Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 4(11): 101245, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37913775

ABSTRACT

Adjuvanted protein vaccines offer high efficacy, yet most potent adjuvants remain proprietary. Several adjuvant compounds are being developed by the Vaccine Formulation Institute in Switzerland for global open access clinical use. In the context of the R21 malaria vaccine, in a mouse challenge model, we characterize the efficacy and mechanism of action of four Vaccine Formulation Institute adjuvants: two liposomal (LQ and LMQ) and two squalene emulsion-based adjuvants (SQ and SMQ), containing QS-21 saponin (Q) and optionally a synthetic TLR4 agonist (M). Two R21 vaccine formulations, R21/LMQ and R21/SQ, offer the highest protection (81%-100%), yet they trigger different innate sensing mechanisms in macrophages with LMQ, but not SQ, activating the NLRP3 inflammasome. The resulting in vivo adaptive responses have a different TH1/TH2 balance and engage divergent innate pathways while retaining high protective efficacy. We describe how modular changes in vaccine formulation allow for the dissection of the underlying immune pathways, enabling future mechanistically informed vaccine design.


Subject(s)
Malaria Vaccines , Malaria , Animals , Mice , Liposomes , Th1 Cells , Emulsions , Adjuvants, Immunologic/pharmacology , Malaria/prevention & control
2.
NPJ Vaccines ; 8(1): 163, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37884538

ABSTRACT

Adjuvants can enhance vaccine immunogenicity, but their mechanism of action is often incompletely understood, hampering rapid applicability for pandemic vaccines. Herein, we characterized the cellular and molecular activity of adjuvant formulations available for pre-clinical evaluation, including several developed for global open access. We applied four complementary human in vitro platforms to assess individual and combined adjuvants in unformulated, oil-in-water, and liposomal delivery platforms. Liposomal co-formulation of MPLA and QS-21 was most potent in promoting dendritic cell maturation, selective production of Th1-polarizing cytokines, and activation of SARS-CoV-2 Spike-specific CD4+ and CD8+ T cells in a co-culture assay. Select formulations also significantly enhanced Spike antigen-specific humoral immunity in vivo. This study confirms the utility of the cumulative use of human in vitro tools to predict adjuvanticity potential. Thus, human in vitro modeling may advance public health by accelerating the development of affordable and scalable adjuvants for vaccines tailored to vulnerable populations.

3.
Front Immunol ; 11: 580974, 2020.
Article in English | MEDLINE | ID: mdl-33262759

ABSTRACT

Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Membrane Glycoproteins/agonists , Monocytes/immunology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , B-Lymphocytes/immunology , Dendritic Cells/immunology , Drug Compounding , Germinal Center/immunology , Heterocyclic Compounds, 3-Ring/administration & dosage , Immunity, Innate , Interferon Type I/immunology , Ligands , Liposomes/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B/immunology , Phosphatidylcholines/administration & dosage , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Signal Transduction/immunology , Stearic Acids/administration & dosage , Th1 Cells/immunology
5.
NPJ Vaccines ; 5(1): 32, 2020.
Article in English | MEDLINE | ID: mdl-32337075

ABSTRACT

The rVSV-ZEBOV Ebolavirus vaccine confers protection within days after immunization, suggesting the contribution of innate immune responses. We report modulation of rVSV-ZEBOV vaccinee blood CD56+ NK cell numbers, NKG2D or NKp30 surface receptor expression, Killer Immunoglobulin-like Receptor (KIR)+ cell percentages and NK-cell-related genes on day 1 post immunization. Inverse correlations existed between the concentration of several plasma cytokines and inhibitory KIR+ CD56dim or cytokine-responsive CD56bright NK cells. Thus, NK cells may contribute to the early protective efficacy of rVSV-ZEBOV in humans.

7.
Front Immunol ; 10: 1650, 2019.
Article in English | MEDLINE | ID: mdl-31396211

ABSTRACT

Dendritic cells (DCs) that drain the gut and skin are known to favor the establishment of T cell populations that home to the original site of DC-antigen (Ag) encounter by providing soluble "imprinting" signals to T cells in the lymph node (LN). To study the induction of lung T cell-trafficking, we used a protein-adjuvant murine intranasal and intramuscular immunization model to compare in vivo-activated Ag+ DCs in the lung and muscle-draining LNs. Higher frequencies of Ag+ CD11b+ DCs were observed in lung-draining mediastinal LNs (MedLN) compared to muscle-draining inguinal LNs (ILN). Ag+ CD11b+ MedLN DCs were qualitatively superior at priming CD4+ T cells, which then expressed CD49a and CXCR3, and preferentially trafficked into the lung parenchyma. CD11b+ DCs from the MedLN expressed higher levels of surface podoplanin, Trem4, GL7, and the known co-stimulatory molecules CD80, CD86, and CD24. Blockade of specific MedLN DC molecules or the use of sorted DC and T cell co-cultures demonstrated that DC surface phenotype influences the ability to prime T cells that then home to the lung. Thus, the density of dLN Ag+ DCs, and DC surface molecule signatures are factors that can influence the output and differentiation of lung-homing CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Lung/immunology , Lymphocyte Activation/immunology , Animals , Cell Differentiation/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL
8.
Cell Rep ; 28(7): 1773-1784.e5, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412246

ABSTRACT

Maternal antibodies (MatAbs) protect offspring from infections but limit their responses to vaccination. The mechanisms of this inhibition are still debated. Using murine early-life immunization models mimicking the condition prevailing in humans, we observed the induction of CD4-T, T follicular helper, and germinal center (GC) B cell responses even when early-life antibody responses were abrogated by MatAbs. GC B cells induced in the presence of MatAbs form GC structures and exhibit canonical GC changes in gene expression but fail to differentiate into plasma cells and/or memory B cells in a MatAb titer-dependent manner. Furthermore, GC B cells elicited in the presence or absence of MatAbs use different VH and Vk genes and show differences in genes associated with B cell differentiation and isotype switching. Thus, MatAbs do not prevent B cell activation but control the output of the GC reaction both quantitatively and qualitatively, shaping the antigen-specific B cell repertoire.


Subject(s)
Antibodies, Viral/immunology , Antibodies/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Orthomyxoviridae/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Animals, Newborn , Antibodies/blood , Antibodies, Viral/blood , Female , Immunization , Male , Maternal-Fetal Exchange/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pregnancy , Vaccination
9.
Front Immunol ; 10: 1520, 2019.
Article in English | MEDLINE | ID: mdl-31333656

ABSTRACT

Pertussis is still observed in many countries despite of high vaccine coverage. Acellular pertussis (aP) vaccination is widely implemented in many countries as primary series in infants and as boosters in school-entry/adolescents/adults (including pregnant women in some). One novel strategy to improve the reactivation of aP-vaccine primed immunity could be to include genetically- detoxified pertussis toxin and novel adjuvants in aP vaccine boosters. Their preclinical evaluation is not straightforward, as it requires mimicking the human situation where T and B memory cells may persist longer than vaccine-induced circulating antibodies. Toward this objective, we developed a novel murine model including two consecutive adoptive transfers of the memory cells induced by priming and boosting, respectively. Using this model, we assessed the capacity of three novel aP vaccine candidates including genetically-detoxified pertussis toxin, pertactin, filamentous hemagglutinin, and fimbriae adsorbed to aluminum hydroxide, supplemented-or not-with Toll-Like-Receptor 4 or 9 agonists (TLR4A, TLR9A), to reactivate aP vaccine-induced immune memory and protection, reflected by bacterial clearance. In the conventional murine immunization model, TLR4A- and TLR9A-containing aP formulations induced similar aP-specific IgG antibody responses and protection against bacterial lung colonization as current aP vaccines, despite IL-5 down-modulation by both TLR4A and TLR9A and IL-17 up-modulation by TLR4A. In the absence of serum antibodies at time of boosting or exposure, TLR4A- and TLR9A-containing formulations both enhanced vaccine antibody recall compared to current aP formulations. Unexpectedly, however, protection was only increased by the TLR9A-containing vaccine, through both earlier bacterial control and accelerated clearance. This suggests that TLR9A-containing aP vaccines may better reactivate aP vaccine-primed pertussis memory and enhance protection than current or TLR4A-adjuvanted aP vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bordetella pertussis/immunology , Pertussis Vaccine , Toll-Like Receptor 4 , Toll-Like Receptor 9 , Animals , Antibodies, Bacterial/immunology , Female , Mice , Mice, Inbred BALB C , Pertussis Vaccine/genetics , Pertussis Vaccine/immunology , Pertussis Vaccine/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/immunology , Whooping Cough/immunology , Whooping Cough/prevention & control
10.
Front Immunol ; 9: 381, 2018.
Article in English | MEDLINE | ID: mdl-29541075

ABSTRACT

Neonates and infants are more vulnerable to infections and show reduced responses to vaccination. Consequently, repeated immunizations are required to induce protection and early life vaccines against major pathogens such as influenza are yet unavailable. Formulating antigens with potent adjuvants, including immunostimulators and delivery systems, is a demonstrated approach to enhance vaccine efficacy. Yet, adjuvants effective in adults may not meet the specific requirements for activating the early life immune system. Here, we assessed the neonatal adjuvanticity of three novel adjuvants including TLR4 (glucopyranosyl lipid adjuvant-squalene emulsion), TLR9 (IC31®), and Mincle (CAF01) agonists, which all induce germinal centers (GCs) and potent antibody responses to influenza hemagglutinin (HA) in adult mice. In neonates, a single dose of HA formulated into each adjuvant induced T follicular helper (TFH) cells. However, only HA/CAF01 elicited significantly higher and sustained antibody responses, engaging neonatal B cells to differentiate into GCs already after a single dose. Although antibody titers remained lower than in adults, HA-specific responses induced by a single neonatal dose of HA/CAF01 were sufficient to confer protection against influenza viral challenge. Postulating that the neonatal adjuvanticity of CAF01 may result from the functionality of the C-type lectin receptor (CLR) Mincle in early life we asked whether other C-type lectin agonists would show a similar neonatal adjuvanticity. Replacing the Mincle agonist trehalose 6,6'-dibehenate by Curdlan, which binds to Dectin-1, enhanced antibody responses through the induction of similar levels of TFH, GCs and bone marrow high-affinity plasma cells. Thus, specific requirements of early life B cells may already be met after a single vaccine dose using CLR-activating agonists, identified here as promising B cell immunostimulators for early life vaccines when included into cationic liposomes.


Subject(s)
Adjuvants, Immunologic , B-Lymphocytes/immunology , Germinal Center/immunology , Glycolipids/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , beta-Glucans/immunology , Adjuvants, Immunologic/pharmacology , Animals , Animals, Newborn , Antibodies, Viral/blood , Female , Glycolipids/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Lectins, C-Type/agonists , Lectins, C-Type/metabolism , Liposomes , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists , Toll-Like Receptor 9/metabolism , beta-Glucans/pharmacology
11.
Sci Rep ; 6: 39117, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27974744

ABSTRACT

Th17 cells play critical roles in host defense and autoimmunity. Emerging data support a role for Notch signaling in Th17 cell differentiation but whether it is a positive or negative regulator remains unclear. We report here that T cell-specific deletion of Notch receptors enhances Th17 cell differentiation in the gut, with a corresponding increase in IL-17 secretion. An increase in Th17 cell frequency was similarly observed following immunization of T cell specific Notch mutant mice with OVA/CFA. However, in this setting, Th17 cytokine secretion was impaired, and increased intracellular retention of IL-17 was observed. Intracellular IL-17 co-localized with the CD71 iron transporter in the draining lymph node of both control and Notch-deficient Th17 cells. Immunization induced CD71 surface expression in control, but not in Notch-deficient Th17 cells, revealing defective CD71 intracellular transport in absence of Notch signaling. Moreover, Notch receptor deficient Th17 cells had impaired mTORC2 activity. These data reveal a context-dependent impact of Notch on vesicular transport during high metabolic demand suggesting a role for Notch signaling in the bridging of T cell metabolic demands and effector functions. Collectively, our findings indicate a prominent regulatory role for Notch signaling in the fine-tuning of Th17 cell differentiation and effector function.


Subject(s)
Immunization/methods , Interleukin-17/metabolism , Receptors, Notch/metabolism , Th17 Cells/cytology , Animals , Antigens, CD/metabolism , Cell Differentiation , Cells, Cultured , Mice , Protein Transport , Receptors, Transferrin/metabolism , Signal Transduction
12.
N Engl J Med ; 374(17): 1647-60, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-25830326

ABSTRACT

BACKGROUND: The replication-competent recombinant vesicular stomatitis virus (rVSV)-based vaccine expressing a Zaire ebolavirus (ZEBOV) glycoprotein was selected for rapid safety and immunogenicity testing before its use in West Africa. METHODS: We performed three open-label, dose-escalation phase 1 trials and one randomized, double-blind, controlled phase 1 trial to assess the safety, side-effect profile, and immunogenicity of rVSV-ZEBOV at various doses in 158 healthy adults in Europe and Africa. All participants were injected with doses of vaccine ranging from 300,000 to 50 million plaque-forming units (PFU) or placebo. RESULTS: No serious vaccine-related adverse events were reported. Mild-to-moderate early-onset reactogenicity was frequent but transient (median, 1 day). Fever was observed in up to 30% of vaccinees. Vaccine viremia was detected within 3 days in 123 of the 130 participants (95%) receiving 3 million PFU or more; rVSV was not detected in saliva or urine. In the second week after injection, arthritis affecting one to four joints developed in 11 of 51 participants (22%) in Geneva, with pain lasting a median of 8 days (interquartile range, 4 to 87); 2 self-limited cases occurred in 60 participants (3%) in Hamburg, Germany, and Kilifi, Kenya. The virus was identified in one synovial-fluid aspirate and in skin vesicles of 2 other vaccinees, showing peripheral viral replication in the second week after immunization. ZEBOV-glycoprotein-specific antibody responses were detected in all the participants, with similar glycoprotein-binding antibody titers but significantly higher neutralizing antibody titers at higher doses. Glycoprotein-binding antibody titers were sustained through 180 days in all participants. CONCLUSIONS: In these studies, rVSV-ZEBOV was reactogenic but immunogenic after a single dose and warrants further evaluation for safety and efficacy. (Funded by the Wellcome Trust and others; ClinicalTrials.gov numbers, NCT02283099, NCT02287480, and NCT02296983; Pan African Clinical Trials Registry number, PACTR201411000919191.).


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Membrane Glycoproteins/immunology , Viral Envelope Proteins/immunology , Adult , Antibodies, Viral/blood , Arthritis/etiology , Dermatitis/etiology , Double-Blind Method , Ebola Vaccines/administration & dosage , Ebola Vaccines/adverse effects , Ebolavirus/isolation & purification , Exanthema/etiology , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Male , Middle Aged , Recombinant Proteins , Vesiculovirus , Viremia , Virus Shedding
13.
Lancet Infect Dis ; 15(10): 1156-1166, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26248510

ABSTRACT

BACKGROUND: Safe and effective vaccines against Ebola could prevent or control outbreaks. The safe use of replication-competent vaccines requires a careful dose-selection process. We report the first safety and immunogenicity results in volunteers receiving 3 × 10(5) plaque-forming units (pfu) of the recombinant vesicular stomatitis virus-based candidate vaccine expressing the Zaire Ebola virus glycoprotein (rVSV-ZEBOV; low-dose vaccinees) compared with 59 volunteers who had received 1 ×10(7) pfu (n=35) or 5 × 10(7) pfu (n=16) of rVSV-ZEBOV (high-dose vaccinees) or placebo (n=8) before a safety-driven study hold. METHODS: The Geneva rVSV-ZEBOV study, an investigator-initiated phase 1/2, dose-finding, placebo-controlled, double-blind trial conducted at the University Hospitals of Geneva, Switzerland, enrolled non-pregnant, immunocompetent, and otherwise healthy adults aged 18-65 years. Participants from the low-dose group with no plans to deploy to Ebola-aff5cted regions (non-deployable) were randomised 9:1 in a double-blind fashion using randomly permuted blocks of varying sizes to a single injection of 3 × 10(5) pfu or placebo, whereas deployable participants received single-injection 3 × 10(5) pfu open-label. Primary safety and immunogenicity outcomes were the incidence of adverse events within 14 days of vaccination and day-28 antibody titres, respectively, analysed by intention to treat. After viral oligoarthritis was observed in 11 of the first 51 vaccinees (22%) receiving 10(7) or 5 × 10(7) pfu, 56 participants were given a lower dose (3 × 10(5) pfu, n=51) or placebo (n=5) to assess the effect of dose reduction on safety and immunogenicity. This trial is ongoing with a follow-up period of 12 months; all reported results are from interim databases. This study is registered with ClinicalTrials.gov, number NCT02287480. FINDINGS: Between Jan 5 and Jan 26, 2015, 43 non-deployable participants received low-dose rVSV-ZEBOV (3 × 10(5) pfu) or placebo in a double-blind fashion, whereas 13 deployable participants received 3 × 10(5) pfu open-label. Altogether, in the low-dose group, 51 participants received rVSV-ZEBOV and five received placebo. No serious adverse events occurred. At 3 × 10(5) pfu, early-onset reactogenicity remained frequent (45 [88%] of 51 compared with 50 [98%] of 51 high dose and two [15%] of 13 placebo recipients), but mild. Objective fever was present in one (2%) of 51 low-dose versus 13 (25%) of 51 high-dose vaccinees receiving at least 1 ×10(7) pfu (p<0·0001). Subjective fever (p<0·0001), myalgia (p=0·036), and chills (p=0·026) were significantly reduced and their time of onset delayed, reflecting significantly lower viraemia (p<0·0001) and blood monocyte-activation patterns (p=0·0233). Although seropositivity rates remained similarly high (48 [94%] of 51), day-28 EBOV-glycoprotein-binding and neutralising antibody titres were lower in low-dose versus high-dose vaccinees (geometric mean titres 344·5 [95% CI 229·7-516·4] vs 1064·2 [757·6-1495·1]; p<0·0001; and 35·1 [24·7-50·7] vs 127·0 [86·0-187·6]; p<0·0001, respectively). Furthermore, oligoarthritis again occurred on day 10 (median; IQR 9-14) in 13 (25%) of 51 low-dose vaccinees, with maculopapular, vesicular dermatitis, or both in seven (54%) of 13; arthritis was associated with increasing age in low-dose but not high-dose vaccinees. Two vaccinees presented with purpura of the lower legs; histological findings indicated cutaneous vasculitis. The presence of rVSV in synovial fluid and skin lesions confirmed causality. INTERPRETATION: Reducing the dose of rVSV-ZEBOV improved its early tolerability but lowered antibody responses and did not prevent vaccine-induced arthritis, dermatitis, or vasculitis. Like its efficacy, the safety of rVSV-ZEBOV requires further definition in the target populations of Africa. FUNDING: Wellcome Trust through WHO.


Subject(s)
Ebola Vaccines/administration & dosage , Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Vaccination/adverse effects , Vaccination/methods , Adolescent , Adult , Aged , Antibodies, Viral/blood , Antigens, Viral/genetics , Arthritis/chemically induced , Arthritis/epidemiology , Arthritis/pathology , Dermatitis/epidemiology , Dermatitis/pathology , Double-Blind Method , Drug Carriers , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Ebola Vaccines/adverse effects , Ebolavirus/genetics , Female , Humans , Male , Middle Aged , Placebos/administration & dosage , Switzerland , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology , Vasculitis/chemically induced , Vasculitis/epidemiology , Vasculitis/pathology , Vesiculovirus/genetics , Young Adult
14.
Vaccine ; 33(30): 3450-5, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26067183

ABSTRACT

The limited durability of pertussis vaccine-induced protection requires novel approaches to reactivate immunity and limit pertussis resurgence in older children and adults. We propose that periodic boosters could be delivered using a novel epicutaneous delivery system (Viaskin) to deliver optimized pertussis antigens such as genetically-detoxified pertussis toxin (rPT). To best mimic the human situation in which vaccine-induced memory cells persist, whereas antibodies wane, we developed a novel adoptive transfer murine model of pertussis immunity. This allowed demonstrating that a single application of Viaskin delivering rPT and/or pertactin and filamentous hemagglutinin effectively reactivates vaccine-induced pertussis immunity and protects against Bordetella pertussis challenge. Recalling pertussis immunity without needles nor adjuvant may considerably facilitate the acceptance and application of periodic boosters.


Subject(s)
Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bordetella pertussis/immunology , Immunization, Secondary/methods , Pertussis Vaccine/administration & dosage , Pertussis Vaccine/immunology , Whooping Cough/prevention & control , Administration, Cutaneous , Animals , Female , Mice, Inbred BALB C
15.
J Immunol ; 194(10): 4836-45, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25870238

ABSTRACT

The early life influenza disease burden calls for more effective vaccines to protect this vulnerable population. Influenza vaccines including the MF59 oil-in-water adjuvant induce higher, broader, and more persistent Ab responses in adults and particularly in young, through yet undefined mechanisms. In this study, we show that MF59 enhances adult murine IgG responses to influenza hemagglutinin (HA) by promoting a potent T follicular helper cells (TFH) response, which directly controls the magnitude of the germinal center (GC) B cell response. Remarkably, this enhancement of TFH and GC B cells is already fully functional in 3-wk-old infant mice, which were fully protected by HA/MF59 but not HA/PBS immunization against intranasal challenge with the homologous H1N1 (A/California/7/2009) strain. In 1-wk-old neonatal mice, MF59 recruits and activates APCs, efficiently induces CD4(+) effector T cells and primes for enhanced infant responses but induces few fully functional TFH cells, which are mostly follicular regulatory T cells, and poor GC and anti-HA responses. The B cell adjuvanticity of MF59 appears to be mediated by the potent induction of TFH cells which directly controls GC responses both in adult and early life, calling for studies assessing its capacity to enhance the efficacy of influenza immunization in young infants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Polysorbates/pharmacology , Squalene/pharmacology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Animals, Newborn , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/immunology , Immunohistochemistry , Influenza A Virus, H1N1 Subtype/immunology , Mice , Real-Time Polymerase Chain Reaction , Squalene/immunology
16.
J Exp Med ; 211(11): 2265-79, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25311507

ABSTRACT

Fibroblast-like cells of secondary lymphoid organs (SLO) are important for tissue architecture. In addition, they regulate lymphocyte compartmentalization through the secretion of chemokines, and participate in the orchestration of appropriate cell-cell interactions required for adaptive immunity. Here, we provide data demonstrating the functional importance of SLO fibroblasts during Notch-mediated lineage specification and immune response. Genetic ablation of the Notch ligand Delta-like (DL)1 identified splenic fibroblasts rather than hematopoietic or endothelial cells as niche cells, allowing Notch 2-driven differentiation of marginal zone B cells and of Esam(+) dendritic cells. Moreover, conditional inactivation of DL4 in lymph node fibroblasts resulted in impaired follicular helper T cell differentiation and, consequently, in reduced numbers of germinal center B cells and absence of high-affinity antibodies. Our data demonstrate previously unknown roles for DL ligand-expressing fibroblasts in SLO niches as drivers of multiple Notch-mediated immune differentiation processes.


Subject(s)
Cellular Microenvironment/immunology , Fibroblasts/metabolism , Immunity , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Receptors, Notch/metabolism , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers/metabolism , Calcium-Binding Proteins , Cell Adhesion Molecules/metabolism , Cell Differentiation , Chemokine CCL19/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Immunophenotyping , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Phenotype , Spleen/immunology , Spleen/metabolism , Stromal Cells/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
17.
J Immunol ; 191(5): 2344-50, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918982

ABSTRACT

Follicular helper T (TFH) cells are specialized in providing help for B cell differentiation and Ab secretion. Several positive and negative regulators of TFH cell differentiation have been described but their control is not fully understood. In this study, we show that Notch signaling in T cells is a major player in the development and function of TFH cells. T cell-specific gene ablation of Notch1 and Notch2 impaired differentiation of TFH cells in draining lymph nodes of mice immunized with T-dependent Ags or infected with parasites. Impaired TFH cell differentiation correlated with deficient germinal center development and the absence of high-affinity Abs. The impact of loss of Notch on TFH cell differentiation was largely independent of its effect on IL-4. These results show a previously unknown role for Notch in the regulation of TFH cell differentiation and function with implications for the control of this T cell population.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Flow Cytometry , Gene Knockdown Techniques , Germinal Center/cytology , Germinal Center/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Helper-Inducer/cytology
18.
Infect Immun ; 81(5): 1575-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23439309

ABSTRACT

Infection with Leishmania braziliensis causes cutaneous or mucocutaneous leishmaniasis in humans. Toll-like receptor 9 (TLR9) expression has been found in granulomas of lesions in L. braziliensis-infected individuals. L. braziliensis inoculation in mice induces very small lesions that are self-healing, whereas deficiency in the TLR adaptor molecule, MyD88, renders mice susceptible to infection. The TLR involved has not been identified, prompting us to investigate if TLR9 triggering by the parasite contributes to the strong resistance to infection observed in L. braziliensis-inoculated mice. The parasites activated wild-type (WT) dendritic cells (DCs) in vitro but not DCs derived from TLR9(-/-) mice. TLR9(-/-) mice inoculated with L. braziliensis exhibited a transient susceptibility characterized by increased lesion size and parasite burden compared to those of WT mice. Surprisingly, elevated levels of gamma interferon (IFN-γ) were measured at the site of infection and in draining lymph node T cells of TLR9(-/-) mice at the peak of susceptibility, suggesting that unlike observations in vitro, the parasite could induce DC activation leading to the development of Th1 cells in the absence of TLR9 expression. Taken together, these data show that TLR9 signaling is important for the early control of lesion development and parasite burden but is dispensable for the differentiation of Th1 cells secreting IFN-γ, and the high levels of this cytokine are not sufficient to control early parasite replication following L. braziliensis infection.


Subject(s)
Leishmania braziliensis , Leishmaniasis, Cutaneous/metabolism , Toll-Like Receptor 9/physiology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Leishmaniasis, Cutaneous/immunology , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Toll-Like Receptor 9/deficiency
19.
J Immunol ; 189(12): 5764-72, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23162125

ABSTRACT

Follicular Th (T(FH)) cells have emerged as a new Th subset providing help to B cells and supporting their differentiation into long-lived plasma cells or memory B cells. Their differentiation had not yet been investigated following neonatal immunization, which elicits delayed and limited germinal center (GC) responses. We demonstrate that neonatal immunization induces CXCR5(high)PD-1(high) CD4(+) T(FH) cells that exhibit T(FH) features (including Batf, Bcl6, c-Maf, ICOS, and IL-21 expression) and are able to migrate into the GCs. However, neonatal T(FH) cells fail to expand and to acquire a full-blown GC T(FH) phenotype, as reflected by a higher ratio of GC T(FH)/non-GC CD4(+) T cells in immunized adults than neonates (3.8 × 10(-3) versus 2.2 × 10(-3), p = 0.01). Following the adoptive transfer of naive adult OT-II CD4(+) T cells, OT-II T(FH) cells expand in the vaccine-draining lymph nodes of immunized adult but not infant recipients, whereas naive 2-wk-old CD4(+) OT-II cells failed to expand in adult hosts, reflecting the influence of both environmental and T cell-intrinsic factors. Postponing immunization to later in life increases the number of T(FH) cells in a stepwise manner, in direct correlation with the numbers of GC B cells and plasma cells elicited. Remarkably, adjuvantation with CpG oligonucleotides markedly increased GC T(FH) and GC B cell neonatal responses, up to adult levels. To our knowledge, this is the first demonstration that the T(FH) cell development limits early life GC responses and that adjuvants/delivery systems supporting T(FH) differentiation may restore adultlike early life GC B cell responses.


Subject(s)
Adjuvants, Immunologic/physiology , Aging/immunology , Cell Communication/immunology , Cell Differentiation/immunology , Cellular Microenvironment/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , Animals, Newborn , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Communication/genetics , Cell Differentiation/genetics , Cellular Senescence/immunology , CpG Islands/immunology , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Immunization , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/transplantation , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology
20.
Curr Top Microbiol Immunol ; 360: 115-34, 2012.
Article in English | MEDLINE | ID: mdl-22653552

ABSTRACT

CD4⁺ T helper cells are playing critical roles in host defense to pathogens and in the maintenance of immune homeostasis. Naïve CD4⁺T cells, upon antigen-specific recognition, receive signals to differentiate into distinct effector T helper cell subsets characterized by their pattern of cytokine production and specific immune functions. A tight balance between these different subsets ensures proper control of the immune response. There is increasing evidence revealing an important role for Notch signaling in the regulation of CD4⁺T helper cell differentiation or function in the periphery. However, the exact mechanisms involved remain unclear and appear contradictory. In this review, we summarize current knowledge and discuss recent advances in the field to reconcile different views on the role of Notch signaling in the differentiation of functional T helper subsets.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Differentiation/immunology , Cytokines/immunology , Cytokines/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Interleukin-4/genetics , Interleukin-4/metabolism , Mice , Promoter Regions, Genetic , Receptors, Notch/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...