Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Am J Respir Crit Care Med ; 210(1): 35-46, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38754132

ABSTRACT

Rationale: Pseudomonas aeruginosa infection is associated with worse outcomes in bronchiectasis. Impaired neutrophil antimicrobial responses contribute to bacterial persistence. Gremubamab is a bivalent, bispecific monoclonal antibody targeting Psl exopolysaccharide and the type 3 secretion system component PcrV. Objectives: This study evaluated the efficacy of gremubamab to enhance killing of P. aeruginosa by neutrophils from patients with bronchiectasis and to prevent P. aeruginosa-associated cytotoxicity. Methods: P. aeruginosa isolates from a global bronchiectasis cohort (n = 100) underwent whole-genome sequencing to determine target prevalence. Functional activity of gremubamab against selected isolates was tested in vitro and in vivo. Patients with bronchiectasis (n = 11) and control subjects (n = 10) were enrolled, and the effect of gremubamab in peripheral blood neutrophil opsonophagocytic killing (OPK) assays against P. aeruginosa was evaluated. Serum antibody titers to Psl and PcrV were determined (n = 30; 19 chronic P. aeruginosa infection, 11 no known P. aeruginosa infection), as was the effect of gremubamab treatment in OPK and anti-cytotoxic activity assays. Measurements and Main Results: Psl and PcrV were conserved in isolates from chronically infected patients with bronchiectasis. Seventy-three of 100 isolates had a full psl locus, and 99 of 100 contained the pcrV gene, with 20 distinct full-length PcrV protein subtypes identified. PcrV subtypes were successfully bound by gremubamab and the monoclonal antibody-mediated potent protective activity against tested isolates. Gremubamab increased bronchiectasis patient neutrophil-mediated OPK (+34.6 ± 8.1%) and phagocytosis (+70.0 ± 48.8%), similar to effects observed in neutrophils from control subjects (OPK, +30.1 ± 7.6%). No evidence of competition between gremubamab and endogenous antibodies was found, with protection against P. aeruginosa-induced cytotoxicity and enhanced OPK demonstrated with and without addition of patient serum. Conclusions: Gremubamab enhanced bronchiectasis patient neutrophil phagocytosis and killing of P. aeruginosa and reduced virulence.


Subject(s)
Antibodies, Bispecific , Bronchiectasis , Neutrophils , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Bronchiectasis/immunology , Bronchiectasis/microbiology , Pseudomonas aeruginosa/immunology , Neutrophils/immunology , Neutrophils/drug effects , Antibodies, Bispecific/therapeutic use , Antibodies, Bispecific/pharmacology , Female , Male , Pseudomonas Infections/immunology , Middle Aged , Aged , Adult , Antigens, Bacterial , Bacterial Toxins , Pore Forming Cytotoxic Proteins
2.
Mucosal Immunol ; 16(5): 593-605, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37392972

ABSTRACT

Young infants frequently experience respiratory tract infections, yet vaccines designed to provide mucosal protection are lacking. Localizing pathogen-specific cellular and humoral immune responses to the lung could provide improved immune protection. We used a well-characterized murine model of respiratory syncytial virus (RSV) to study the development of lung-resident memory T cells (TRM) in neonatal compared to adult mice. We demonstrated that priming with RSV during the neonatal period failed to retain RSV-specific clusters of differentiation (CD8) TRM 6 weeks post infection, in contrast to priming during adulthood. The reduced development of RSV-specific TRM was associated with poor acquisition of two key markers of tissue residence: CD69 and CD103. However, by augmenting both innate immune activation and antigen exposure, neonatal RSV-specific CD8 T cells increased expression of tissue-residence markers and were maintained in the lung at memory time points. Establishment of TRM correlated with more rapid control of the virus in the lungs upon reinfection. This is the first strategy to effectively establish RSV-specific TRM in neonates providing new insight into neonatal memory T cell development and vaccine strategies.

3.
Infect Immun ; 90(10): e0020322, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36069593

ABSTRACT

The Gram-negative pathogen Pseudomonas aeruginosa is a common cause of pneumonia in hospitalized patients. Its increasing antibiotic resistance and widespread occurrence present a pressing need for vaccines. We previously showed that a P. aeruginosa type III secretion system protein, PopB, elicits a strong Th17 response in mice after intranasal (IN) immunization and confers antibody-independent protection against pneumonia in mice. In the current study, we evaluated the immunogenicity and protective efficacy in mice of the combination of PopB (purified with its chaperone protein PcrH) and OprF/I, an outer membrane hybrid fusion protein, compared with immunization with the proteins individually either by the intranasal (IN) or subcutaneous (SC) routes. Our results show that after vaccination, a Th17 recall response from splenocytes was detected only in mice vaccinated with PopB/PcrH, either alone or in combination with OprF/I. Mice immunized with the combination of PopB/PcrH and OprF/I had enhanced protection in an acute lethal P. aeruginosa pneumonia model, regardless of vaccine route, compared with mice vaccinated with either alone or adjuvant control. Immunization generated IgG titers against the vaccine proteins and whole P. aeruginosa cells. Interestingly, none of these antisera had opsonophagocytic killing activity, but antisera from mice immunized with vaccines containing OprF/I, had the ability to block IFN-γ binding to OprF/I, a known virulence mechanism. Hence, vaccines combining PopB/PcrH with OprF/I that elicit functional antibodies lead to a broadly and potently protective vaccine against P. aeruginosa pulmonary infections.


Subject(s)
Pneumonia , Pseudomonas Infections , Mice , Animals , Pseudomonas Vaccines , Pseudomonas aeruginosa , Pseudomonas Infections/prevention & control , Th17 Cells , Type III Secretion Systems , Antibody Formation , Antibodies, Bacterial , Bacterial Proteins , Immunoglobulin G , Immune Sera
4.
Mil Med ; 186(Suppl 1): 76-81, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33499495

ABSTRACT

BACKGROUND: Respiratory viruses are an important cause of nonbattle injury disease and contribute to the top seven reasons for medical encounters. In the absence of vaccines that provide complete protection against these viruses, viral surveillance can identify disease burden and target virus-specific preventative measures. Influenza infection, in particular, has significant adverse effects on force readiness. METHODS: We tracked the frequency of 16 respiratory viruses at Walter Reed National Military Medical Center tested for during routine patient care using multiplex polymerase chain reaction and rapid antigen testing. We collected data on the date and location of the testing, as well as the age of the individual tested from two consecutive respiratory viral seasons. RESULTS: During the first year of data compilation (2017-2018), 2556 tests were performed; 342 (13.4%) were positive for influenza A and 119 (4.7%) were positive for influenza B. After influenza, the most common families of viruses identified were rhino/enterovirus (490 [19.2%]). During the second year (2018-2019), 4,458 tests were run; 564 (12.7%) were positive for influenza A and 35 (0.79%) were positive for influenza B, while rhino/enterovirus was identified in 690 (15.4%). Influenza peaked early during the 2017-2018 season and later during the 2018-2019 season. Importantly, during the 2017-2018 season, the vaccine was less effective for the H3N2 strain circulating that year and viral surveillance quickly identified a hospital-specific outbreak and a larger disease burden. This was in contrast to the 2018-2019 vaccine which exhibited higher effectiveness for circulating strains. CONCLUSION: Our data highlight the seasonality of respiratory viruses with a focus on influenza. By tracking respiratory viruses in Department of Defense communities, we may be able to predict when influenza may cause the greatest burden for distinct organizational regions and prescribe with greater precision preventative protocols by location, as well as rapidly determine vaccine efficacy. Our current data suggest that when vaccine strains are mismatched, rapid upfront targeting of antivirals may be warranted, but when the vaccine strains are better matched, late season peaks of disease may indicate waning immunity and should be monitored.


Subject(s)
Influenza, Human , Cost of Illness , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Public Health , Seasons
5.
Mucosal Immunol ; 13(2): 371-380, 2020 03.
Article in English | MEDLINE | ID: mdl-31797910

ABSTRACT

Insufficient T-cell responses contribute to the increased burden of viral respiratory disease in infancy. Neonatal dendritic cells (DCs) often provide defective activation of pathogen-specific T cells through mechanisms that are incompletely understood, which hinders vaccine design for this vulnerable age group. Enhancing our characterization of neonatal DC sub-specialization and function is therefore critical to developing their potential for immunomodulation of T-cell responses. In this study, we engineered respiratory syncytial virus (RSV) to express a model protein, ovalbumin, to track antigen-presenting DCs in vivo. We found that murine neonatal conventional DC1s (cDC1s) efficiently migrated and presented RSV-derived antigen, challenging the paradigm that neonatal DCs are globally immature. In a key observation, however, we discovered that during infection neonatal cDC1s presenting viral antigen were unable to upregulate costimulatory molecules in response to type I interferons (IFN-I), contributing to poor antiviral T-cell responses. Importantly, we showed that the deficient response to IFN-I was also exhibited by human neonatal cDC1s, independent of infection. These findings reveal a functionally distinct response to IFN-I by neonatal cDC1s that may leave young infants susceptible to viral infections, and provide a new target for exploration, in light of failed efforts to design neonatal RSV vaccines.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Infant, Newborn, Diseases/immunology , Interferon Type I/metabolism , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/physiology , T-Lymphocytes/immunology , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Female , Humans , Infant, Newborn , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
6.
PLoS One ; 7(6): e38733, 2012.
Article in English | MEDLINE | ID: mdl-22723880

ABSTRACT

Cell signaling for T-cell growth, differentiation, and apoptosis is initiated in the cholesterol-rich microdomains of the plasma membrane known as lipid rafts. Herein, we investigated whether enrichment of membrane cholesterol in lipid rafts affects antigen-specific CD4 T-helper cell functions. Enrichment of membrane cholesterol by 40-50% following squalene administration in mice was paralleled by an increased number of resting CD4 T helper cells in periphery. We also observed sensitization of the Th1 differentiation machinery through co-localization of IL-2Rα, IL-4Rα, and IL-12Rß2 subunits with GM1 positive lipid rafts, and increased STAT-4 and STAT-5 phosphorylation following membrane cholesterol enrichment. Antigen stimulation or CD3/CD28 polyclonal stimulation of membrane cholesterol-enriched, resting CD4 T-cells followed a path of Th1 differentiation, which was more vigorous in the presence of increased IL-12 secretion by APCs enriched in membrane cholesterol. Enrichment of membrane cholesterol in antigen-specific, autoimmune Th1 cells fostered their organ-specific reactivity, as confirmed in an autoimmune mouse model for diabetes. However, membrane cholesterol enrichment in CD4(+)Foxp3(+) T-reg cells did not alter their suppressogenic function. These findings revealed a differential regulatory effect of membrane cholesterol on the function of CD4 T-cell subsets. This first suggests that membrane cholesterol could be a new therapeutic target to modulate the immune functions, and second that increased membrane cholesterol in various physiopathological conditions may bias the immune system toward an inflammatory Th1 type response.


Subject(s)
Cholesterol/metabolism , Inflammation/immunology , Inflammation/metabolism , Membrane Microdomains/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Female , Gene Expression Regulation/drug effects , Inflammation/genetics , Male , Membrane Microdomains/drug effects , Mice , Mice, Transgenic , Protein Transport , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Signal Transduction , Squalene/administration & dosage , Squalene/pharmacology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
7.
Vaccine ; 28(45): 7319-30, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20832492

ABSTRACT

The T-regulatory (T-reg) cells restrict the T-cell functions in various viral infections including influenza infection. However little is known about the effect of T-regs in influenza vaccination. Herein, we found that immunization of BALB/c mice with a prototype of UV-inactivated influenza PR8/A/34 virus vaccine expanded the CD4(+)Foxp3(+) T-reg pool and fostered the development of virus-specific CD4(+)Foxp3(+) T-reg cells. Increasing the size of Foxp3(+) T-reg pool did not alter the primary PR8-specific B-cell response, but it did suppress the primary and memory PR8-specific T helper responses induced by vaccination. In contrast, the vaccination-induced T helper cell response was augmented in the absence of CD4(+)Foxp3(+) T-reg cells. Since CD4 T helper cells contribute to anti-influenza protection, therapeutic "quenching" of T-reg function prior to vaccination may enhance the efficacy of influenza vaccination.


Subject(s)
B-Lymphocytes/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/immunology , Immunity, Cellular , Immunologic Memory , Influenza A virus/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/immunology
8.
PLoS One ; 5(7): e11427, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20625402

ABSTRACT

BACKGROUND: Double negative CD3(+)4(-)8(-) TCR alphabeta splenic cells (DNCD3) can suppress the immune responses to allo and xenografts, infectious agents, tumors, and some autoimmune disorders. However, little is known about their role in autoimmune diabetes, a disease characterized by the reduction of insulin production subsequent to destruction of pancreatic beta-cells by a polyclonal population of self-reactive T-cells. Herein, we analyzed the function and phenotype of DNCD3 splenic cells in young NOD mice predisposed to several autoimmune disorders among which, the human-like autoimmune diabetes. METHODOLOGY/PRINCIPAL FINDINGS: DNCD3 splenic cells from young NOD mice (1) provided long-lasting protection against diabetes transfer in NOD/Scid immunodeficient mice, (2) proliferated and differentiated in the spleen and pancreas of NOD/Scid mice and pre-diabetic NOD mice into IL-10-secreting T(R)-1 like cells in a Th2-like environment, and (3) their anti-diabetogenic phenotype is CD3(+)(CD4(-)CD8(-))CD28(+)CD69(+)CD25(low) Foxp3(-) iCTLA-4(-)TCR alphabeta(+) with a predominant Vbeta13 gene usage. CONCLUSIONS/SIGNIFICANCE: These findings delineate a new T regulatory component in autoimmune diabetes apart from that of NKT and CD4(+)CD25(high) Foxp3(+)T-regulatory cells. DNCD3 splenic cells could be potentially manipulated towards the development of autologous cell therapies in autoimmune diabetes.


Subject(s)
CD3 Complex/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Diabetes Mellitus, Type 1/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Cycle/physiology , Cell Proliferation , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology
9.
Eur J Immunol ; 40(8): 2277-88, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20540111

ABSTRACT

Polyclonality of self-reactive CD4(+) T cells is the hallmark of several autoimmune diseases like type 1 diabetes. We have previously reported that a soluble dimeric MHC II-peptide chimera prevents and reverses type 1 diabetes induced by a monoclonal diabetogenic T-cell population in double Tg mice [Casares, S. et al., Nat. Immunol. 2002. 3: 383-391]. Since most of the glutamic acid decarboxylase 65 (GAD65)-specific CD4(+) T cells in the NOD mouse are tolerogenic but unable to function in an autoimmune environment, we have activated a silent, monoclonal T-regulatory cell population (GAD65(217-230)-specific CD4(+) T cells) using a soluble I-A(αß) (g7)/GAD65(217-230)/Fcγ2a dimer, and measured the effect on the ongoing polyclonal diabetogenic T-cell process. Activated GAD65(217-230)-specific T cells and a fraction of the diabetogenic (B(9-23)-specific) T cells were polarized toward the IL-10-secreting T-regulatory type 1-like function in the pancreas of diabetic NOD mice. More importantly, this led to the reversal of hyperglycemia for more than 2 months post-therapy in 80% of mice in the context of stabilization of pancreatic insulitis and improved insulin secretion by the ß cells. These findings argue for the stabilization of a polyclonal self-reactive T-cell process by a single epitope-mediated bystander suppression. Dimeric MHC class II-peptide chimeras-like approach may provide rational grounds for the development of more efficient antigen-specific therapies in type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/immunology , Interleukin-10/biosynthesis , Pancreas/pathology , T-Lymphocytes, Regulatory/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Clone Cells , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/therapy , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Histocompatibility Antigens Class II/metabolism , Hyperglycemia , Immunosuppression Therapy , Insulin/genetics , Insulin/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
10.
Mutat Res ; 650(1): 15-29, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18006367

ABSTRACT

The particulate fraction of cigarette smoke, cigarette smoke condensate (CSC), is genotoxic in many short-term in vitro tests and is carcinogenic in rodents. However, no study has evaluated a series of CSCs prepared from a diverse set of cigarettes and produced with different smoking machine regimens in several short-term genotoxicity tests. Here we report on the genotoxicity of 10 CSCs prepared from commercial cigarettes that ranged from ultra-low tar per cigarette (< or =6.5 mg) to full flavor (>14.5 mg) as determined by the Federal Trade Commission (FTC) smoking regimen, a reference cigarette blended to be representative of a U.S. FTC-regimen low-tar cigarette, and experimental cigarettes constructed of single tobacco types. CSCs were tested in the presence of rat liver S9 in the Salmonella plate-incorporation assay using frameshift strains TA98 and YG1041; in micronucleus and comet assays in L5178Y/Tk(+/-) 7.3.2C mouse lymphoma cells, and in CHO-K(1) cells for chromosome aberrations. All 10 CSCs were mutagenic in both strains of Salmonella, and the rank order of their mutagenic potencies was similar. Their mutagenic potencies in Salmonella spanned 7-fold when expressed as rev/mug CSC but 158-fold when expressed as rev/mg nicotine; the range of genotoxic potencies of the CSCs in the other assays was similar regardless of how the data were expressed. All 10 CSCs induced micronuclei with a 3-fold range in their potency. All but one CSC induced DNA damage over a 20-fold range, and all but one CSC induced chromosome aberrations over a 4-fold range. There was no relation among the genotoxic potencies of the CSCs across the assays, and a qualitative advantage of the addition of the other assays to the Salmonella assay was not supported by our findings. Although consideration of nicotine levels may improve the relevance of the quantitative data obtained in the Salmonella and possibly comet assays, compensatory smoking habits and other factors may make the data from the assays used here have qualitative but not quantitative value in assessing risk of cigarette types and cigarette smoking to human health.


Subject(s)
Mutagens/toxicity , Nicotiana , Smoke/adverse effects , Animals , CHO Cells , Cricetinae , Cricetulus , Mice , Mutagenicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...