Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978810

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a global health threat, compounded by the emergence of drug-resistant strains. A hallmark of pulmonary tuberculosis (TB) is the formation of hypoxic necrotic granulomas, which upon disintegration, release infectious Mtb. Furthermore, hypoxic necrotic granulomas are associated with increased disease severity and provide a niche for drug-resistant Mtb. However, the host immune responses that promote the development of hypoxic TB granulomas are not well described. Using a necrotic Mtb mouse model, we show that loss of Mtb virulence factors, such as phenolic glycolipids, decreases the production of the proinflammatory cytokine IL-17 (also referred to as IL-17A). IL-17 production negatively regulates the development of hypoxic TB granulomas by limiting the expression of the transcription factor hypoxia-inducible factor 1α (HIF1α). In human TB patients, HIF1α mRNA expression is increased. Through genotyping and association analyses in human samples, we identified a link between the single nucleotide polymorphism rs2275913 in the IL-17 promoter (-197G/G), which is associated with decreased IL-17 production upon stimulation with Mtb cell wall. Together, our data highlight a potentially novel role for IL-17 in limiting the development of hypoxic necrotic granulomas and reducing disease severity in TB.


Subject(s)
Granuloma/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Interleukin-17/immunology , Tuberculosis, Pulmonary/immunology , Adult , Aged , Animals , Cell Hypoxia/immunology , Female , Gene Expression Regulation/immunology , Granuloma/microbiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Male , Mice, Inbred Strains , Middle Aged , RNA, Messenger/genetics , Tuberculosis, Pulmonary/complications , Young Adult
2.
J Leukoc Biol ; 101(6): 1373-1381, 2017 06.
Article in English | MEDLINE | ID: mdl-28258153

ABSTRACT

Pulmonary tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is a leading cause of global morbidity and mortality. The only licensed TB vaccine, Mycobacterium bovis bacillus Calmette-Guerin (BCG), has variable efficacy in protecting against pulmonary TB. Thus, the development of more effective TB vaccines is critical to control the TB epidemic. Specifically, vaccines delivered through the mucosal route are known to induce Th17 responses and provide superior protection against Mtb infection. However, already tested Th17-inducing mucosal adjuvants, such as heat-labile enterotoxins and cholera toxins, are not considered safe for use in humans. In the current study, we rationally screened adjuvants for their ability to induce Th17-polarizing cytokines in dendritic cells (DCs) and determined whether they could be used in a protective mucosal TB vaccine. Our new studies show that monophosphoryl lipid A (MPL), when used in combination with chitosan, potently induces Th17-polarizing cytokines in DCs and downstream Th17/Th1 mucosal responses and confers significant protection in mice challenged with a clinical Mtb strain. Additionally, we show that both TLRs and the inflammasome pathways are activated in DCs by MPL-chitosan to mediate induction of Th17-polarizing cytokines. Together, our studies put forward the potential of a new, protective mucosal TB vaccine candidate, which incorporates safe adjuvants already approved for use in humans.


Subject(s)
BCG Vaccine/therapeutic use , Mucous Membrane/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Administration, Intranasal , Animals , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/drug effects , Mucous Membrane/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
3.
Nat Commun ; 7: 13894, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28004802

ABSTRACT

The development of a tuberculosis (TB) vaccine that induces sterilizing immunity to Mycobacterium tuberculosis infection has been elusive. Absence of sterilizing immunity induced by TB vaccines may be due to delayed activation of mucosal dendritic cells (DCs), and subsequent delay in antigen presentation and activation of vaccine-induced CD4+ T-cell responses. Here we show that pulmonary delivery of activated M. tuberculosis antigen-primed DCs into vaccinated mice, at the time of M. tuberculosis exposure, can overcome the delay in accumulation of vaccine-induced CD4+ T-cell responses. In addition, activating endogenous host CD103+ DCs and the CD40-CD40L pathway can similarly induce rapid accumulation of vaccine-induced lung CD4+ T-cell responses and limit early M. tuberculosis growth. Thus, our study provides proof of concept that targeting mucosal DCs can accelerate vaccine-induced T-cell responses on M. tuberculosis infection, and provide insights to overcome bottlenecks in TB vaccine efficacy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Tuberculosis Vaccines/immunology , Adoptive Transfer , Animals , Antigen Presentation , Antigens, CD/metabolism , Female , Humans , Integrin alpha Chains/metabolism , Lymphocyte Activation , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/prevention & control
4.
Hum Vaccin Immunother ; 12(10): 2649-2653, 2016 10 02.
Article in English | MEDLINE | ID: mdl-27322875

ABSTRACT

Mycobacterium tuberculosis infects one third of the world's population. Due to variable efficacy of the Bacille Calmette Guerin (BCG) vaccine, development of novel TB vaccines remains a priority. Here, we demonstrate the protective efficacy of a novel multivalent DNA vaccine, which contains 15 synthetic antigens targeting the Mtb ESX secretion system.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/isolation & purification , Tuberculosis/prevention & control , Animals , Disease Models, Animal , Humans , Mice, Inbred C57BL , Tuberculosis/immunology
5.
J Clin Invest ; 125(12): 4699-713, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26571397

ABSTRACT

Parasitic helminth worms, such as Schistosoma mansoni, are endemic in regions with a high prevalence of tuberculosis (TB) among the population. Human studies suggest that helminth coinfections contribute to increased TB susceptibility and increased rates of TB reactivation. Prevailing models suggest that T helper type 2 (Th2) responses induced by helminth infection impair Th1 immune responses and thereby limit Mycobacterium tuberculosis (Mtb) control. Using a pulmonary mouse model of Mtb infection, we demonstrated that S. mansoni coinfection or immunization with S. mansoni egg antigens can reversibly impair Mtb-specific T cell responses without affecting macrophage-mediated Mtb control. Instead, S. mansoni infection resulted in accumulation of high arginase-1-expressing macrophages in the lung, which formed type 2 granulomas and exacerbated inflammation in Mtb-infected mice. Treatment of coinfected animals with an antihelminthic improved Mtb-specific Th1 responses and reduced disease severity. In a genetically diverse mouse population infected with Mtb, enhanced arginase-1 activity was associated with increased lung inflammation. Moreover, in patients with pulmonary TB, lung damage correlated with increased serum activity of arginase-1, which was elevated in TB patients coinfected with helminths. Together, our data indicate that helminth coinfection induces arginase-1-expressing type 2 granulomas, thereby increasing inflammation and TB disease severity. These results also provide insight into the mechanisms by which helminth coinfections drive increased susceptibility, disease progression, and severity in TB.


Subject(s)
Arginase/blood , Lung/metabolism , Macrophages/enzymology , Mycobacterium tuberculosis , Schistosoma mansoni , Schistosomiasis mansoni/blood , Tuberculosis, Pulmonary/blood , Animals , Female , Granuloma/enzymology , Granuloma/microbiology , Granuloma/parasitology , Granuloma/pathology , Humans , Lung/microbiology , Lung/parasitology , Lung/pathology , Macrophages/pathology , Male , Mice , Mice, Transgenic , Schistosomiasis mansoni/microbiology , Schistosomiasis mansoni/pathology , Tuberculosis, Pulmonary/parasitology , Tuberculosis, Pulmonary/pathology
6.
J Cell Mol Med ; 19(8): 2019-31, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26130503

ABSTRACT

Escherichia coli's heat-labile enterotoxin (Etx) and its non-toxic B subunit (EtxB) have been characterized as adjuvants capable of enhancing T cell responses to co-administered antigen. Here, we investigate the direct effect of intravenously administered EtxB on the size of the dendritic and myeloid cell populations in spleen. EtxB treatment appears to enhance the development and turnover of dendritic and myeloid cells from precursors within the spleen. EtxB treatment also gives a dendritic cell (DC) population with higher viability and lower activation status based on the reduced expression of MHC-II, CD80 and CD86. In this respect, the in vivo effect of EtxB differs from that of the highly inflammatory mediator lipopolysaccharide. In in vitro bone marrow cultures, EtxB treatment was also found to enhance the development of DC from precursors dependent on Flt3L. In terms of the in vivo effect of EtxB on CD4 and CD8 T cell responses in mice, the interaction of EtxB directly with DC was demonstrated following conditional depletion of CD11c(+) DC. In summary, all results are consistent with EtxB displaying adjuvant ability by enhancing the turnover of DC in spleen, leading to newly mature myeloid and DC in spleen, thereby increasing DC capacity to perform as antigen-presenting cells on encounter with T cells.


Subject(s)
Antigen Presentation/immunology , Bacterial Toxins/pharmacology , Dendritic Cells/immunology , Enterotoxins/pharmacology , Escherichia coli Proteins/pharmacology , Protein Subunits/pharmacology , Animals , Antigen Presentation/drug effects , Bone Marrow Cells/cytology , Cell Count , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Female , Lymphocyte Activation/drug effects , Membrane Proteins/metabolism , Mice, Inbred C57BL , Myeloid Cells/cytology , Spleen/cytology
8.
Oncotarget ; 5(22): 11064-80, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25415223

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant desmoplastic reaction driven by pancreatic stellate cells (PSCs) that contributes to tumor progression. Here we sought to characterize the interactions between pancreatic cancer cells (PCCs) and PSCs that affect the inflammatory and immune response in pancreatic tumors. Conditioned media from mono- and cocultures of PSCs and PCCs were assayed for expression of cytokines and growth factors. IP-10/CXCL10 was the most highly induced chemokine in coculture of PSCs and PCCs. Its expression was induced in the PSCs by PCCs. IP-10 was elevated in human PDAC specimens, and positively correlated with high stroma content. Furthermore, gene expression of IP-10 and its receptor CXCR3 were significantly associated with the intratumoral presence of regulatory T cells (Tregs). In an independent cohort of 48 patients with resectable pancreatic ductal adenocarcinoma, high IP-10 expression levels correlated with decreased median overall survival. Finally, IP-10 stimulated the ex vivo recruitment of CXCR3+ effector T cells as well as CXCR3+ Tregs derived from patients with PDAC. Our findings suggest that, in pancreatic cancer, CXCR3+ Tregs can be recruited by IP-10 expressed by PSCs in the tumor stroma, leading to immunosuppressive and tumor-promoting effects.


Subject(s)
Carcinoma, Pancreatic Ductal/immunology , Chemokine CXCL10/biosynthesis , Pancreatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , HEK293 Cells , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , T-Lymphocytes, Regulatory/pathology
9.
Lancet Infect Dis ; 14(10): 939-46, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25151225

ABSTRACT

BACKGROUND: Intradermal MVA85A, a candidate vaccine against tuberculosis, induces high amounts of Ag85A-specific CD4 T cells in adults who have already received the BCG vaccine, but aerosol delivery of this vaccine might offer immunological and logistical advantages. We did a phase 1 double-blind trial to compare the safety and immunogenicity of aerosol-administered and intradermally administered MVA85A METHODS: In this phase 1, double-blind, proof-of-concept trial, 24 eligible BCG-vaccinated healthy UK adults were randomly allocated (1:1) by sequentially numbered, sealed, opaque envelopes into two groups: aerosol MVA85A and intradermal saline placebo or intradermal MVA85A and aerosol saline placebo. Participants, the bronchoscopist, and immunologists were masked to treatment assignment. The primary outcome was safety, assessed by the frequency and severity of vaccine-related local and systemic adverse events. The secondary outcome was immunogenicity assessed with laboratory markers of cell-mediated immunity in blood and bronchoalveolar lavage samples. Safety and immunogenicity were assessed for 24 weeks after vaccination. Immunogenicity to both insert Ag85A and vector modified vaccinia virus Ankara (MVA) was assessed by ex-vivo interferon-γ ELISpot and serum ELISAs. Since all participants were randomised and vaccinated according to protocol, our analyses were per protocol. This trial is registered with ClinicalTrials.gov, number NCT01497769. FINDINGS: Both administration routes were well tolerated and immunogenic. Respiratory adverse events were rare and mild. Intradermal MVA85A was associated with expected mild local injection-site reactions. Systemic adverse events did not differ significantly between the two groups. Three participants in each group had no vaccine-related systemic adverse events; fatigue (11/24 [46%]) and headache (10/24 [42%]) were the most frequently reported symptoms. Ag85A-specific systemic responses were similar across groups. Ag85A-specific CD4 T cells were detected in bronchoalveolar lavage cells from both groups and responses were higher in the aerosol group than in the intradermal group. MVA-specific cellular responses were detected in both groups, whereas serum antibodies to MVA were only detectable after intradermal administration of the vaccine. INTERPRETATION: Further clinical trials assessing the aerosol route of vaccine delivery are merited for tuberculosis and other respiratory pathogens. FUNDING: The Wellcome Trust and Oxford Radcliffe Hospitals Biomedical Research Centre.


Subject(s)
Antigens, Bacterial/immunology , BCG Vaccine/immunology , Mycobacterium bovis/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis/immunology , Vaccination/adverse effects , Administration, Inhalation , Adult , Aerosols , CD4-Positive T-Lymphocytes/immunology , Double-Blind Method , Female , Humans , Immunity, Cellular , Injections, Intradermal , Male , Middle Aged , Safety , Tuberculosis/prevention & control , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/standards , Vaccines, DNA , Young Adult
10.
J Cell Mol Med ; 18(9): 1908-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913604

ABSTRACT

The Gram-negative bacterial endotoxin lipopolysaccharide (LPS) is a potent inflammatory mediator and a leading cause of bacterial sepsis. While LPS is known to activate antigen-presenting cells, here we find that LPS down-regulates expression of CD11c and CD11b on splenic dendritic cell subsets, thus confounding the ability to identify these subsets following treatment. This has implications with regard to tracking the response to LPS in terms of the cell subsets involved, and should be considered whenever such studies are undertaken.


Subject(s)
Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Spleen/immunology , Animals , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cells, Cultured , Down-Regulation , Endocytosis , Female , Mice, Inbred C57BL
11.
Curr Stem Cell Res Ther ; 9(4): 354-63, 2014.
Article in English | MEDLINE | ID: mdl-24745998

ABSTRACT

Hematopoietic stem/progenitor cells (HSPC) differentiate in the context of stromal niches producing cells of multiple lineages. Limited success has been achieved in the past with induction of hematopoiesis in vitro. Previously, spleen long-term stromal cultures (LTC) were shown to continuously support restricted hematopoiesis for production of novel dendritic-like cells (LTC-DC). An in vivo equivalent dendritic cell type was then described which is specific for spleen. The in vivo counterpart cell was termed 'L-DC' and represents a dendritic-like CD11c(lo)CD11b(hi)CD8α-MHC-II- cell which differs phenotypically and functionally from monocytes/macrophages and conventional and plasmacytoid DC. Splenic stroma is now shown to maintain HSPC and to support their restricted in vitro differentiation to give this 'L-DC' subset. In order to characterise progenitors of this distinct cell type, LTC were analysed for cell subsets produced, and these subsets sorted and assessed for hematopoietic potential in subsequent co-cultures over STX3 stroma. Progenitors were defined as a lineage (Lin)(-)ckit(lo) subset reflecting HSPC. Furthermore, when Lin(-)ckit(hi)Sca1(+)Flt3(-) HSPC were sorted from bone marrow, they colonised splenic stroma with long-term production of L-DC. The maintenance of HSPC by splenic stroma was confirmed when non-adherent cells collected from LTC showed oligopotent reconstitution of the hematopoietic compartment of lethally irradiated mice. All data support a model whereby spleen houses a niche for HSPC in the resting state, with production of progenitors, and their differentiation to give tissue-specific antigen presenting cells.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/physiology , Spleen/cytology , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Coculture Techniques , Hematopoietic Stem Cell Transplantation , Immune System/cytology , Mice, Inbred C57BL , Stem Cell Niche
12.
Curr Opin Immunol ; 28: 58-63, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24608070

ABSTRACT

Vaccination against intracellular pathogens requires generation of a pool of memory T cells able to respond upon infection and mediate either killing of the infected cell or induce killing mechanisms in the infected cell. T cell-inducing vaccines must aim to target the antigen to antigen-presenting cells (APCs) so that it can be presented on MHC molecules on the cell surface. Methods to do this include making use of vectors such as plasmid DNA or viruses, live attenuated pathogens or subunit vaccines targeted and enhanced using adjuvants. The choice of approach should be guided by the phenotype and localization of the desired T cell response. This review will discuss current approaches in the pipeline for the development of T cell-inducing vaccines, including vectored, live attenuated, and subunit vaccines.


Subject(s)
Vaccines/immunology , Animals , Genetic Vectors , Humans , Intracellular Space/immunology , T-Lymphocytes/immunology
13.
Tuberculosis (Edinb) ; 94(3): 226-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24572168

ABSTRACT

The efficacy of Bacillus Calmette-Guerin (BCG) vaccination in protection against pulmonary tuberculosis (TB) is highly variable between populations. One possible explanation for this variability is increased exposure of certain populations to non-tuberculous mycobacteria (NTM). This study used a murine model to determine the effect that exposure to NTM after BCG vaccination had on the efficacy of BCG against aerosol Mycobacterium tuberculosis challenge. The effects of administering live Mycobacterium avium (MA) by an oral route and killed MA by a systemic route on BCG-induced protection were evaluated. CD4+ and CD8+ T cell responses were profiled to define the immunological mechanisms underlying any effect on BCG efficacy. BCG efficacy was enhanced by exposure to killed MA administered by a systemic route; T helper 1 and T helper 17 responses were associated with increased protection. BCG efficacy was reduced by exposure to live MA administered by the oral route; T helper 2 cells were associated with reduced protection. These findings demonstrate that exposure to NTM can induce opposite effects on BCG efficacy depending on route of exposure and viability of NTM. A reproducible model of NTM exposure would be valuable in the evaluation of novel TB vaccine candidates.


Subject(s)
Adjuvants, Immunologic/pharmacology , BCG Vaccine/pharmacology , Mycobacterium avium Complex/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Inhalation , Administration, Oral , Aerosols , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Immunity, Cellular/immunology , Lymph Nodes/immunology , Mice, Inbred C57BL , Spleen/immunology , Th1 Cells/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/pharmacology , Vaccines, Live, Unattenuated/immunology , Vaccines, Live, Unattenuated/pharmacology
14.
PLoS One ; 8(10): e78312, 2013.
Article in English | MEDLINE | ID: mdl-24194918

ABSTRACT

Interleukin (IL)-17 is emerging as an important cytokine in vaccine-induced protection against tuberculosis disease in animal models. Here we show that compared to parenteral delivery, BCG delivered mucosally enhances cytokine production, including interferon gamma and IL-17, in the lungs. Furthermore, we find that cholera toxin, delivered mucosally along with BCG, further enhances IL-17 production by CD4(+) T cells over mucosal BCG alone both in the lungs and systemically. This boosting effect of CT is also observed using a vaccine regimen of BCG followed by the candidate vaccine MVA85A. Using a murine Mycobacterium tuberculosis (M.tb) aerosol challenge model, we demonstrate the ability of cholera toxin delivered at the time of a priming BCG vaccination to improve protection against tuberculosis disease in a manner at least partially dependent on the observed increase in IL-17. This observed increase in IL-17 in the lungs has no adverse effect on lung pathology following M.tb challenge, indicating that IL-17 can safely be boosted in murine lungs in a vaccine/M.tb challenge setting.


Subject(s)
BCG Vaccine/pharmacology , Cholera Toxin/pharmacology , Interleukin-17/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Analysis of Variance , Animals , BCG Vaccine/immunology , Mice , Statistics, Nonparametric , Tuberculosis/immunology
15.
J Cell Mol Med ; 17(12): 1608-18, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24251878

ABSTRACT

A novel dendritic-like cell subset termed L-DC was recently identified in murine spleen based on marker expression of a homogeneous cell population derived from long-term culture of neonatal spleen. The function of L-DC is distinct from other splenic dendritic and myeloid cell subsets because of their high endocytic capacity and their ability to cross-present antigen to CD8(+) T cells. This paper shows the subset to be unique to spleen and blood, with a similar, but possibly functionally distinct subset also present in bone marrow. The prevalence of the subset is low; ~6% of all dendritic and myeloid cells in the spleen and ~5% in blood. However, they are a distinct cell type on the basis of marker expression, and endocytic and T-cell stimulatory capacity. Attempts to identify an enriched population of these cells in mutant mouse strains with reported increases in myelopoiesis showed either a lack of L-DC or an altered phenotype reflective of the phenotype of the mouse strain.


Subject(s)
Dendritic Cells/cytology , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , Endocytosis , Female , Histocompatibility Antigens Class II/metabolism , Homeodomain Proteins/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/cytology , Monocytes/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , Phenotype , Spleen/cytology , Spleen/metabolism
16.
PLoS One ; 8(7): e67922, 2013.
Article in English | MEDLINE | ID: mdl-23844129

ABSTRACT

A better understanding of the relationships between vaccine, immunogenicity and protection from disease would greatly facilitate vaccine development. Modified vaccinia virus Ankara expressing antigen 85A (MVA85A) is a novel tuberculosis vaccine candidate designed to enhance responses induced by BCG. Antigen-specific interferon-γ (IFN-γ) production is greatly enhanced by MVA85A, however the variability between healthy individuals is extensive. In this study we have sought to characterize the early changes in gene expression in humans following vaccination with MVA85A and relate these to long-term immunogenicity. Two days post-vaccination, MVA85A induces a strong interferon and inflammatory response. Separating volunteers into high and low responders on the basis of T cell responses to 85A peptides measured during the trial, an expansion of circulating CD4+ CD25+ Foxp3+ cells is seen in low but not high responders. Additionally, high levels of Toll-like Receptor (TLR) 1 on day of vaccination are associated with an increased response to antigen 85A. In a classification model, combined expression levels of TLR1, TICAM2 and CD14 on day of vaccination and CTLA4 and IL2Rα two days post-vaccination can classify high and low responders with over 80% accuracy. Furthermore, administering MVA85A in mice with anti-TLR2 antibodies may abrogate high responses, and neutralising antibodies to TLRs 1, 2 or 6 or HMGB1 decrease CXCL2 production during in vitro stimulation with MVA85A. HMGB1 is released into the supernatant following atimulation with MVA85A and we propose this signal may be the trigger activating the TLR pathway. This study suggests an important role for an endogenous ligand in innate sensing of MVA and demonstrates the importance of pattern recognition receptors and regulatory T cell responses in determining the magnitude of the antigen specific immune response to vaccination with MVA85A in humans.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HMGB1 Protein/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptors/metabolism , Tuberculosis Vaccines/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Animals , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Mice , Middle Aged , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Toll-Like Receptors/genetics , Vaccination , Vaccines, DNA , Young Adult
17.
Tuberculosis (Edinb) ; 93(5): 551-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23726784

ABSTRACT

Development of an improved vaccine against tuberculosis (TB) is hindered by the lack of a surrogate of protection. Efficacy of new TB vaccines in humans can only be evaluated by expensive and time consuming efficacy trials within TB endemic areas. It is critical that vaccines with the greatest potential to protect are selected for these trials. Mycobacterial growth inhibition assays (MGIAs) have been developed with the hope that these in-vitro functional assays will correlate with protection, which could aid in the selection of the best vaccine candidates. The present study describes the use of the BACTEC system to perform MGIAs in mice. We demonstrate reproducible mycobacterial growth inhibition in splenocytes from BCG immunised mice compared with unimmunised mice (P < 0.023), which corresponded with in-vivo efficacy against Mycobacterium tuberculosis (M. tb) challenge. Microarray data showed extensive differential gene expression in splenocyte responses to ex-vivo BCG stimulation between unimmunised and BCG-immunised mice. TH1 responses, including IFN-γ, nitric oxide synthase (NOS2) and Interleukin -17 (IL-17) expression were enhanced in BCG immunised mice, indicating a possible mechanism for mycobacterial growth inhibition. Further investigation into whether the BACTEC MGIA can be used as a surrogate of protection in humans and preclinical animal models is now warranted.


Subject(s)
BCG Vaccine/immunology , Mycobacterium bovis/growth & development , Spleen/microbiology , Tuberculosis/prevention & control , Animals , Cells, Cultured , Colony Count, Microbial , Cytokines/biosynthesis , Cytokines/genetics , Disease Models, Animal , Female , Gene Expression Regulation/immunology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/isolation & purification , Spleen/cytology , Spleen/metabolism , Tuberculosis/genetics , Tuberculosis/immunology
18.
Vaccine ; 31(7): 1026-33, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23266342

ABSTRACT

BACKGROUND: New vaccines to prevent tuberculosis are urgently needed. MVA85A is a novel viral vector TB vaccine candidate designed to boost BCG-induced immunity when delivered intradermally. To date, intramuscular delivery has not been evaluated. Skin and muscle have distinct anatomical and immunological properties which could impact upon vaccine-mediated cellular immunity. METHODS: We conducted a randomised phase I trial comparing the safety and immunogenicity of 1×10(8)pfu MVA85A delivered intramuscularly or intradermally to 24 healthy BCG-vaccinated adults. RESULTS: Intramuscular and intradermal MVA85A were well tolerated. Intradermally-vaccinated subjects experienced significantly more local adverse events than intramuscularly-vaccinated subjects, with no difference in systemic adverse events. Both routes generated strong and sustained Ag85A-specific IFNγ T cell responses and induced multifunctional CD4+ T cells. The frequencies of CD4+ T cells expressing chemokine receptors CCR4, CCR6, CCR7 and CXCR3 induced by vaccination was similar between routes. CONCLUSIONS: In this phase I trial the intramuscular delivery of MVA85A was well tolerated and induced strong, durable cellular immune responses in healthy BCG vaccinated adults, comparable to intradermal delivery. These findings are important for TB vaccine development and are of relevance to HIV, malaria, influenza and other intracellular pathogens for which T cell-inducing MVA-based vaccine platforms are being evaluated.


Subject(s)
Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Adolescent , Adult , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Gene Expression , Humans , Injections, Intradermal , Injections, Intramuscular , Interferon-gamma/metabolism , Male , Middle Aged , Receptors, Chemokine/biosynthesis , T-Lymphocytes/immunology , Tuberculosis Vaccines/administration & dosage , Vaccines, DNA , Viral Vaccines/administration & dosage , Young Adult
19.
Stem Cells Int ; 2011: 954275, 2011.
Article in English | MEDLINE | ID: mdl-22190965

ABSTRACT

While spleen and other secondary tissue sites contribute to hematopoiesis, the nature of cells produced and the environment under which this happens are not fully defined. Evidence is reviewed here for hematopoiesis occurring in the spleen microenvironment leading to the production of tissue-specific antigen presenting cells. The novel dendritic-like cell identified in spleen is phenotypically and functionally distinct from other described antigen presenting cells. In order to identify these cells as distinct, it has been necessary to show that their lineage origin and progenitors differ from that of other known dendritic and myeloid cell types. The spleen therefore represents a distinct microenvironment for hematopoiesis of a novel myeloid cell arising from self-renewing hematopoietic stem cells (HSC) or progenitors endogenous to spleen.

20.
BMJ Open ; 1(2): e000223, 2011.
Article in English | MEDLINE | ID: mdl-22102640

ABSTRACT

Objectives Control of the tuberculosis (TB) epidemic is a global health priority and one that is likely to be achieved only through vaccination. The critical overlap with the HIV epidemic requires any effective TB vaccine regimen to be safe in individuals who are infected with HIV. The objectives of this clinical trial were to evaluate the safety and immunogenicity of a leading candidate TB vaccine, MVA85A, in healthy, HIV-infected adults. Design This was an open-label Phase I trial, performed in 20 healthy HIV-infected, antiretroviral-naïve subjects. Two different doses of MVA85A were each evaluated as a single immunisation in 10 subjects, with 24 weeks of follow-up. The safety of MVA85A was assessed by clinical and laboratory markers, including regular CD4 counts and HIV RNA load measurements. Vaccine immunogenicity was assessed by ex vivo interferon γ (IFN-γ) ELISpot assays and flow-cytometric analysis. Results MVA85A was safe in subjects with HIV infection, with an adverse-event profile comparable with historical data from previous trials in HIV-uninfected subjects. There were no clinically significant vaccine-related changes in CD4 count or HIV RNA load in any subjects, and no evidence from qPCR analyses to indicate that MVA85A vaccination leads to widespread preferential infection of vaccine-induced CD4 T cell populations. Both doses of MVA85A induced an antigen-specific IFN-γ response that was durable for 24 weeks, although of a lesser magnitude compared with historical data from HIV-uninfected subjects. The functional quality of the vaccine-induced T cell response in HIV-infected subjects was remarkably comparable with that observed in healthy HIV-uninfected controls, but less durable. Conclusion MVA85A is safe and immunogenic in healthy adults infected with HIV. Further safety and efficacy evaluation of this candidate vaccine in TB- and HIV-endemic areas is merited.

SELECTION OF CITATIONS
SEARCH DETAIL
...