Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1494: 353-368, 2017.
Article in English | MEDLINE | ID: mdl-27718207

ABSTRACT

Adjuvants in vaccine formulations are designed to enhance immune responses against a target antigen or pathogen. The ability of these vaccines to induce activation and differentiation of mature naïve B cells to produce pathogen-specific antibodies (immunoglobulins; Ig) helps guarantee long-lived humoral immunity. This process involves clonal expansion of antigen-specific B cells, genomic rearrangement of Ig heavy (IgH) and light (IgL) loci, somatic hypermutation (SHM), and clonal selection for affinity-matured antibody, resulting in a vast but directed repertoire of B cells expressing highly specific antibody proteins. High-throughput sequencing of the IgH and IgL complementary determining regions (CDRs) derived from various B cell populations provides an unprecedented way to observe dynamic responses of the humoral immune repertoire in response to vaccination. However, applying high-throughput sequencing (HTS) methodologies to multi-armed in vivo experiments requires careful coordination of sample preparation with downstream bioinformatics, particularly with regard to issues of quantitation, sequence fidelity, bar-coding, and multiplexing strategies. Here, we overview strategies of high-throughput sequencing and analysis of the adaptive immune complex loci applied to multi-armed, multiplexed experiments.


Subject(s)
Adjuvants, Immunologic/pharmacology , B-Lymphocytes/immunology , Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Vaccines/pharmacology , Animals , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Mice
2.
Clin Transl Immunology ; 4(4): e35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26175894

ABSTRACT

Key antigens of Leishmania species identified in the context of host responses in Leishmania-exposed individuals from disease-endemic areas were prioritized for the development of a subunit vaccine against visceral leishmaniasis (VL), the most deadly form of leishmaniasis. Two Leishmania proteins-nucleoside hydrolase and a sterol 24-c-methyltransferase, each of which are protective in animal models of VL when properly adjuvanted- were produced as a single recombinant fusion protein NS (LEISH-F3) for ease of antigen production and broad coverage of a heterogeneous major histocompatibility complex population. When formulated with glucopyranosyl lipid A-stable oil-in-water nanoemulsion (GLA-SE), a Toll-like receptor 4 TH1 (T helper 1) promoting nanoemulsion adjuvant, the LEISH-F3 polyprotein induced potent protection against both L. donovani and L. infantum in mice, measured as significant reductions in liver parasite burdens. A robust immune response to each component of the vaccine with polyfunctional CD4 TH1 cell responses characterized by production of antigen-specific interferon-γ, tumor necrosis factor and interleukin-2 (IL-2), and low levels of IL-5 and IL-10 was induced in immunized mice. We also demonstrate that CD4 T cells, but not CD8 T cells, are sufficient for protection against L. donovani infection in immunized mice. Based on the sum of preclinical data, we prepared GMP materials and performed a phase 1 clinical study with LEISH-F3+GLA-SE in healthy, uninfected adults in the United States. The vaccine candidate was shown to be safe and induced a strong antigen-specific immune response, as evidenced by cytokine and immunoglobulin subclass data. These data provide a strong rationale for additional trials in Leishmania-endemic countries in populations vulnerable to VL.

3.
J Immunol ; 194(2): 505-13, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505290

ABSTRACT

Chemokine-dependent localization of specific B cell subsets within the immune microarchitecture is essential to ensure successful cognate interactions. Although cognate interactions between T cells and memory B cells (B(mem)) are essential for the secondary humoral immune responses, the chemokine response patterns of B(mem) cells are largely unknown. In contrast to naive B cells, this study shows that Ag-specific B(mem) cells have heightened expression of CCR6 and a selective chemotactic response to the CCR6 ligand, CCL20. Although CCR6 appears be nonessential for the initial clonal expansion and maintenance of B(mem), CCR6 is essential for the ability of B(mem) to respond to a recall response to their cognate Ag. This dependency was deemed intrinsic by studies in CCR6-deficient mice and in bone marrow chimeric mice where CCR6 deficiency was limited to the B cell lineage. Finally, the mis-positioning of CCR6-deficient B(mem) was revealed by immunohistological analysis with an altered distribution of CCR6-deficient B(mem) from the marginal and perifollicular to the follicular/germinal center area.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Chemotaxis/immunology , Immunologic Memory/physiology , Receptors, CCR6/immunology , Allografts , Animals , B-Lymphocytes/cytology , Bone Marrow Transplantation , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Chemotaxis/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Germinal Center/cytology , Germinal Center/immunology , Mice , Mice, Knockout , Receptors, CCR6/genetics , Transplantation Chimera/immunology
4.
Infect Immun ; 81(7): 2318-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23589575

ABSTRACT

In livers of susceptible but self-curing C57BL/6 mice, intracellular Leishmania donovani infection enhanced Toll-like receptor 4 (TLR4) and TLR2 gene expression. In the liver, infected TLR4(-/-) mice showed reduced gamma interferon (IFN-γ), tumor necrosis factor (TNF), and inducible nitric oxide synthase (iNOS) mRNA expression, higher-level and slowly resolving infection, delayed granuloma formation, and little response to low-dose chemotherapy; in serum, the ratio of IFN-γ to interleukin 10 (IL-10) activity was decreased by 50%. In contrast, in TLR2(-/-) mice, control of liver infection, parasite killing, and granuloma assembly were accelerated and chemotherapy's efficacy enhanced. In livers of infected TLR2(-/-) mice, mRNA expression was not increased for inflammatory cytokines or iNOS or decreased for IL-10; however, the serum IFN-γ/IL-10 ratio was increased 6.5-fold and minimal responses to IL-10 receptor blockade suggested downregulated IL-10. In established infection in wild-type mice, blockading TLR2 induced parasite killing and triggering TLR4 strengthened resistance and promoted chemotherapy's effect. Thus, in experimental L. donovani infection in the liver, TLR4 signaling upregulates and TLR2 signaling downregulates macrophage antileishmanial activity, making both receptors potential therapeutic targets in visceral leishmaniasis for engagement (TLR4) or blockade (TLR2).


Subject(s)
Gene Expression Regulation , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/parasitology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Animals , Antiprotozoal Agents/pharmacology , Disease Models, Animal , Female , Interferon-gamma/blood , Interleukin-10/genetics , Interleukin-10/immunology , Leishmaniasis, Visceral/drug therapy , Lipopeptides/pharmacology , Liver/drug effects , Liver/immunology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
5.
Vaccine ; 31(5): 813-9, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23228811

ABSTRACT

Despite the advances toward the elimination of leprosy through widespread provision of multi-drug therapy to registered patients over the last 2 decades, new case detection rates have stabilized and leprosy remains endemic in a number of localized regions. A vaccine could overcome the inherent limitations of the drug treatment program by providing protection in individuals who are not already harboring the Mycobacterium leprae bacilli at the time of administration and effectively interrupt the transmission cycle over a wider timespan. In this report we present data validating the production of 73f, a chimeric fusion protein incorporating the M. leprae antigens ML2028, ML2346 and ML2044. The 73f protein was recognized by IgG in multibacillary (MB) leprosy patient sera and stimulated IFNγ production within whole blood assays of paucibacillary (PB) leprosy patient and healthy household contacts of MB patients (HHC). When formulated with a TLR4L-containing adjuvant (GLA-SE), 73f stimulated a strong and pluripotent Th1 response that inhibited M. leprae-induced inflammation in mice. We are using these data to develop new vaccine initiatives for the continued and long-term control of leprosy.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Leprosy/prevention & control , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Female , Humans , Immunoglobulin G/blood , Interferon-gamma/metabolism , Leprosy/immunology , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mycobacterium leprae/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Th1 Cells/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Young Adult
6.
Front Immunol ; 3: 144, 2012.
Article in English | MEDLINE | ID: mdl-22701453

ABSTRACT

Two million new cases of leishmaniasis occur every year, with the cutaneous leishmaniasis (CL) presentation accounting for approximately two-thirds of all cases. Despite the high incidence rates and geographic expansion of the disease, CL remains a neglected tropical disease without effective intervention strategies. Efforts to address this deficit have given rise to the experimental murine model of CL. By virtue of its simplicity and pliability, the CL model has been used to provide substantial information regarding cellular immunity, as well as in the discovery and evaluation of various vaccine adjuvants. The CL model has facilitated in vivo studies of the mechanism of action of many adjuvants, including the TLR4 agonist monophosphoryl lipid A, the TLR7/8 agonist imiquimod, the TLR9 agonist CpG, adenoviral vectors, and the immunostimulatory complexes. Together, these studies have helped to unveil the requirement for certain types of immune responses at specific stages of CL disease and provide a basis to aid the design of effective second-generation vaccines for human CL. This review focuses on adjuvants that have been tested in experimental CL, outlining how they have helped advance our understanding of the disease and ultimately, how they have performed when applied within clinical trials against human CL.

7.
Vaccine ; 30(2): 134-41, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22085553

ABSTRACT

Infection with Leishmania parasites results in a range of clinical manifestations and outcomes. Control of Leishmania parasite transmission is extremely difficult due to the large number of vectors and potential reservoirs, and none of the current treatments are ideal. Vaccination could be an effective strategy to provide sustained control. In this review, the current global situation with regard to leishmaniasis, the immunology of Leishmania infection and various efforts to identify second generation vaccine candidates are briefly discussed. The variety of clinical trials conducted using the only current second generation vaccine approved for clinical use, LEISH-F1+MPL-SE, are described. Given that epidemiological evidence suggests that reducing the canine reservoir also positively impacts human incidence, efforts at providing a vaccine for leishmaniasis in dogs are highlighted. Finally, potential refinements and surrogate markers that could expedite the introduction of a vaccine that can limit the severity and incidence of leishmaniasis are discussed.


Subject(s)
Leishmaniasis Vaccines/administration & dosage , Leishmaniasis Vaccines/immunology , Leishmaniasis/epidemiology , Leishmaniasis/prevention & control , Animals , Clinical Trials as Topic , Disease Reservoirs , Dog Diseases/prevention & control , Dog Diseases/transmission , Dogs , Global Health , Humans , Incidence , Leishmaniasis Vaccines/adverse effects
8.
Sci Transl Med ; 3(93): 93ra69, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21795589

ABSTRACT

Vaccination with an isolated antigen is frequently not sufficient to elicit a protective immune response. The addition of adjuvants to the antigen can increase the magnitude and breadth of the response generated, but quantification of this increase as a function of adjuvant has been intractable. We have directly determined the variation of the immunoglobulin G variable-chain repertoire of an entire organism as a function of vaccination. Using the well-established Plasmodium vivax antigen, PvRII, and massively parallel sequencing, we showed that the use of a Toll-like receptor (TLR) agonist in the vaccine formulation increased the diversity of the variable region sequences in comparison to the use of an oil-in-water emulsion adjuvant alone. Moreover, increased variable domain diversity in response to the use of TLR agonist-based adjuvants correlated with improved antigen neutralization. The use of TLR agonists also broadened the range of polymorphic variants against which these antibodies could be effective. In addition, a peptide microarray demonstrated that inclusion of adjuvants changed the profile of linear epitopes from PvRII that were recognized by serum from immunized animals. The results of these studies have broad implications for vaccine design--they may enable tailored adjuvants that elicit the broad spectrum of antibodies required to neutralize drifted and polymorphic pathogen strains as well as provide a method for rapid determination of correlates of adjuvant-induced humoral immunity.


Subject(s)
Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Toll-Like Receptors/immunology , Adjuvants, Immunologic , Amino Acid Sequence , Animals , Antibodies, Protozoan/chemistry , Antibody Formation/immunology , DNA, Complementary/genetics , Epitopes/immunology , Immunization , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Mice , Mice, Inbred BALB C , Peptides/immunology
9.
Clin Vaccine Immunol ; 18(7): 1118-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21632891

ABSTRACT

A subunit vaccine using a defined antigen(s) may be one effective solution for controlling leishmaniasis. Because of genetic diversity in target populations, including both dogs and humans, a multiple-antigen vaccine will likely be essential. However, the cost of a vaccine to be used in developing countries must be considered. We describe herein a multiantigen vaccine candidate comprised of antigens known to be protective in animal models, including dogs, and to be recognized by humans immune to visceral leishmaniasis. The polyprotein (KSAC) formulated with monophosphoryl lipid A, a widely used adjuvant in human vaccines, was found to be immunogenic and capable of inducing protection against Leishmania infantum, responsible for human and canine visceral leishmaniasis, and against L. major, responsible for cutaneous leishmaniasis. The results demonstrate the feasibility of producing a practical, cost-effective leishmaniasis vaccine capable of protecting both humans and dogs against multiple Leishmania species.


Subject(s)
Leishmaniasis, Visceral/prevention & control , Polyproteins/therapeutic use , Adjuvants, Immunologic , Animals , Antibodies, Protozoan , Antigens, Protozoan , Dogs , Humans , Leishmania infantum/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/veterinary , Lipid A/analogs & derivatives , Polyproteins/immunology , Protozoan Vaccines
10.
Vaccine ; 29(17): 3284-92, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21349366

ABSTRACT

GMZ2 adjuvanted by aluminum hydroxide is a candidate malaria vaccine that has successfully passed phase 1 clinical testing in adult German and Gabonese volunteers and Gabonese children under five. Here we report a preclinical study screening a series of adjuvant vehicles and Toll-like receptor (TLR) agonists in CB6F1 mice to identify an improved formulation of GMZ2 suitable for further human clinical studies. GMZ2 formulated in an oil-in-water emulsion plus the synthetic TLR4 agonist GLA elicits the highest (a) vaccine-specific IgG2a and total IgG titers, (b) parasite-specific IFA titers, (c) levels of Type 1 cytokine responses (IFN-γ), and (d) number of long-lived-plasma cells (LLPC) secreting antibodies against both the GMZ2 fusion and its two components. Thus, GLA helps to elicit a vaccine-specific Type 1 antibody profile together with high levels of LLPC, both of which are thought to be essential for the development of long-term protective immunity against clinical malaria.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Protozoan/blood , Lipid A/administration & dosage , Malaria Vaccines/immunology , Th1 Cells/immunology , Toll-Like Receptor 4/agonists , Animals , Female , Immunoglobulin G/blood , Malaria Vaccines/administration & dosage , Mice , Mice, Inbred C57BL
11.
PLoS One ; 5(10): e13677, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-21060869

ABSTRACT

Safe, effective adjuvants that enhance vaccine potency, including induction of neutralizing Abs against a broad range of variant strains, is an important strategy for the development of seasonal influenza vaccines which can provide optimal protection, even during seasons when available vaccines are not well matched to circulating viruses. We investigated the safety and ability of Glucopyranosyl Lipid Adjuvant-Stable Emulsion (GLA-SE), a synthetic Toll-like receptor (TLR)4 agonist formulation, to adjuvant Fluzone® in mice and non-human primates. The GLA-SE adjuvanted Fluzone vaccine caused no adverse reactions, increased the induction of T helper type 1 (T(H)1)-biased cytokines such as IFNγ, TNF and IL-2, and broadened serological responses against drifted A/H1N1 and A/H3N2 influenza variants. These results suggest that synthetic TLR4 adjuvants can enhance the magnitude and quality of protective immunity induced by influenza vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza Vaccines/immunology , Animals , Cross Reactions , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Cellular , Mice , Mice, Inbred BALB C , Primates
12.
J Immunol ; 185(3): 1701-10, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20601594

ABSTRACT

Therapy of intracellular pathogens can be complicated by drug toxicity, drug resistance, and the need for prolonged treatment regimens. One approach that has shown promise is immunotherapy. Leishmaniasis, a vector-borne disease ranked among the six most important tropical infectious diseases by the World Health Organization, has been treated clinically with crude or defined vaccine preparations or cytokines, such as IFN-gamma and GM-CSF, in combination with chemotherapy. We have attempted to develop an improved and defined immunotherapeutic using a mouse model of cutaneous leishmaniasis. We hypothesized that immunotherapy may be improved by using TLR synergy to enhance the parasite-specific immune response. We formulated L110f, a well-established Leishmania poly-protein vaccine candidate, in conjunction with either monophosphoryl lipid A, a TLR4 agonist, or CpG, a TLR9 agonist, or a combination of these, and evaluated anti-Leishmania immune responses in absence or presence of active disease. Only mice treated with L110f plus monophosphoryl lipid A-CpG were able to induce a strong effective T cell response during disease and subsequently cured lesions and reduced parasite burden when compared with mice treated with L110f and either single adjuvant. Our data help to define a correlate of protection during active infection and indicate TLR synergy to be a potentially valuable tool in treating intracellular infections.


Subject(s)
Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/therapy , Toll-Like Receptors/physiology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Cytoskeletal Proteins/physiology , Drug Therapy, Combination , Female , Interleukin-12/biosynthesis , Leishmaniasis, Cutaneous/microbiology , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Pyrin , Toll-Like Receptors/agonists
13.
Infect Immun ; 77(12): 5623-30, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19786561

ABSTRACT

Leprosy elimination has been a goal of the WHO for the past 15 years. Widespread BCG vaccination and multidrug therapy have dramatically reduced worldwide leprosy prevalence, but new case detection rates have remained relatively constant. These data suggest that additional control strategies, such as a subunit vaccine, are required to block transmission and to improve leprosy control. We recently identified several Mycobacterium leprae antigens that stimulate gamma interferon (IFN-gamma) secretion upon incubation with blood from paucibacillary leprosy patients, a group who limit M. leprae growth and dissemination. In this study, we demonstrate that M. leprae-specific mouse T-cell lines recognize several of these antigens, with the ML0276 protein stimulating the most IFN-gamma secretion. We then examined if the ML0276 protein could be used in a subunit vaccine to provide protection against experimental M. leprae infection. Our data demonstrate that combining ML0276 with either a Toll-like receptor 4 (TLR4) (EM005), TLR7 (imiquimod), or TLR9 (CpG DNA) agonist during immunization induces Th1 responses that limit local inflammation upon experimental M. leprae infection. Our data indicate that only the ML0276/EM005 regimen is able to elicit a response that is transferable to recipient mice. Despite the potent Th1 response induced by this regimen, it could not provide protection in terms of limiting bacterial growth. We conclude that EM005 is the most potent adjuvant for stimulating a Th1 response and indicate that while a subunit vaccine containing the ML0276 protein may be useful for the prevention of immune pathology during leprosy, it will not control bacterial burden and is therefore unlikely to interrupt disease transmission.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Inflammation/prevention & control , Leprosy/immunology , Leprosy/prevention & control , Mycobacterium leprae/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Colony Count, Microbial , Female , Inflammation/immunology , Interferon-gamma/metabolism , Leprosy/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Toll-Like Receptors/agonists , Vaccines, Subunit/immunology
14.
Vaccine ; 27(43): 5956-63, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19679214

ABSTRACT

Impairments in anti-influenza T helper 1 (Th1) responses are associated with greater risk of influenza-related mortality in the elderly. Addition of adjuvants to existing influenza vaccines could improve immune responses in the elderly. In this study, the activity of three adjuvants, an oil-in-water emulsion and a synthetic lipid A adjuvant formulated with or without the emulsion, is compared. Our results show that Fluzone combined with lipid A plus an emulsion effectively leads to greater vaccine-specific IgG2a and IgG titers, enhances hemagglutination-inhibition titers and induces Type 1 cytokine responses (IFN-gamma and IL-2) to each of the Fluzone components.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Lipid A/pharmacology , Orthomyxoviridae Infections/prevention & control , Toll-Like Receptor 4/agonists , Animals , Antibodies, Viral/blood , Antibody Formation , Cell Line , Emulsions/pharmacology , Female , Hemagglutination Inhibition Tests , Immunity, Cellular , Immunoglobulin G/immunology , Interferon-gamma/immunology , Interleukin-2/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Th1 Cells/immunology
15.
Vaccine ; 27(21): 2884-90, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19428898

ABSTRACT

The enzyme sterol 24-c-methyltranferase (SMT) is required for the biosynthesis of ergosterol, the major membrane sterol in Leishmania parasites. SMT and ergosterol are not found in mammals, so this protein may be an attractive target for anti-leishmanial vaccines and drugs. We have previously demonstrated that SMT from L. infantum, which causes visceral leishmaniasis, is a protective antigen against this parasite. Because this protein is highly conserved among Leishmania species, we evaluated the potential of SMT to cross-protect against a different form of leishmaniasis. Here, we show that immunization with L. infantum SMT, formulated with monophosphoryl lipid A in stable emulsion (MPL-SE), protects mice from cutaneous leishmaniasis caused by L. major. In BALB/c mice the vaccine preparation induced antigen-specific multi-functional CD4(+) T cells capable of producing IFN-gamma, IL-2, and/or TNF-alpha upon antigen re-exposure, and MPL-SE was indispensable to direct immune responses to SMT towards Th1. Mice immunized with the SMT/MPL-SE vaccine developed significantly smaller lesions following ear challenge with L. major. These results suggest that SMT is a promising vaccine antigen for multiple forms of leishmaniasis.


Subject(s)
Leishmania infantum/enzymology , Leishmania infantum/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Methyltransferases/immunology , Methyltransferases/metabolism , Animals , Antigens, Protozoan/immunology , Cells, Cultured , Cricetinae , Cross Reactions/immunology , Emulsions , Female , Methyltransferases/genetics , Mice , Th1 Cells/immunology , Th2 Cells/immunology
16.
Infect Immun ; 75(11): 5290-7, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17724073

ABSTRACT

Leprosy is caused by infection with Mycobacterium leprae. The immune response of leprosy patients can be highly diverse, ranging from strong cellular responses accompanied by an apparent deficit of M. leprae-specific antibodies to strong humoral responses with a deficit of cell-mediated responses. Leprosy takes many years to manifest, and this has precluded analyses of disease and immune response development in infected humans. In an attempt to better define development of the immune response during leprosy we have developed an M. leprae ear infection model. Intradermal inoculation of M. leprae into the ear supported not only infection but also the development of a chronic inflammatory response. The inflammatory response was localized, comprising a T-cell infiltration into the ear and congestion of cells in the draining lymph nodes. The development of local chronic inflammation was prevented by rifampin treatment. Importantly, and in contrast to subcutaneous M. leprae footpad infection, systemic M. leprae-specific gamma interferon and antibody responses were detected following intradermal ear infection. These results indicate the utility of intradermal ear infection for both induction and understanding of the immune response during M. leprae infection and the identification or testing of new leprosy treatments.


Subject(s)
Antibody Formation , Disease Models, Animal , Ear/microbiology , Immunity, Cellular , Mycobacterium leprae/immunology , Animals , Antibodies, Bacterial/blood , Female , Inflammation/immunology , Inflammation/pathology , Injections, Intradermal , Interferon-gamma/biosynthesis , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocytes/immunology
17.
Infect Immun ; 75(9): 4648-54, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17606603

ABSTRACT

The Leishmania-derived recombinant polyprotein Leish-111f or its three component proteins, thiol-specific antioxidant (TSA), Leishmania major stress-inducible protein 1 (LmSTI1), and Leishmania elongation initiation factor (LeIF), have previously been demonstrated to be efficacious against cutaneous or mucosal leishmaniasis in mice, nonhuman primates, and humans. In this study we demonstrate that Leish-111f is also a vaccine antigen candidate against visceral leishmaniasis (VL) caused by Leishmania infantum. We evaluated the immune response and protection induced by Leish-111f formulated with monophosphoryl lipid A in a stable emulsion (Leish-111f+MPL-SE) and demonstrated that mice developed strong humoral and T-cell responses to the vaccine antigen. Analysis of the cellular immune responses of immunized, uninfected mice demonstrated that the vaccine induced a significant increase in CD4(+) T cells producing gamma interferon, interleukin 2, and tumor necrosis factor cytokines, indicating a Th1-type immune response. Experimental infection of immunized mice and hamsters demonstrated that Leish-111f+MPL-SE induced significant protection against L. infantum infection, with reductions in parasite loads of 99.6%, a level of protection greater than that reported for other vaccine candidates in animal models of VL. Taken together, our results suggest that this vaccine represents a good candidate for use against several Leishmania species. The Leish-111f+MPL-SE product we report here is the first defined vaccine for leishmaniasis in human clinical trials and has completed phase 1 and 2 safety and immunogenicity testing in normal, healthy human subjects.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Leishmaniasis, Visceral/prevention & control , Lymphocyte Activation/immunology , Polyproteins/immunology , Protozoan Vaccines/immunology , Animals , Cells, Cultured , Cricetinae , Female , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/immunology , Leishmaniasis, Visceral/immunology , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Lipid A/immunology , Mesocricetus , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Initiation Factors/administration & dosage , Peptide Initiation Factors/immunology , Peroxidases/administration & dosage , Peroxidases/immunology , Peroxiredoxins , Polyproteins/administration & dosage , Protozoan Proteins/administration & dosage , Protozoan Proteins/immunology , Protozoan Vaccines/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
18.
Blood ; 110(1): 193-200, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17360936

ABSTRACT

The recruitment of tumor necrosis factor receptor-associated factors (TRAFs) 1, 2, 3, 5, and 6 to the CD40 cytoplasmic tail upon CD40 trimerization results in downstream signaling events that ultimately lead to CD40-dependent, thymus-dependent (TD) humoral immune responses. Previously, we have shown signaling through the C-terminal tail of CD40 in the absence of canonical TRAF-binding sites is capable of signaling through an alternative TRAF2-binding site. Here, we demonstrate that B cells from mice harboring CD40 with only the C-terminal tail can activate both canonical and noncanonical NFkappaB signaling pathways. Moreover, while lacking germinal center formation, several hallmarks of humoral immune responses including clonal B-cell activation/expansion, antibody isotype switching, and affinity maturation remain normal. This study demonstrates a new functional domain in CD40 that controls critical aspects of B-cell immunity in an in vivo setting.


Subject(s)
B-Lymphocytes/cytology , CD40 Antigens/physiology , TNF Receptor-Associated Factor 2/metabolism , Animals , Antibody Formation , Binding Sites , CD40 Antigens/chemistry , CD40 Antigens/immunology , Cell Differentiation , Cell Proliferation , Mice , NF-kappa B/metabolism , Signal Transduction
19.
Immunol Lett ; 109(2): 93-100, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17374402

ABSTRACT

Long-term humoral immunity elicited by pathogens and vaccines alike relies upon the generation of both memory B cells (B(mem)) and long-lived plasma cells (PCs). Virtually all vaccine formulations induce the concomitant emergence of both B(mem) and PCs, suggesting that the emergence of these two differentiated B cells subsets is commonly controlled. Evidence presented shows specific Toll-like receptor (TLR) agonists coupled with soluble protein antigen (sAg) can selectively induce the expansion of antigen specific B(mem) in the absence of PC generation. The co-administration of either TLR 3 or 9 agonists with sAg induced germinal centre (GC) formation, antigen-specific B(mem), but failed to substantively induce the generation of long-lived bone marrow (BM) PCs. Upon re-challenge, high levels of PCs were induced with concomitant high titres of antigen-specific serum IgG. Hence, vaccines can be developed that can prime and protect the host to subsequent infectious agents without initial, high levels of antibody production. Furthermore, these studies suggest that the signals that govern the expansion and differentiation of B(mem) can be uncoupled from those that induce long-lived BM PCs.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Toll-Like Receptors/agonists , Vaccines/immunology , Animals , Antigens/immunology , Epitopes, B-Lymphocyte/immunology , Freund's Adjuvant/immunology , Germinal Center/immunology , Lipids/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasma Cells/immunology
20.
J Exp Med ; 199(1): 91-8, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14707116

ABSTRACT

Long-lived humoral immunity is manifested by the ability of bone marrow plasma cells (PCs) to survive for extended periods of time. Recent studies have underscored the importance of BLyS and APRIL as factors that can support the survival of B lineage lymphocytes. We show that BLyS can sustain PC survival in vitro, and this survival can be further enhanced by interleukin 6. Selective up-regulation of Mcl-1 in PCs by BLyS suggests that this alpha-apoptotic gene product may play an important role in PC survival. Blockade of BLyS, via transmembrane activator and cyclophilin ligand interactor-immunoglobulin treatment, inhibited PC survival in vitro and in vivo. Heightened expression of B cell maturation antigen (BCMA), and lowered expression of transmembrane activator and cyclophilin ligand interactor and BAFF receptor in PCs relative to resting B cells suggests a vital role of BCMA in PC survival. Affirmation of the importance of BCMA in PC survival was provided by studies in BCMA-/- mice in which the survival of long-lived bone marrow PCs was impaired compared with wild-type controls. These findings offer new insights into the molecular basis for the long-term survival of PCs.


Subject(s)
Bone Marrow Cells/cytology , Cell Survival/physiology , Plasma Cells/cytology , Receptors, Tumor Necrosis Factor/physiology , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , Base Sequence , Bone Marrow Cells/physiology , DNA Primers , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/physiology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...