Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 5.580
1.
Front Immunol ; 12: 753371, 2021.
Article En | MEDLINE | ID: mdl-34721427

Many pathogens enter the host via the gut, causing disease in animals and humans. A robust intestinal immune response is necessary to protect the host from these gut pathogens. Despite being best suited for eliciting intestinal immunity, oral vaccination remains a challenge due to the gastrointestinal environment, a poor uptake of vaccine antigens by the intestinal epithelium and the tolerogenic environment pervading the gut. To improve uptake, efforts have focused on targeting antigens towards the gut mucosa. An interesting target is aminopeptidase N (APN), a conserved membrane protein present on small intestinal epithelial cells shown to mediate epithelial transcytosis. Here, we aimed to further optimize this oral vaccination strategy in a large animal model. Porcine APN-specific monoclonal antibodies were generated and the most promising candidate in terms of epithelial transcytosis was selected to generate antibody fusion constructs, comprising a murine IgG1 or porcine IgA backbone and a low immunogenic antigen: the F18-fimbriated E. coli tip adhesin FedF. Upon oral delivery of these recombinant antibodies in piglets, both mucosal and systemic immune responses were elicited. The presence of the FedF antigen however appeared to reduce these immune responses. Further analysis showed that F18 fimbriae were able to disrupt the antigen presenting capacity of intestinal antigen presenting cells, implying potential tolerogenic effects of FedF. Altogether, these findings show that targeted delivery of molecules to epithelial aminopeptidase N results in their transcytosis and delivery to the gut immune systems. The results provide a solid foundation for the development of oral subunit vaccines to protect against gut pathogens.


Adhesins, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , CD13 Antigens/immunology , Escherichia coli Proteins/immunology , Immunoconjugates/immunology , Immunoglobulin A/biosynthesis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Swine/immunology , Transcytosis , Vaccines, Synthetic/immunology , Adhesins, Bacterial/administration & dosage , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/administration & dosage , Antibody Affinity , Antigen-Presenting Cells/immunology , Antigens, Bacterial/administration & dosage , CD13 Antigens/physiology , Enterotoxigenic Escherichia coli/immunology , Epithelial Cells/metabolism , Escherichia coli Proteins/administration & dosage , Female , Fimbriae, Bacterial/immunology , Immunoconjugates/administration & dosage , Immunoglobulin A/administration & dosage , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Intestine, Small/enzymology , Mice , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Transcytosis/physiology , Vaccination/veterinary
2.
Front Immunol ; 12: 757811, 2021.
Article En | MEDLINE | ID: mdl-34745131

Induction of broadly neutralizing antibodies (bNAbs) is a major goal for HIV vaccine development. HIV envelope glycoprotein (Env)-specific bNAbs isolated from HIV-infected individuals exhibit substantial somatic hypermutation and correlate with T follicular helper (Tfh) responses. Using the VC10014 DNA-protein co-immunization vaccine platform consisting of gp160 plasmids and gp140 trimeric proteins derived from an HIV-1 infected subject that developed bNAbs, we determined the characteristics of the Env-specific humoral response in vaccinated rhesus macaques in the context of CD4+ T cell depletion. Unexpectedly, both CD4+ depleted and non-depleted animals developed comparable Tier 1 and 2 heterologous HIV-1 neutralizing plasma antibody titers. There was no deficit in protection from SHIV challenge, no diminution of titers of HIV Env-specific cross-clade binding antibodies, antibody dependent cellular phagocytosis, or antibody-dependent complement deposition in the CD4+ depleted animals. These collective results suggest that in the presence of diminished CD4+ T cell help, HIV neutralizing antibodies were still generated, which may have implications for developing effective HIV vaccine strategies.


AIDS Vaccines , Broadly Neutralizing Antibodies/biosynthesis , HIV Antibodies/biosynthesis , Macaca mulatta/immunology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , Broadly Neutralizing Antibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Female , Germinal Center/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp160/immunology , HIV-1/immunology , Immunization, Secondary , Male , Phagocytosis , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vaccine Development , Vaccines, Synthetic , Viral Load , env Gene Products, Human Immunodeficiency Virus/immunology
3.
Front Immunol ; 12: 712632, 2021.
Article En | MEDLINE | ID: mdl-34335629

Lymphotoxin beta receptor (LTßR) is a promising therapeutic target in autoimmune and infectious diseases as well as cancer. Mice with genetic inactivation of LTßR display multiple defects in development and organization of lymphoid organs, mucosal immune responses, IgA production and an autoimmune phenotype. As these defects are imprinted in embryogenesis and neonate stages, the impact of LTßR signaling in adulthood remains unclear. Here, to overcome developmental defects, we generated mice with inducible ubiquitous genetic inactivation of LTßR in adult mice (iLTßRΔ/Δ mice) and redefined the role of LTßR signaling in organization of lymphoid organs, immune response to mucosal bacterial pathogen, IgA production and autoimmunity. In spleen, postnatal LTßR signaling is required for development of B cell follicles, follicular dendritic cells (FDCs), recruitment of neutrophils and maintenance of the marginal zone. Lymph nodes of iLTßRΔ/Δ mice were reduced in size, lacked FDCs, and had disorganized subcapsular sinus macrophages. Peyer`s patches were smaller in size and numbers, and displayed reduced FDCs. The number of isolated lymphoid follicles in small intestine and colon were also reduced. In contrast to LTßR-/- mice, iLTßRΔ/Δ mice displayed normal thymus structure and did not develop signs of systemic inflammation and autoimmunity. Further, our results suggest that LTßR signaling in adulthood is required for homeostasis of neutrophils, NK, and iNKT cells, but is dispensable for the maintenance of polyclonal IgA production. However, iLTßRΔ/Δ mice exhibited an increased sensitivity to C. rodentium infection and failed to develop pathogen-specific IgA responses. Collectively, our study uncovers new insights of LTßR signaling in adulthood for the maintenance of lymphoid organs, neutrophils, NK and iNKT cells, and IgA production in response to mucosal bacterial pathogen.


Aging/immunology , Lymphoid Tissue/immunology , Lymphotoxin beta Receptor/physiology , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Autoimmunity , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Citrobacter rodentium/immunology , Crosses, Genetic , Gene Expression Regulation, Developmental , Homeostasis/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Inflammation , Killer Cells, Natural/immunology , Lymphoid Tissue/cytology , Lymphotoxin beta Receptor/biosynthesis , Lymphotoxin beta Receptor/deficiency , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , Neutrophils/immunology , Sequence Deletion , Specific Pathogen-Free Organisms , Splenomegaly/immunology
4.
Front Immunol ; 12: 696003, 2021.
Article En | MEDLINE | ID: mdl-34177963

Antiviral, antibacterial, and antiparasitic drugs and vaccines are essential to maintaining the health of humans and animals. Yet, their production can be slow and expensive, and efficacy lost once pathogens mount resistance. Chicken immunoglobulin Y (IgY) is a highly conserved homolog of human immunoglobulin G (IgG) that has shown benefits and a favorable safety profile, primarily in animal models of human infectious diseases. IgY is fast-acting, easy to produce, and low cost. IgY antibodies can readily be generated in large quantities with minimal environmental harm or infrastructure investment by using egg-laying hens. We summarize a variety of IgY uses, focusing on their potential for the detection, prevention, and treatment of human and animal infections.


Antibodies, Neutralizing/therapeutic use , Bacterial Infections/drug therapy , Chickens/immunology , Immunoassay , Immunoglobulins/therapeutic use , Parasitic Diseases/drug therapy , Virus Diseases/drug therapy , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Antibody Formation , Antibody Specificity , Bacterial Infections/diagnosis , Bacterial Infections/immunology , Bacterial Infections/virology , Humans , Immunoglobulins/biosynthesis , Immunoglobulins/immunology , Parasitic Diseases/diagnosis , Parasitic Diseases/immunology , Parasitic Diseases/virology , Predictive Value of Tests , Virus Diseases/diagnosis , Virus Diseases/immunology , Virus Diseases/virology
5.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article En | MEDLINE | ID: mdl-34001624

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.


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
6.
Microbes Infect ; 23(4-5): 104801, 2021.
Article En | MEDLINE | ID: mdl-33582283

Acinetobacter baumannii is an important nosocomial pathogen. BamA is a protein that belongs to a complex responsible for organizing the proteins on the bacterial outer membrane. In this work, we aimed to evaluate murine immune responses to BamA recombinant protein (rAbBamA) from A. baumannii in an animal model of infection, and to assess cross-reactivity of this target for the development of anti-A. baumannii vaccines or diagnostics. Immunization of mice with rAbBamA elicited high antibody titers and antibody recognition of native A. baumannii BamA. Immunofluorescence also detected binding to the bacterial surface. After challenge, immunized mice demonstrated a 40% survival increase and better bacterial clearance in kidneys. Immunoblot of anti-rAbBamA against other medically relevant bacteria showed binding to proteins of approximately 35 kDa in Klebsiella pneumoniae and Escherichia coli lysates, primarily identified as OmpA and OmpC, respectively. Altogether, our data show that anti-rAbBamA antibodies provide a protective response against A. baumannii infection in mice. However, the response elicited by immunization with rAbBamA is not completely specific to A. baumannii. Although a broad-spectrum vaccine that protects against various pathogens is an appealing strategy, antibody reactivity against the human microbiota is undesired. In fact, immunization with rAbBamA produced noticeable effects on the gut microbiota. However, the changes elicited were small and non-specific, given that no significant changes in the abundance of Proteobacteria were observed. Overall, rAbBamA is a promising target, but specificity must be considered in the development of immunological tools against A. baumannii.


Acinetobacter Infections/prevention & control , Acinetobacter baumannii/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Acinetobacter baumannii/immunology , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/chemistry , Cloning, Molecular , DNA, Bacterial/chemistry , Feces/chemistry , Female , Gastrointestinal Microbiome , Gene Expression Regulation, Bacterial , Mice , Mice, Inbred C57BL , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/chemistry , Recombinant Proteins/immunology
7.
Arch Microbiol ; 203(5): 2719-2725, 2021 Jul.
Article En | MEDLINE | ID: mdl-33606039

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.


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
8.
Carbohydr Polym ; 255: 117475, 2021 Mar 01.
Article En | MEDLINE | ID: mdl-33436239

Extraintestinal pathogenic Escherichia coli (ExPEC) has presented a major clinical infection emerged in the past decades. O-polysaccharide (OPS)-based glycoconjugate vaccines produced using the bacterial glycosylation machinery can be utilized to confer protection against such infection. However, constructing a low-cost microbial cell factory for high-efficient production of OPS-based glycoconjugate vaccines remains challenging. Here, we engineered a glyco-optimized chassis strain by reprogramming metabolic network. The yield was enhanced to 38.6 mg L-1, the highest level reported so far. MS analysis showed that designed glycosylation sequon was modified by target polysaccharide with high glycosylation efficiency of 90.7 % and 76.7 % for CTB-O5 and CTB-O7, respectively. The glycoconjugate vaccines purified from this biosystem elicited a marked increase in protection against ExPEC infection in mouse model, compared to a non-optimized system. The glyco-optimized platform established here is broadly suitable for polysaccharide-based conjugate production against ExPEC and other surface-polysaccharide-producing pathogens.


Cell Engineering/methods , Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/biosynthesis , Extraintestinal Pathogenic Escherichia coli/immunology , Glycoconjugates/biosynthesis , O Antigens/biosynthesis , Amino Acid Sequence , Animals , Animals, Outbred Strains , Antibodies, Bacterial/biosynthesis , Carbohydrate Sequence , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/mortality , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/genetics , Escherichia coli Vaccines/immunology , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Female , Glycoconjugates/administration & dosage , Glycoconjugates/genetics , Glycoconjugates/immunology , Glycosylation , Immunization , Immunogenicity, Vaccine , Immunoglobulin G/biosynthesis , Metabolic Networks and Pathways/genetics , Mice , O Antigens/genetics , O Antigens/immunology , Plasmids/chemistry , Plasmids/metabolism , Survival Analysis , Vaccines, Conjugate
9.
Int Immunopharmacol ; 89(Pt B): 107100, 2020 Dec.
Article En | MEDLINE | ID: mdl-33091812

The role of nontreponemal antibodies in the Treponema pallidum infection course is unclear. We investigated the effect of immunization with nontreponemal antigen on T. pallidum-challenged rabbits. Nontreponemal antigen was injected intravenously into rabbits in the nontreponemal group (n = 12) to elicit antibodies (≥1:64), and normal saline-injected rabbits were used as controls (n = 12). Then, rabbits were challenged with 106T. pallidum per site along their back. Lesion development was observed, and the injection sites were biopsied for mRNA analysis every week. Six rabbits from both groups were euthanized at 14 d and 28 d. The popliteal lymph nodes were extracted to assess infectivity using a rabbit infectivity test. The maximum lesion diameters were not different between the two groups (12.4 ± 0.9 mm in the nontreponemal group vs. 12.5 ± 1.0 mm in the control group, P = 0.386), but the time to maximum diameter appearance was delayed by approximately 4 d in the nontreponemal group (14.4 ± 1.6 d vs. 10.8 ± 1.9 d, P = 0.000). There were no significant differences in the proportions of lesions (58/60 (96.7%) vs. 59/60 (98.3%), P = 0.500) or ulcers (55/60 (91.7%) vs. 57/60 (95.0%), P = 0.359) between the two groups. An ulcer development delay of 5 d was observed in the nontreponemal group (19.3 ± 2.0 d vs. 14.0 ± 1.8 d, P = 0.000). IL-2 and IFN-γ mRNA expression in the nontreponemal group was significantly higher than that in the control group at 7 d and 14 d post-challenge. flaA mRNA expression and the rabbit infectivity test positive rate were not different between the two groups. Immunization with nontreponemal antigen altered the syphilis course in rabbits, resulting in delayed maximal lesion diameter and ulcer development, but it could not inhibit the spread of T. pallidum from primary lesion sites to viscera.


Antigens, Bacterial/immunology , Immune Sera/immunology , Immunization/methods , Syphilis/prevention & control , Treponema pallidum/immunology , Administration, Intravenous , Animals , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/administration & dosage , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Flagellin/blood , Flagellin/drug effects , Flagellin/genetics , Humans , Immune Sera/administration & dosage , Injections, Intradermal , Liver/drug effects , Liver/microbiology , Lymph Nodes/transplantation , Male , Rabbits , Skin Diseases, Infectious/microbiology , Skin Diseases, Infectious/prevention & control , Spleen/drug effects , Spleen/microbiology , Syphilis/blood , Testis/drug effects , Testis/microbiology , Treponema pallidum/drug effects , Ulcer/microbiology , Ulcer/prevention & control
10.
Bull Exp Biol Med ; 169(4): 474-477, 2020 Aug.
Article En | MEDLINE | ID: mdl-32910375

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.


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
11.
Front Immunol ; 11: 1069, 2020.
Article En | MEDLINE | ID: mdl-32655550

Acinetobacter baumannii (A. baumannii) is becoming a common global concern due to the emergence of multi-drug or pan-drug resistant strains. Confronting the issue of antimicrobial resistance by developing vaccines against the resistant pathogen is becoming a common strategy. In this study, different methods for preparing A. baumannii outer membrane vesicles (AbOMVs) vaccines were developed. sOMV (spontaneously released AbOMV) was extracted from the culture supernatant, while SuOMV (sucrose-extracted AbOMV) and nOMV (native AbOMV) were prepared from the bacterial cells. Three AbOMVs exhibited significant differences in yield, particle size, protein composition, and LPS/DNA content. To compare the protective efficacy of the three AbOMVs, groups of mice were immunized either intramuscularly or intranasally with each AbOMV. Vaccination via both routes conferred significant protection against lethal and sub-lethal A. baumannii challenge. Moreover, intranasal vaccination provided more robust protection, which may be attributed to the induction of significant sIgA response in mucosal sites. Among the three AbOMVs, SuOMV elicited the highest level of protective immunity against A. baumannii infection, whether intramuscular or intranasal immunization, which was characterized by the expression of the most profound specific serum IgG or mucosal sIgA. Taken together, the preparation method had a significant effect on the yield, morphology, and composition of AbOMVs, that further influenced the protective effect against A. baumannii infection.


Acinetobacter baumannii/immunology , Bacterial Vaccines/isolation & purification , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/pathogenicity , Acinetobacter baumannii/ultrastructure , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibody Specificity , Bacterial Outer Membrane/immunology , Bacterial Outer Membrane/ultrastructure , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/classification , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission
12.
Front Immunol ; 11: 1083, 2020.
Article En | MEDLINE | ID: mdl-32582177

Human cytomegalovirus (HCMV) infection has a profound effect on the human immune system, causing massive clonal expansion of CD8, and to a lesser extend CD4 T cells. The few human trials that have explored the effect of HCMV infection on responses to vaccination are conflicting, with some studies suggesting no effect whilst others suggest decreased or increased immune responses. Recent studies indicate substantial differences in overall immune system reactivity to vaccines based on age and sex, particularly cellular immunity. 225 nine-month old Gambian infants were immunized with diphtheria-tetanus-whole cell pertussis and/or measles vaccines. HCMV infection status was determined by the presence of CMV DNA by PCR of urine samples prior to vaccination. The effect of HCMV infection on either protective antibody immunity or vaccine-specific and overall cellular immune responses 4 weeks post-vaccination was determined, further stratified by sex. Tetanus toxoid-specific antibody responses were significantly lower in HCMV+ infants compared to their HCMV- counterparts, while pertussis, diphtheria and measles antibody responses were generally comparable between the groups. Responses to general T cell stimulation with anti-CD3/anti-CD28 as well as antigen-specific cytokine responses to purified protein derivative (PPD) were broadly suppressed in infants infected with HCMV but, perhaps surprisingly, there was only a minimal impact on antigen-specific cellular responses to vaccine antigens. There was evidence for subtle sex differences in the effects of HCMV infection, in keeping with the emerging evidence suggesting sex differences in homeostatic immunity and in responses to vaccines. This study reassuringly suggests that the high rates of HCMV infection in low income settings have little clinically significant impact on antibody and cellular responses to early life vaccines, while confirming the importance of sex stratification in such studies.


Cytomegalovirus Infections/immunology , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Measles Vaccine/immunology , Antibodies, Bacterial/biosynthesis , Antibodies, Viral/biosynthesis , Cohort Studies , Cytokines/blood , Female , Gambia , Humans , Immune Tolerance , Immunity, Cellular , Immunoglobulin G/blood , Infant , Male , Prospective Studies , Sex Characteristics , T-Lymphocytes/immunology , Tetanus Toxoid/immunology
13.
Infect Immun ; 88(8)2020 07 21.
Article En | MEDLINE | ID: mdl-32393506

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.


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
14.
Bull Exp Biol Med ; 168(4): 485-487, 2020 Feb.
Article En | MEDLINE | ID: mdl-32146631

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.


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
15.
Life Sci ; 248: 117444, 2020 May 01.
Article En | MEDLINE | ID: mdl-32084433

AIMS: Nonhuman primates have been used to investigate pathogenic mechanisms and evaluate immune responses following Chlamydia trachomatis inoculation. This study aimed to systemically profile antibody responses to C. trachomatis infection in nonhuman primates. MATERIALS AND METHODS: Sera were obtained from 4 pig-tailed and 8 long-tailed macaques which were intravaginally or ocularly infected with live C. trachomatis organisms, and analyzed by C. trachomatis proteome array of antigens. KEY FINDINGS: The sera from 12 macaques recognized total 172 C. trachomatis antigens. While 84 antigens were recognized by pig-tailed macaques intravaginally infected with serovar D strain, 125 antigens were recognized by long-tailed macaques ocularly infected with serovar A, and 37 antigens were recognized by both. Ocular inoculation with virulent A2497 strain induced antibodies to more antigens. Among the antigens uniquely recognized by A2497 strain infected macaques, outer membrane complex B antigen (OmcB) induced robust antibody response. Although macaques infected by less virulent A/HAR-13 strain failed to develop antibodies to OmcB, reinfection by A2497 strain induced high levels of antibodies to OmcB. SIGNIFICANCE: Proteome array has revealed a correlation of chlamydial infection invasiveness with chlamydial antigen immunogenicity, and identified antibody responses to OmcB potentially as biomarkers for invasive infection with C. trachomatis.


Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/blood , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Reproductive Tract Infections/immunology , Trachoma/immunology , Animals , Antibodies, Bacterial/classification , Antigens, Bacterial/classification , Bacterial Outer Membrane Proteins/blood , Chlamydia Infections/blood , Chlamydia Infections/microbiology , Chlamydia trachomatis/pathogenicity , Eye/immunology , Eye/microbiology , Female , Immune Sera/chemistry , Macaca fascicularis , Macaca nemestrina , Male , Protein Array Analysis , Proteome/chemistry , Proteome/immunology , Reproductive Tract Infections/blood , Reproductive Tract Infections/microbiology , Trachoma/blood , Trachoma/microbiology , Vagina/immunology , Vagina/microbiology
16.
Eur J Immunol ; 50(5): 676-684, 2020 05.
Article En | MEDLINE | ID: mdl-32026472

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.


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
17.
Med Microbiol Immunol ; 209(2): 125-137, 2020 Apr.
Article En | MEDLINE | ID: mdl-31811379

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.


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
18.
Sci Rep ; 9(1): 18404, 2019 12 05.
Article En | MEDLINE | ID: mdl-31804525

Typhoid fever caused by Salmonella enterica serovar Typhi (S.Typhi) continues to be a major problem, especially in developing countries. Due to the rapid emergence of multi-drug-resistant (MDR) strains, which limits the efficacy of conventional antibiotics as well as problems associated with the existing vaccines, efforts are being made to develop effective prophylactic agents. CdtB subunit of typhoid toxin was selected for assessing its vaccine potential due to its high conservation throughout the Typhi strains. In-vitro assessment of DNase activity of cloned and purified CdtB protein showed a significant decrease in the band intensity of DNA. The measure of metabolic activity and morphological alterations assessed using different cell lines in the presence of CdtB protein showed no significant signs of toxicity. These observations were further strengthened by cell cycle analysis, assessed by flow cytometry. Keeping these observations in mind, the immunoprotective potential of CdtB was assessed using S.Typhi induced mouse peritonitis model. A significant titer of IgG antibodies (>128000) against CdtB protein was recorded in the immunized mice by enzyme-linked immunosorbent assay (ELISA), which was also validated by immunoblotting. Active immunization with the protein protected 75% mice against a lethal dose of S.Typhi Ty2. The data indicated a significant (up to 5 log) reduction in the bacterial load in the spleen and liver of immunized-infected mice compared to control (unimmunized-infected) mice which might have resulted in the modulation of histoarchitecture of spleen and liver and the levels of cytokines (IL-6, TNF-α and IL-10) production; thereby indicating the effectiveness of the subunit. The observations deduced from the study give the proof of concept of immunogenic potential of protein. However, further studies involving the immunoreactivity of CdtB with the statistically significant number of sera samples obtained from the human patients would be helpful in establishing the relevance of CdtB protein in humans and for making the strategies to develop it as an effective vaccine candidate.


Antibodies, Bacterial/biosynthesis , Bacterial Toxins/administration & dosage , Immunoglobulin G/biosynthesis , Peritonitis/prevention & control , Salmonella typhi/drug effects , Typhoid Fever/prevention & control , Typhoid-Paratyphoid Vaccines/administration & dosage , Animals , Bacterial Toxins/immunology , Colony Count, Microbial , Disease Models, Animal , Gene Expression , Humans , Immunization/methods , Immunogenicity, Vaccine , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Liver/drug effects , Liver/immunology , Liver/microbiology , Mice , Peritonitis/immunology , Peritonitis/microbiology , Peritonitis/mortality , Salmonella typhi/immunology , Salmonella typhi/pathogenicity , Spleen/drug effects , Spleen/immunology , Spleen/microbiology , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Typhoid Fever/immunology , Typhoid Fever/microbiology , Typhoid Fever/mortality , Typhoid-Paratyphoid Vaccines/immunology
19.
Sci Rep ; 9(1): 17586, 2019 11 26.
Article En | MEDLINE | ID: mdl-31772196

The most commonly used biopesticides to control agricultural, forest and insect vectors of human diseases are derived from the bacterium Bacillus thuringiensis, which begins to produce Cry and Cyt insecticidal proteins during the onset of the sporulation phase. Some B. thuringiensis strains also produce S-layer proteins that are toxic to certain pests. S-layer proteins are the most abundant proteins in bacteria and archaea. This proteins' key trait to design high performace processes for mass production is their continuous expression during the vegetative phase, unlike Cry and Cyt, which are restricted to the sporulation phase. In this work, a S-layer protein expressed by the GP543 strain of B. thuringiensis that is toxic to the cattle tick Rhipicephalus microplus was mass produced using the batch culture fermentation technique. In addition, the spore-protein complex showed a mortality rate of 75% with a dose of 300 µg·mL-1 on adult females of R. microplus after fourteen days. The lethal concentration 50 was 69.7 µg·mL-1. The treatment also caused a decrease of 13% in the weight of the mass of oviposited eggs with 200 µg·mL-1 of the spore-protein complex and inhibition of the hatching of eggs from 80 to 92%. Therefore, this could be a good option for controlling this parasite. The advantages of S-layer protein synthesis are focused on the production of a new generation of proteins in pest control. This is the first report on the mass production of an S-layer protein that is responsible for toxicity.


Bacillus thuringiensis/chemistry , Bacteriological Techniques/methods , Biological Control Agents/isolation & purification , Industrial Microbiology/methods , Membrane Glycoproteins/isolation & purification , Rhipicephalus/drug effects , Animals , Antibodies, Bacterial/biosynthesis , Bacillus thuringiensis/drug effects , Bacillus thuringiensis/growth & development , Bacillus thuringiensis/metabolism , Biological Control Agents/toxicity , Biomass , Bioreactors , Cattle , Culture Media/pharmacology , Female , Fermentation , Membrane Glycoproteins/immunology , Membrane Glycoproteins/toxicity , Oviposition/drug effects , Ovum/drug effects , Rabbits , Spores, Bacterial
20.
Infect Immun ; 87(10)2019 10.
Article En | MEDLINE | ID: mdl-31308086

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.


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
...