production, bactericidal potential, and production of IL-1ß by mouse peritoneal cells after intraperitoneal and intramuscular injection and stimulates the production of IL-10 after intraperitoneal injection. The data of in vitro experiments attest to an important role of bacterial glycolipids in the regulation of the functions of splenocytes and peritoneal macrophages.


Subject(s)
Immunologic Factors/pharmacology , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Rhodococcus/chemistry , Surface-Active Agents/pharmacology , Trehalose/pharmacology , Adaptive Immunity/drug effects , Animals , Antibodies, Bacterial/biosynthesis , Gene Expression , Immunity, Innate/drug effects , Immunologic Factors/chemistry , Immunologic Factors/isolation & purification , Injections, Intraperitoneal , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Male , Mice , Primary Cell Culture , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Surface-Active Agents/chemistry , Surface-Active Agents/isolation & purification , Trehalose/analogs & derivatives , Trehalose/isolation & purification
9.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31331958

ABSTRACT

Infection with Coxiella burnetii, the causative agent of Q fever, can result in life-threatening persistent infection. Reactogenicity hinders worldwide implementation of the only licensed human Q fever vaccine. We previously demonstrated long-lived immunoreactivity in individuals with past symptomatic and asymptomatic Coxiella infection (convalescents) to promiscuous HLA class II C. burnetii epitopes, providing the basis for a novel T-cell targeted subunit vaccine. In this study, we investigated in a cohort of 22 individuals treated for persistent infection (chronic Q fever) whether they recognize the same set of epitopes or distinct epitopes that could be candidates for a therapeutic vaccine or aid in the diagnosis of persistent infection. In cultured enzyme-linked immunosorbent spot (ELISpot) assays, individuals with chronic Q fever showed strong class II epitope-specific responses that were largely overlapping with the peptide repertoire identified previously for convalescents. Five additional peptides were recognized more frequently by chronic subjects, but there was no combination of epitopes uniquely recognized by or nonreactive in subjects with chronic Q fever. Consistent with more recent/prolonged exposure, we found, however, stronger ex vivo responses by direct ELISpot to both whole-cell C. burnetii and individual peptides in chronic patients than in convalescents. In conclusion, we have validated and expanded a previously published set of candidate epitopes for a novel T-cell targeted subunit Q fever vaccine in treated patients with chronic Q fever and demonstrated that they successfully mounted a T-cell response comparable to that of convalescents. Finally, we demonstrated that individuals treated for chronic Q fever mount a broader ex vivo response to class II epitopes than convalescents, which could be explored for diagnostic purposes.


Subject(s)
Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Coxiella burnetii/immunology , Epitopes, T-Lymphocyte/immunology , Q Fever/immunology , Aged , Anti-Bacterial Agents/therapeutic use , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Chronic Disease , Convalescence , Coxiella burnetii/pathogenicity , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Female , Gene Expression , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Testing , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Male , Middle Aged , Peptides/genetics , Peptides/immunology , Q Fever/drug therapy , Q Fever/genetics , Q Fever/prevention & control , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
10.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31308088

ABSTRACT

Acute otitis media is one of the most common childhood infections worldwide. Currently licensed vaccines against the common otopathogen Streptococcus pneumoniae target the bacterial capsular polysaccharide and confer no protection against nonencapsulated strains or capsular types outside vaccine coverage. Mucosal infections such as acute otitis media remain prevalent, even those caused by vaccine-covered serotypes. Here, we report that a protein-based vaccine, a fusion construct of epitopes of CbpA to pneumolysin toxoid, confers effective protection against pneumococcal acute otitis media for non-PCV-13 serotypes and enhances protection for PCV-13 serotypes when coadministered with PCV-13. Having cross-reactive epitopes, the fusion protein also induces potent antibody responses against nontypeable Haemophilus influenzae and S. pneumoniae, engendering protection against acute otitis media caused by emerging unencapsulated otopathogens. These data suggest that augmenting capsule-based vaccination with conserved, cross-reactive protein-based vaccines broadens and enhances protection against acute otitis media.


Subject(s)
Antibodies, Bacterial/biosynthesis , Haemophilus Infections/prevention & control , Haemophilus influenzae/immunology , Otitis Media/prevention & control , Pneumococcal Vaccines/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Streptococcus pneumoniae/immunology , Acute Disease , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Cross Protection , Cross Reactions , Female , Gene Expression , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus influenzae/drug effects , Haemophilus influenzae/pathogenicity , Humans , Immunogenicity, Vaccine , Mice , Mice, Inbred BALB C , Otitis Media/immunology , Otitis Media/microbiology , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity , Streptolysins/biosynthesis , Streptolysins/genetics , Toxoids/biosynthesis , Toxoids/genetics , Vaccination , Vaccines, Synthetic
11.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31308086

ABSTRACT

As important players in the host defense system, commensal microbes and the microbiota influence multiple aspects of host physiology. Bordetella pertussis infection is highly contagious among humans. However, the roles of the microbiota in B. pertussis pathogenesis are poorly understood. Here, we show that antibiotic-mediated depletion of the microbiota results in increased susceptibility to B. pertussis infection during the early stage. The increased susceptibility was associated with a marked impairment of the systemic IgG, IgG2a, and IgG1 antibody responses to B. pertussis infection after antibiotic treatment. Furthermore, the microbiota impacted the short-lived plasma cell responses as well as the recall responses of memory B cells to B. pertussis infection. Finally, we found that the dysbiosis caused by antibiotic treatment affects CD4+ T cell generation and PD-1 expression on CD4+ T cells and thereby perturbs plasma cell differentiation. Our results have revealed the importance of commensal microbes in modulating host immune responses to B. pertussis infection and support the possibility of controlling the severity of B. pertussis infection in humans by manipulating the microbiota.


Subject(s)
Bordetella pertussis/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity, Humoral , Symbiosis/immunology , Whooping Cough/immunology , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/classification , Bacteroidetes/classification , Bacteroidetes/drug effects , Bacteroidetes/growth & development , Bacteroidetes/immunology , Bordetella pertussis/growth & development , Bordetella pertussis/pathogenicity , Dysbiosis/microbiology , Dysbiosis/physiopathology , Female , Firmicutes/classification , Firmicutes/drug effects , Firmicutes/growth & development , Firmicutes/immunology , Gastrointestinal Microbiome/drug effects , Immunity, Innate , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Metronidazole/pharmacology , Mice , Mice, Inbred BALB C , Neomycin/pharmacology , Proteobacteria/classification , Proteobacteria/drug effects , Proteobacteria/growth & development , Proteobacteria/immunology , Symbiosis/drug effects , Vancomycin/pharmacology , Whooping Cough/microbiology , Whooping Cough/physiopathology
12.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31331960

ABSTRACT

In this study, a novel recombinant attenuated Yersinia pseudotuberculosis PB1+ strain (χ10069) engineered with ΔyopK ΔyopJ Δasd triple mutations was used to deliver a Y. pestis fusion protein, YopE amino acid 1 to 138-LcrV (YopENt138-LcrV), to Swiss Webster mice as a protective antigen against infections by yersiniae. χ10069 bacteria harboring the pYA5199 plasmid constitutively synthesized the YopENt138-LcrV fusion protein and secreted it via the type 3 secretion system (T3SS) at 37°C under calcium-deprived conditions. The attenuated strain χ10069(pYA5199) was manifested by the establishment of controlled infection in different tissues without developing conspicuous signs of disease in histopathological analysis of microtome sections. A single-dose oral immunization of χ10069(pYA5199) induced strong serum antibody titers (log10 mean value, 4.2), secretory IgA in bronchoalveolar lavage (BAL) fluid from immunized mice, and Yersinia-specific CD4+ and CD8+ T cells producing high levels of tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), and interleukin 2 (IL-2), as well as IL-17, in both lungs and spleens of immunized mice, conferring comprehensive Th1- and Th2-mediated immune responses and protection against bubonic and pneumonic plague challenges, with 80% and 90% survival, respectively. Mice immunized with χ10069(pYA5199) also exhibited complete protection against lethal oral infections by Yersinia enterocolitica WA and Y. pseudotuberculosis PB1+. These findings indicated that χ10069(pYA5199) as an oral vaccine induces protective immunity to prevent bubonic and pneumonic plague, as well as yersiniosis, in mice and would be a promising oral vaccine candidate for protection against plague and yersiniosis for human and veterinary applications.


Subject(s)
Antibodies, Bacterial/biosynthesis , Immunoglobulin A/biosynthesis , Plague Vaccine/administration & dosage , Plague/prevention & control , Recombinant Fusion Proteins/administration & dosage , Yersinia pestis/drug effects , Yersinia pseudotuberculosis Infections/prevention & control , Yersinia pseudotuberculosis/drug effects , Administration, Oral , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cross Protection , Female , Gene Expression , Humans , Immunization , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Male , Mice , Plague/immunology , Plague/microbiology , Plague/mortality , Plague Vaccine/biosynthesis , Plague Vaccine/genetics , Plague Vaccine/immunology , Plasmids/chemistry , Plasmids/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccines, Synthetic , Yersinia pestis/immunology , Yersinia pestis/pathogenicity , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis/pathogenicity , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/microbiology , Yersinia pseudotuberculosis Infections/mortality
13.
Rheumatology (Oxford) ; 58(9): 1585-1596, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30877773

ABSTRACT

OBJECTIVES: We aimed to assess the safety and immunogenicity of a diphtheria/tetanus vaccine booster dose in three different patient groups with rheumatic diseases on a variety of immunosuppressive/immunomodulatory medications compared with healthy controls (HCs). METHODS: We conducted a multi-centre prospective cohort study in Switzerland. We enrolled patients with RA, axial SpA/PsA, vasculitis (Behçet's disease, ANCA-associated vasculitis) and HCs. Diphtheria/tetanus vaccination was administered according to the Swiss vaccination recommendations. Blood samples were drawn before vaccination, and 1 month and 3 months afterwards. Antibody concentrations against vaccine antigens were measured by ELISA. Immunogenicity was compared between patient and medication groups. A mixed model was applied for multivariate analysis. Missing data were dealt with using multiple imputation. RESULTS: Between January 2014 and December 2015, we enrolled 284 patients with rheumatic diseases (131 RA, 114 SpA/PsA, 39 vasculitis) and 253 HCs. Of the patients, 89% were on immunosuppressive/immunomodulatory medication. Three months post-vaccination 100% of HCs vs 98% of patients were protected against tetanus and 84% vs 73% against diphtheria. HCs and SpA/PsA patients had significantly higher responses than RA and vasculitis patients. Assessing underlying diseases and medications in a multivariate model, rituximab was the only factor negatively influencing tetanus immunogenicity, whereas only MTX treatment had a negative influence on diphtheria antibody responses. No vaccine-related serious adverse events were recorded. CONCLUSION: Diphtheria/tetanus booster vaccination was safe. Tetanus vaccination was immunogenic; the diphtheria component was less immunogenic. Vaccine responses were blunted by rituximab and MTX. TRIAL REGISTRATION: ClinicalTrials.gov, http://clinicaltrials.gov, Identifier: NCT01947465.


Subject(s)
Antibodies, Bacterial/biosynthesis , Diphtheria-Tetanus Vaccine/adverse effects , Immunogenicity, Vaccine/drug effects , Rheumatic Diseases/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Clostridium tetani/immunology , Corynebacterium diphtheriae/immunology , Diphtheria/prevention & control , Diphtheria-Tetanus Vaccine/immunology , Female , Humans , Immunization, Secondary , Immunogenicity, Vaccine/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Prospective Studies , Rheumatic Diseases/drug therapy , Tetanus/prevention & control , Vaccination , Young Adult
14.
Glycoconj J ; 36(5): 429-438, 2019 10.
Article in English | MEDLINE | ID: mdl-31230165

ABSTRACT

Enterococcus faecium (E. faecium) has emerged as one of today's leading causes of health care-associated infections that is difficult to treat with the available antibiotics. These pathogens produce capsular polysaccharides on the cell surface which play a significant role in adhesion, virulence and evasion. Therefore, we aimed at the identification and characterization of bacterial polysaccharide antigens which are central for the development of vaccine-based prophylactic approaches. The crude cell wall-associated polysaccharides from E. faecium, its mutant and complemented strains were purified and analyzed by a primary antibody raised against lipoteichoic acid (LTA) and diheteroglycan (DHG). The resistant E. faecium strains presumably possess novel capsular polysaccharides that allow them to avoid the evasion from opsonic killing. The E. faecium U0317 strain was very well opsonized by anti-U0317 (~95%), an antibody against the whole bacterial cell. The deletion mutant showed a significantly increased susceptibility to opsonophagocytic killing (90-95%) against the penicillin binding protein (anti-PBP-5). By comparison, in a mouse urinary tract and rat endocarditis infection model, respectively, there were no significant differences in virulence. In this study we explored the biological role of the capsule of E. faecium. Our findings showed that the U0317 strain is not only sensitive to anti-LTA but also to antibodies against other enterococcal surface proteins. Our findings demonstrate that polysaccharides capsule mediated-resistance to opsonophagocytosis. We also found that the capsular polysaccharides do not play an important role in bacterial virulence in urinary tract and infective endocarditis in vivo models.


Subject(s)
Antibodies, Bacterial/pharmacology , Antigens, Bacterial/isolation & purification , Cell Wall/chemistry , Enterococcus faecium/chemistry , Lipopolysaccharides/isolation & purification , Polysaccharides, Bacterial/isolation & purification , Teichoic Acids/isolation & purification , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Bacterial Capsules/chemistry , Bacterial Capsules/immunology , Cell Wall/immunology , Disease Models, Animal , Drug Resistance, Bacterial , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/microbiology , Enterococcus faecium/drug effects , Enterococcus faecium/immunology , Enterococcus faecium/pathogenicity , Female , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Opsonin Proteins/pharmacology , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/immunology , Penicillin-Binding Proteins/isolation & purification , Penicillin-Binding Proteins/pharmacology , Phagocytosis/drug effects , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/immunology , Primary Cell Culture , Rats , Rats, Wistar , Teichoic Acids/chemistry , Teichoic Acids/immunology , Teichoic Acids/pharmacology , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology
15.
J Immunol ; 199(7): 2547-2554, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28801359

ABSTRACT

Immune mechanisms responsible for pathogen clearance from the female reproductive tract (FRT) are incompletely defined; in particular, the contribution of lymphocyte trafficking to this process is unclear. CCR7-deficient mice have profoundly altered lymphocyte recirculation and display ectopic formation of lymphocyte aggregates within mucosal nonlymphoid tissues, including the FRT. In this study, we investigated how altered lymphocyte distribution in CCR7-deficient mice would affect host responses to Chlamydia muridarum within the reproductive tract. As expected, CCR7-deficient mice exhibited reduced lymphocyte trafficking to lymph nodes and a corresponding increase in T cell populations within the FRT. After intravaginal infection with Chlamydia, CCR7-deficient mice displayed markedly reduced Ag-specific CD4 T cell responses within the local draining iliac lymph nodes, yet robust Th1 and Th17 responses were prominent in the FRT. In addition, Chlamydia-specific Ab responses were dysregulated in CCR7-deficient mice, displaying an unexpected increase in the systemic IgA responses. Importantly, prominent mucosal immune responses in CCR7-deficient mice increased the efficiency of bacteria clearance from the FRT while reducing tissue-associated inflammation and pathology. Thus, increased numbers of lymphocytes within the FRT result in pathogen clearance with reduced immune-mediated pathology.


Subject(s)
Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia muridarum/immunology , Receptors, CCR7/immunology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/microbiology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Chlamydia muridarum/isolation & purification , Female , Immunoglobulin A/blood , Inflammation/microbiology , Lymph Nodes/immunology , Mice , Mice, Knockout , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Th1 Cells/immunology , Th17 Cells/immunology
16.
Proc Natl Acad Sci U S A ; 113(25): 6898-903, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27274071

ABSTRACT

Immunization strategies against commensal bacterial pathogens have long focused on eradicating asymptomatic carriage as well as disease, resulting in changes in the colonizing microflora with unknown future consequences. Additionally, current vaccines are not easily adaptable to sequence diversity and immune evasion. Here, we present a "smart" vaccine that leverages our current understanding of disease transition from bacterial carriage to infection with the pneumococcus serving as a model organism. Using conserved surface proteins highly expressed during virulent transition, the vaccine mounts an immune response specifically against disease-causing bacterial populations without affecting carriage. Aided by a delivery technology capable of multivalent surface display, which can be adapted easily to a changing clinical picture, results include complete protection against the development of pneumonia and sepsis during animal challenge experiments with multiple, highly variable, and clinically relevant pneumococcal isolates. The approach thus offers a unique and dynamic treatment option readily adaptable to other commensal pathogens.


Subject(s)
Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Animals , Antibodies, Bacterial/biosynthesis , Biofilms , Humans , Mice , Pneumococcal Infections/immunology , Pneumococcal Vaccines/immunology
17.
Proc Natl Acad Sci U S A ; 113(20): 5718-23, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27140614

ABSTRACT

A hallmark of Staphylococcus aureus disease in humans is persistent infections without development of protective immune responses. Infected patients generate VH3 plasmablast expansions and increased VH3 idiotype Ig; however, the mechanisms for staphylococcal modification of immune responses are not known. We report here that S. aureus-infected mice generate VH3 antibody expansions via a mechanism requiring MHC-restricted antigen presentation to CD4(+) T cells and staphylococcal protein A (SpA), a cell wall-anchored surface molecule that binds Fcγ and VH3 variant heavy chains of Ig. VH3 expansion occurred with peptidoglycan-linked SpA from the bacterial envelope but not with recombinant SpA, and optimally required five tandem repeats of its Ig-binding domains. Signaling via receptor-interacting serine/threonine protein kinase 2 (RIPK2) was essential for implementing peptidoglycan-linked SpA superantigen activity. VH3 clan IgG from S. aureus-infected or SpA-treated animals was not pathogen-specific, suggesting that SpA cross-linking of VH3 idiotype B-cell receptors and activation via attached peptidoglycan are the determinants of staphylococcal escape from adaptive immune responses.


Subject(s)
Antibodies, Bacterial/biosynthesis , Staphylococcal Infections/immunology , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Bacterial/blood , Antibody Formation , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptidoglycan/immunology , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
18.
Proc Natl Acad Sci U S A ; 113(11): 3042-7, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929342

ABSTRACT

The bacterial type 6 secretion system (T6SS) is a dynamic apparatus that translocates proteins between cells by a mechanism analogous to phage tail contraction. T6SS sheaths are cytoplasmic tubular structures composed of stable VipA-VipB (named for ClpV-interacting protein A and B) heterodimers. Here, the structure of the VipA/B sheath was exploited to generate immunogenic multivalent particles for vaccine delivery. Sheaths composed of VipB and VipA fused to an antigen of interest were purified from Vibrio cholerae or Escherichia coli and used for immunization. Sheaths displaying heterologous antigens generated better immune responses against the antigen and different IgG subclasses compared with soluble antigen alone. Moreover, antigen-specific antibodies raised against sheaths presenting Neisseria meningitidis factor H binding protein (fHbp) antigen were functional in a serum bactericidal assay. Our results demonstrate that multivalent nanoparticles based on the T6SS sheath represent a versatile scaffold for vaccine applications.


Subject(s)
Antigens/administration & dosage , Nanoparticles/chemistry , Type VI Secretion Systems/ultrastructure , Vaccines/administration & dosage , Acinetobacter/chemistry , Acinetobacter/genetics , Aminoacyltransferases/metabolism , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antigens/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cell Line , Cysteine Endopeptidases/metabolism , Dimerization , Drug Delivery Systems , Escherichia coli/chemistry , Female , Genes, Reporter , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Macrophages/physiology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Type VI Secretion Systems/chemistry , Vaccination , Vaccines/immunology , Vibrio cholerae/chemistry
19.
J Proteome Res ; 17(9): 2987-2994, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30095909

ABSTRACT

It is widely accepted that live vaccines elicit higher immune protection than inactivated vaccines. However, the mechanisms are largely unknown. Here, an array with 64 recombinant outer membrane proteins of Vibrio parahemolyticus was developed to explore antibody responses of live and inactivated V. parahemolyticus post immunization of the 8th, 12th, 16th and 20th day. Among the 64 outer membrane proteins, 28 elicited antibody generation. They were all detected in live vaccine-induced immunity but only 15 antibodies were found in inactivated vaccine-induced immunity. Passive immunization showed that higher percent survival was detected in live than inactivated vaccine-induced immunities. Active immunization indicated that out of 19 randomly selected outer membrane proteins, 5 stimulated immune protection against V. parahemolyticus infection. Among them, antibodies to VP2309 and VPA0526 were shared in mice immunized by live or inactivated vaccines, whereas antibodies to VPA0548, VPA1745, and VP1667 were only found in mice immunized by live vaccine. In addition, live V. parahemolyticus stimulated earlier antibody response than inactivated bacteria. These results indicate that not all of the outer membrane proteins elicited antibody responses when they work together in the form of live or inactivated bacteria; live vaccine elicits more protective antibodies, which contribute to higher immune protection in live vaccine than inactivated vaccine. Notably, the recombinant proteins might be different from those separated from live bacteria, and they might be different in their immunogenic potencies.


Subject(s)
Antibodies, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Immunity, Humoral/drug effects , Vibrio Infections/prevention & control , Animals , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Hot Temperature , Immune Sera/administration & dosage , Immunization, Passive/methods , Immunogenicity, Vaccine , Mice , Protein Array Analysis , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Survival Analysis , Vaccines, Inactivated , Vibrio Infections/immunology , Vibrio Infections/microbiology , Vibrio Infections/mortality , Vibrio parahaemolyticus/drug effects , Vibrio parahaemolyticus/growth & development , Vibrio parahaemolyticus/immunology , Zebrafish
20.
Article in English | MEDLINE | ID: mdl-29133571

ABSTRACT

The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.


Subject(s)
Anthrax , Anti-Bacterial Agents , Antibodies, Monoclonal , Antitoxins , Bacillus anthracis , Levofloxacin , Respiratory Tract Infections , Spores, Bacterial , Animals , Female , Male , Rabbits , Anthrax/immunology , Anthrax/microbiology , Anthrax/mortality , Anthrax/prevention & control , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/biosynthesis , Antibodies, Monoclonal/pharmacology , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Antitoxins/pharmacology , Bacillus anthracis/drug effects , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/blood , Bacterial Toxins/immunology , Drug Therapy, Combination , Immunization, Passive/methods , Immunoglobulin G/biosynthesis , Immunologic Memory/drug effects , Levofloxacin/pharmacology , Random Allocation , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Respiratory Tract Infections/prevention & control , Spores, Bacterial/drug effects , Spores, Bacterial/immunology , Spores, Bacterial/pathogenicity , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL