Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Immunology ; 131(1): 128-40, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20465573

ABSTRACT

A better understanding of similarities and differences in the composition of the cellular immune system in non-human primates (NHPs) compared with human subjects will improve the interpretation of preclinical studies. It will also aid in addressing the usefulness of NHPs as subjects for studying chronic diseases, vaccine development and immune reconstitution. We employed high content colour flow cytometry and analysed simultaneously the expression of CD3, CD4, CD8alpha, CD8beta, CD16/CD56, CD45RA, CCR7, CD27, CD28, CD107a and the interleukin-7 receptor alpha-chain (IL-7Ralpha) in peripheral blood mononuclear cells (PBMCs) of 27 rhesus macaques and 16 healthy human subjects. Regulatory T cells (Tregs) were identified using anti-CD3, -CD4, -CD25, -FoxP3, and -IL-7Ralpha monoclonal antibodies. Responsiveness to IL-7 was gauged in a signal transducer and activation of transcription 5 (STAT-5) phosphorylation assay. Human and NHP PBMCs showed a similar T-cell composition pattern with some remarkable differences. Similarities: human and NHP CD4(+) and CD8(+) cells showed a similar STAT-5 phosphorylation pattern in response to IL-7. Multicolour flow cytometric analysis identified a CD4(+) CD8alphaalpha(+) CD8alphabeta(+) T-cell population in NHPs as well as in human subjects that expressed the degranulation marker CD107a and may represent a unique CD4(+) T-cell subset endowed with cytotoxic capacity. Differences: we identified in PBMCs from NHPs a higher proportion (5.16% in CD3(+) T cells) of CD8alphaalpha(+) T cells when compared with human donors (1.22% in CD3(+) T cells). NHP CD8alphaalpha(+) T cells produced tumour necrosis factor-alpha / interferon-gamma (TNF-alpha/IFN-gamma) or TNF-alpha, whereas human CD8alphaalpha(+) T cells produced simultaneously TNF-alpha/IFN-gamma and IL-2. A minor percentage of human CD8(+) T cells expressed CD25(bright) and FoxP3 (0.01%). In contrast, 0.07% of NHP CD8(+) T cells exhibited the CD25(bright) FoxP3(+) phenotype. PBMCs from NHPs showed less IL-7Ralpha-positive events in all T-cell subsets including CD4(+) Tregs (median 5%) as compared with human (median 12%). The data visualize commonalities and differences in immune cell subsets in humans and NHPs, most of them in long-lived memory cells and cells with suppressive functions. This provides a matrix to assess future efforts to study diseases and vaccines in NHPs.


Subject(s)
Immunity, Cellular , Macaca mulatta/immunology , T-Lymphocyte Subsets/immunology , Adult , Animals , Cytokines/metabolism , Female , Flow Cytometry , Humans , Interleukin-7/immunology , Phosphorylation , STAT5 Transcription Factor/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/metabolism , Young Adult
2.
Vaccine ; 28(4): 1084-93, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19896449

ABSTRACT

Despite the extensive success with the introduction of M. bovis Bacille Calmette-Guérin (BCG), tuberculosis (TB) remains a major global epidemic infecting between 8 and 9 million people annually with an estimated 1.7 million deaths each year. However, because of its demonstrated effectiveness against some of the most severe forms of childhood TB, it is now realized that BCG vaccination of newborns is unlikely to be replaced. Therefore, BCG or an improved BCG will continue to be used as a prime TB vaccine and there is a need to develop effective boost vaccines that would enhance and prolong the protective immunity induced by BCG prime immunization. We report on a heterologous booster approach using two highly immunogenic TB antigens comprising Ag85B and TB10.4 (HyVac4) delivered as a fusion molecule and formulated in the proprietary adjuvant IC31. This vaccine was found to be immunogenic and demonstrated greater protection in the more stringent guinea pig model of pulmonary tuberculosis than BCG alone when used in a prime/boost regimen. Significant difference in lung involvement was observed for all animals in the HyVac4 boosted group compared to BCG alone regardless of time to death or sacrifice. A vaccine toxicology study of the HyVac4:IC31 regimen was performed and it was judged safe to advance the vaccine into clinical trials. Therefore, all non-clinical data supports the suitability of HyVac4 as a safe, immunogenic, and effective vaccination in a prime-boost regimen with BCG.


Subject(s)
Immunization, Secondary/methods , Oligodeoxyribonucleotides/adverse effects , Oligopeptides/adverse effects , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Animals , Drug Combinations , Female , Guinea Pigs , Lung/immunology , Lung/pathology , Oligodeoxyribonucleotides/administration & dosage , Oligopeptides/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Tuberculosis Vaccines/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology
3.
Immunology ; 129(4): 496-505, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20002212

ABSTRACT

The molecular definition of major histocompatibility complex (MHC) class I-presented CD8(+) T-cell epitopes from clinically relevant Mycobacterium tuberculosis (Mtb) target proteins will aid in the rational design of T-cell-based diagnostics of tuberculosis (TB) and the measurement of TB vaccine-take. We used an epitope discovery system, based on recombinant MHC class I molecules that cover the most frequent Caucasian alleles [human leucocyte antigen (HLA)-A*0101, A*0201, A*0301, A*1101, A*2402, B*0702, B*0801 and B*1501], to identify MHC class I-binding peptides from overlapping 9-mer peptides representing the Mtb protein TB10.4. A total of 33 MHC class I-binding epitopes were identified, spread across the entire amino acid sequence, with some clustering at the N- and C-termini of the protein. Binding of individual peptides or closely related peptide species to different MHC class I alleles was frequently observed. For instance, the common motif of xIMYNYPAMx bound to six of eight alleles. Affinity (50% effective dose) and off-rate (half life) analysis of candidate Mtb peptides will help to define the conditions for CD8(+) T-cell interaction with their nominal MHC class I-peptide ligands. Subsequent construction of tetramers allowed us to confirm the recognition of some of the epitopes by CD8(+) T cells from patients with active pulmonary TB. HLA-B alleles served as the dominant MHC class I restricting molecules for anti-Mtb TB10.4-specific CD8(+) T-cell responses measured in CD8(+) T cells from patients with pulmonary TB.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , HLA-B Antigens/immunology , Histocompatibility Antigens Class I/immunology , Mycobacterium tuberculosis/immunology , Peptides/immunology , Binding Sites , HLA-B7 Antigen , HLA-B8 Antigen , Humans , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/immunology
4.
Vaccine ; 27(33): 4412-23, 2009 Jul 16.
Article in English | MEDLINE | ID: mdl-19500523

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), has infected approximately two billion individuals worldwide with approximately 9.2 million new cases and 1.6 million deaths annually. Current efforts are focused on making better BCG priming vaccines designed to induce a comprehensive and balanced immunity followed by booster(s) targeting a specific set of relevant antigens in common with the BCG prime. We describe the generation and immunological characterization of recombinant BCG strains with properties associated with lysis of the endosome compartment and over-expression of key Mtb antigens. The endosome lysis strain, a derivative of BCG SSI-1331 (BCG(1331)) expresses a mutant form of perfringolysin O (PfoA(G137Q)), a cytolysin normally secreted by Clostridium perfringens. Integration of the PfoA(G137Q) gene into the BCG genome was accomplished using an allelic exchange plasmid to replace ureC with pfoA(G137Q) under the control of the Ag85B promoter. The resultant BCG construct, designated AERAS-401 (BCG(1331) DeltaureC::OmegapfoA(G137Q)) secreted biologically active Pfo, was well tolerated with a good safety profile in immunocompromised SCID mice. A second rBCG strain, designated AFRO-1, was generated by incorporating an expression plasmid encoding three mycobacterial antigens, Ag85A, Ag85B and TB10.4, into AERAS-401. Compared to the parental BCG strain, vaccination of mice and guinea pigs with AFRO-1 resulted in enhanced immune responses. Mice vaccinated with AFRO-1 and challenged with the hypervirulent Mtb strain HN878 also survived longer than mice vaccinated with the parental BCG. Thus, we have generated improved rBCG vaccine candidates that address many of the shortcomings of the currently licensed BCG vaccine strains.


Subject(s)
Antigens, Bacterial/immunology , BCG Vaccine/immunology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Immunodominant Epitopes/immunology , Tuberculosis/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/genetics , Bacterial Toxins/genetics , Cell Line , Erythrocytes/microbiology , Female , Genes, Bacterial , Guinea Pigs , Hemolysin Proteins/genetics , Hemolysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Mycobacterium bovis/genetics , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Plasmids , Sheep , Tuberculosis/immunology
5.
Proc Natl Acad Sci U S A ; 106(7): 2301-6, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19188599

ABSTRACT

The development of a vaccine for tuberculosis requires a combination of antigens and adjuvants capable of inducing appropriate and long-lasting T cell immunity. We evaluated Mtb72F formulated in AS02A in the cynomolgus monkey model. The vaccine was immunogenic and caused no adverse reactions. When monkeys were immunized with bacillus Calmette-Guérin (BCG) and then boosted with Mtb72F in AS02A, protection superior to that afforded by using BCG alone was achieved, as measured by clinical parameters, pathology, and survival. We observed long-term survival and evidence of reversal of disease progression in monkeys immunized with the prime-boost regimen. Antigen-specific responses from protected monkeys receiving BCG and Mtb72F/AS02A had a distinctive cytokine profile characterized by an increased ratio between 3 Th1 cytokines, IFN-gamma, TNF, and IL-2 and an innate cytokine, IL-6. To our knowledge, this is an initial report of a vaccine capable of inducing long-term protection against tuberculosis in a nonhuman primate model, as determined by protection against severe disease and death, and by other clinical and histopathological parameters.


Subject(s)
Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Cytokines/metabolism , Disease Progression , Haplorhini , Immune System , Interferon-gamma/metabolism , Interleukin-6/metabolism , Macaca fascicularis , Mycobacterium tuberculosis/metabolism , Time Factors , Treatment Outcome , Tuberculosis/microbiology , Tuberculosis Vaccines/chemistry
6.
Vaccine ; 27(2): 223-33, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19000730

ABSTRACT

To identify Mycobacterium tuberculosis (Mtb) antigens as candidates for a subunit vaccine against tuberculosis (TB), we have employed a CD4+ T-cell expression screening method. Mtb-specific CD4+ T-cell lines from nine healthy PPD positive donors were stimulated with different antigenic substrates including autologous dendritic cells (DC) infected with Mtb, or cultured with culture filtrate proteins (CFP), and purified protein derivative of Mtb (PPD). These lines were used to screen a genomic Mtb library expressed in Escherichia coli and processed and presented by autologous DC. This screening led to the recovery of numerous T-cell antigens, including both novel and previously described antigens. One of these novel antigens, referred to as Mtb9.8 (Rv0287), was recognized by multiple T-cell lines, stimulated with either Mtb-infected DC or CFP. Using the mouse and guinea pig models of TB, high levels of IFN-gamma were produced, and solid protection from Mtb challenge was observed following immunization with Mtb9.8 formulated in either AS02A or AS01B Adjuvant Systems. These results demonstrate that T-cell screening of the Mtb genome can be used to identify CD4+ T-cell antigens that are candidates for vaccine development.


Subject(s)
Antigens, Bacterial/genetics , CD4-Positive T-Lymphocytes/metabolism , Cloning, Molecular/methods , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Amino Acid Sequence , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Line , Epitope Mapping , Escherichia coli/genetics , Escherichia coli/metabolism , Guinea Pigs , Humans , Lymphocyte Activation , Mice , Molecular Sequence Data , Sequence Alignment , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/prevention & control , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics
7.
PLoS One ; 3(11): e3790, 2008.
Article in English | MEDLINE | ID: mdl-19023426

ABSTRACT

BACKGROUND: BCG vaccination, combined with adenoviral-delivered boosts, represents a reasonable strategy to augment, broaden and prolong immune protection against tuberculosis (TB). We tested BCG (SSI1331) (in 6 animals, delivered intradermally) and a recombinant (rBCG) AFRO-1 expressing perfringolysin (in 6 animals) followed by two boosts (delivered intramuscullary) with non-replicating adenovirus 35 (rAd35) expressing a fusion protein composed of Ag85A, Ag85B and TB10.4, for the capacity to induce antigen-specific cellular immune responses in rhesus macaques (Macaca mulatta). Control animals received diluent (3 animals). METHODS AND FINDINGS: Cellular immune responses were analyzed longitudinally (12 blood draws for each animal) using intracellular cytokine staining (TNF-alpha, IL-2 and IFN-gamma), T cell proliferation was measured in CD4(+), CD8alpha/beta(+), and CD8alpha/alpha(+) T cell subsets and IFN-gamma production was tested in 7 day PBMC cultures (whole blood cell assay, WBA) using Ag85A, Ag85B, TB10.4 recombinant proteins, PPD or BCG as stimuli. Animals primed with AFRO-1 showed i) increased Ag85B-specific IFN-gamma production in the WBA assay (median >400 pg/ml for 6 animals) one week after the first boost with adenoviral-delivered TB-antigens as compared to animals primed with BCG (<200 pg/ml), ii) stronger T cell proliferation in the CD8alpha/alpha(+) T cell subset (proliferative index 17%) as compared to BCG-primed animals (proliferative index 5% in CD8alpha/alpha(+) T cells). Polyfunctional T cells, defined by IFN-gamma, TNF-alpha and IL-2 production were detected in 2/6 animals primed with AFRO-1 directed against Ag85A/b and TB10.4; 4/6 animals primed with BCG showed a Ag85A/b responses, yet only a single animal exhibited Ag85A/b and TB10.4 reactivity. CONCLUSION: AFRO-1 induces qualitatively and quantitatively different cellular immune responses as compared with BCG in rhesus macaques. Increased IFN-gamma-responses and antigen-specific T cell proliferation in the CD8alpha/alpha+ T cell subset represents a valuable marker for vaccine-take in BCG-based TB vaccine trials.


Subject(s)
BCG Vaccine/administration & dosage , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Antigens, Bacterial/genetics , BCG Vaccine/genetics , Bacterial Toxins/genetics , Female , Genetic Vectors , Hemolysin Proteins/genetics , Immunity, Cellular , Immunization, Secondary , Interferon-gamma/biosynthesis , Lymphocyte Activation , Macaca mulatta , Tuberculosis Vaccines/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics
8.
Infect Immun ; 75(8): 4105-15, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526747

ABSTRACT

There is an urgent need for an efficacious vaccine against tuberculosis (TB). Cellular immune responses are key to an effective protective response against TB. Recombinant adenovirus (rAd) vectors are especially suited to the induction of strong T-cell immunity and thus represent promising vaccine vehicles for the prevention of TB. We have previously reported on rAd vector serotype 35, the serotype of choice due to low preexisting immunity worldwide, which expresses a unique fusion protein of Mycobacterium tuberculosis antigens Ag85A, Ag85B, and TB10.4 (Ad35-TBS). Here, we demonstrate that Ad35-TBS confers protection against M. tuberculosis when administered to mice through either an intranasal or an intramuscular route. Histological evaluation of lung tissue corroborated the protection and, in addition, demonstrated differences between two mouse strains, with diffuse inflammation in BALB/c mice and distinct granuloma formation in C57BL/6 mice. Epitope mapping analysis in these mouse strains showed that the major T-cell epitopes are conserved in the artificial fusion protein, while three novel CD8 peptides were discovered. Using a defined set of T-cell epitopes, we reveal differences between the two mouse strains in the type of protective immune response, demonstrating that different antigen-specific gamma interferon (IFN-gamma)-producing T cells can provide protection against M. tuberculosis challenge. While in BALB/c (H-2(d)) mice, a dominant CD8 T-cell response was detected, in C57BL/6 (H-2(b)) mice, more balanced CD4/CD8 T-cell responses were observed, with a more pronounced CD4 response in the lungs. These results unify conflicting reports on the relative importance of CD4 versus CD8 T-cell responses in protection and emphasize the key role of IFN-gamma.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Interferon-gamma/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Viral Vaccines/immunology , Acyltransferases/genetics , Acyltransferases/immunology , Adenoviridae/genetics , Administration, Intranasal , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Colony Count, Microbial , Disease Models, Animal , Epitope Mapping , Female , Genetic Vectors , Injections, Intramuscular , Liver/microbiology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spleen/microbiology , Tuberculosis Vaccines/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/genetics
9.
Microbiology (Reading) ; 152(Pt 12): 3667-3681, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17159220

ABSTRACT

Despite accumulating data implicating Propionibacterium acnes in a variety of diseases, its precise role in infection remains to be determined. P. acnes antigen-specific CD4(+) T cells are present in early inflamed acne lesions and may be involved in the inflammatory response; however, little is known about the specific antigens involved. In this study, B cell and T cell antigens from P. acnes expression libraries were cloned and evaluated and the four predominant proteins identified were investigated. Two of these antigens share some homology with an M-like protein of Streptococcus equi and have dermatan-sulphate-binding activity (PA-25957 and 5541). The remaining two antigens, PA-21693 and 4687, are similar to the product of the Corynebacterium diphtheriae htaA gene from the hmu ABC transport locus, although only one of these (PA-21693) is encoded within an hmu-like operon and conserved amongst a range of clinical isolates. All four proteins contain an LPXTG motif, although only PA-21693 contains a characteristic sortase-sorting signal. Variation in the expression of PA-4687, 25957 and 5541 is evident amongst clinical isolates and is generated both by frameshifts associated with the putative signal peptide and by variable numbers of repeat regions toward the carboxy-terminus, potentially generating heterogeneity of molecular mass and antigenic variation. In addition, in the case of PA-25957, a frameshift in a C-rich region at the extreme carboxy-terminus eliminates the LPXTG motif in some isolates. For the dermatan-sulphate-binding PA-25957, IgG1 antibody in serum from acne-positive donors was shown to be specific for the amino-terminal region of the protein, which also contains a CD4(+) T cell epitope. In contrast, serum from acne-negative donors shows an IgG2 and IgG3 antibody subclass response to the carboxy-terminal region. These data have implications for the potential role of P. acnes in inflammatory acne and other diseases.


Subject(s)
Acne Vulgaris/immunology , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Propionibacterium acnes/immunology , Propionibacterium acnes/metabolism , Amino Acid Sequence , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/genetics , Cloning, Molecular , Conserved Sequence , Corynebacterium diphtheriae/genetics , Dermatan Sulfate/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Frameshift Mutation , Gene Expression , Genetic Variation , Humans , Immunoblotting , Immunoglobulin G/immunology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Propionibacterium acnes/genetics , Protein Binding , Protein Sorting Signals/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sequence Homology, Amino Acid , Streptococcus equi/genetics
10.
Nat Rev Microbiol ; 4(6): 469-76, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16710326

ABSTRACT

Tuberculosis (TB), an ancient human scourge, is a growing health problem in the developing world. Approximately two million deaths each year are caused by TB, which is the leading cause of death in HIV-infected individuals. Clearly, an improved TB vaccine is desperately needed. Heterologous prime-boost regimens probably represent the best hope for an improved vaccine regimen to prevent TB. This first generation of new vaccines might also complement drug treatment regimens and be effective against reactivation of TB from the latent state, which would significantly enhance their usefulness.


Subject(s)
Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/standards , Tuberculosis/prevention & control , Vaccination , BCG Vaccine/immunology , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Time , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccination/standards , Vaccination/trends , Vaccines, Attenuated/immunology , Vaccines, Subunit/immunology
11.
Infect Immun ; 74(4): 2392-401, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16552069

ABSTRACT

Using a rabbit model of tuberculous meningitis, we evaluated the protective efficacy of vaccination with the recombinant polyprotein Mtb72F, which is formulated in two alternative adjuvants, AS02A and AS01B, and compared this to vaccination with Mycobacterium bovis bacillus Calmette-Guérin (BCG) alone or as a BCG prime/Mtb72F-boost regimen. Vaccination with Mtb72F formulated in AS02A (Mtb72F+AS02A) or Mtb72F formulated in AS01B (Mtb72F+AS01B) was protective against central nervous system (CNS) challenge with Mycobacterium tuberculosis H37Rv to an extent comparable to that of vaccination with BCG. Similar accelerated clearances of bacilli from the cerebrospinal fluid, reduced leukocytosis, and less pathology of the brain and lungs were noted. Weight loss of infected rabbits was less extensive for Mtb72F+AS02A-vaccinated rabbits. In addition, protection against M. tuberculosis H37Rv CNS infection afforded by BCG/Mtb72F in a prime-boost strategy was similar to that by BCG alone. Interestingly, Mtb72F+AS01B induced better protection against leukocytosis and weight loss, suggesting that the polyprotein in this adjuvant may boost immunity without exacerbating inflammation in previously BCG-vaccinated individuals.


Subject(s)
Bacterial Proteins/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Meningeal/immunology , Tuberculosis, Meningeal/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Proteins/administration & dosage , Body Weight/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Disease Models, Animal , Drug Combinations , Drug Evaluation, Preclinical , Immunity, Active , Immunization, Secondary , Lipid A/administration & dosage , Lipid A/analogs & derivatives , Lipid A/immunology , Lung/immunology , Lung/pathology , Mycobacterium bovis/immunology , Rabbits , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Saponins/administration & dosage , Saponins/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Meningeal/pathology
12.
Vaccine ; 23(30): 3937-45, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15917115

ABSTRACT

MTB41 is a Mycobacterium antigen that is recognized by CD4+ T cells early after experimental infection of mice with Mycobacterium tuberculosis and by PBMC from healthy PPD positive individuals. Immunization of mice with plasmid DNA encoding the MTB41 gene sequence results in the development of antigen-specific CD4+ and CD8+ T cells, and protection against challenge with virulent M. tuberculosis. In the present studies, in contrast to DNA immunization, we show, that a strong MTB41-specific CD4+ T cell response, but no MHC class I restricted cytotoxic T lymphocyte (CTL) activity is detected in the spleen cells of infected mice. Therefore, this data suggests that the induction of CD8+ T cell response to MTB41 epitopes by DNA immunization may not be relevant to protection because these epitopes are not recognized during the infectious process. We also compared the repertoire of rMTB41 epitope recognition by CD4+ T cells of M. tuberculosis-infected mice with the recognition repertoire of mice immunized with the recombinant rMTB41 protein. Both regimens of sensitization lead to the recognition of the same molecular epitope. Coincidentally, immunization with the soluble recombinant protein plus adjuvant, a regimen known to generate primarily CD4+ T cells, resulted in induction of protection comparable to BCG in two well-established animal models of tuberculosis (mice and guinea pigs).


Subject(s)
Antigens, Bacterial/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , Colony Count, Microbial , Cytokines/analysis , Cytokines/immunology , Epitopes/immunology , Guinea Pigs , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Spleen/cytology , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccines, Synthetic/immunology
13.
Vaccine ; 23(17-18): 2132-5, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15755583

ABSTRACT

We have developed two novel tuberculosis (TB) vaccines: a DNA vaccine combination expressing mycobacterial heat shock protein 65 (Hsp65) and interleukin-12 (IL-12) by using the hemagglutinating virus of Japan (HVJ)-liposome (HSP65+IL-12/HVJ) and a recombinant BCG harboring the 72f fusion gene (72f rBCG). These vaccines provide remarkable protective efficacy in mouse and guinea pig models, as compared to the current by available BCG vaccine. In the present study, we extended our studies to a cynomolgus monkey model, which is currently the best animal model of human tuberculosis, to evaluate the HSP65+IL-12/HVJ and 72f rBCG vaccines. Vaccination with HSP65+IL-12/HVJ as well as 72f rBCG vaccines provided better protective efficacy as assessed by the Erythrocyte Sedimentation Rate, chest X-ray findings and immune responses than BCG. Most importantly, HSP65+IL-12/HVJ resulted in an increased survival for over a year. This is the first report of successful DNA vaccination and recombinant BCG vaccination against M. tuberculosis in the monkey model.


Subject(s)
BCG Vaccine/pharmacology , Tuberculosis Vaccines/pharmacology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Chaperonin 60 , Chaperonins/genetics , Chaperonins/immunology , Disease Models, Animal , Guinea Pigs , Humans , Interleukin-12/genetics , Liposomes , Macaca fascicularis , Mice , Sendai virus/genetics , Tuberculosis Vaccines/genetics , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology
14.
Infect Immun ; 72(11): 6622-32, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501795

ABSTRACT

A tuberculosis vaccine candidate consisting of a 72-kDa polyprotein or fusion protein based upon the Mtb32 and Mtb39 antigens of Mycobacterium tuberculosis and designated Mtb72F was tested for its protective capacity as a potential adjunct to the Mycobacterium bovis BCG vaccine in the mouse and guinea pig models of this disease. Formulation of recombinant Mtb72F (rMtb72F) in an AS02A adjuvant enhanced the Th1 response to BCG in mice but did not further reduce the bacterial load in the lungs after aerosol challenge infection. In the more stringent guinea pig disease model, rMtb72F delivered by coadministration with BCG vaccination significantly improved the survival of these animals compared to BCG alone, with some animals still alive and healthy in their appearance at >100 weeks post-aerosol challenge. A similar trend was observed with guinea pigs in which BCG vaccination was boosted by DNA vaccination, although this increase was not statistically significant due to excellent protection conferred by BCG alone. Histological examination of the lungs of test animals indicated that while BCG controls eventually died from overwhelming lung consolidation, the majority of guinea pigs receiving BCG mixed with rMtb72F or boosted twice with Mtb72F DNA had mostly clear lungs with minimal granulomatous lesions. Lesions were still prominent in guinea pigs receiving BCG and the Mtb72F DNA boost, but there was considerable evidence of lesion healing and airway remodeling and reestablishment. These data support the hypothesis that the coadministration or boosting of BCG vaccination with Mtb72F may limit the lung consolidation seen with BCG alone and may promote lesion resolution and healing. Collectively, these data suggest that enhancing BCG is a valid vaccination strategy for tuberculosis that is worthy of clinical evaluation.


Subject(s)
BCG Vaccine/immunology , Bacterial Proteins/immunology , Immunization, Secondary , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Guinea Pigs , Humans , Lung/pathology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Polyproteins/administration & dosage , Polyproteins/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/mortality , Tuberculosis, Pulmonary/pathology , Vaccination , Vaccines, Synthetic/immunology
15.
J Immunol ; 172(12): 7618-28, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15187142

ABSTRACT

Key Ags of Mycobacterium tuberculosis initially identified in the context of host responses in healthy purified protein derivative-positive donors and infected C57BL/6 mice were prioritized for the development of a subunit vaccine against tuberculosis. Our lead construct, Mtb72F, codes for a 72-kDa polyprotein genetically linked in tandem in the linear order Mtb32(C)-Mtb39-Mtb32(N). Immunization of C57BL/6 mice with Mtb72F DNA resulted in the generation of IFN-gamma responses directed against the first two components of the polyprotein and a strong CD8(+) T cell response directed exclusively against Mtb32(C). In contrast, immunization of mice with Mtb72F protein formulated in the adjuvant AS02A resulted in the elicitation of a moderate IFN-gamma response and a weak CD8(+) T cell response to Mtb32c. However, immunization with a formulation of Mtb72F protein in AS01B adjuvant generated a comprehensive and robust immune response, resulting in the elicitation of strong IFN-gamma and Ab responses encompassing all three components of the polyprotein vaccine and a strong CD8(+) response directed against the same Mtb32(C) epitope identified by DNA immunization. All three forms of Mtb72F immunization resulted in the protection of C57BL/6 mice against aerosol challenge with a virulent strain of M. tuberculosis. Most importantly, immunization of guinea pigs with Mtb72F, delivered either as DNA or as a rAg-based vaccine, resulted in prolonged survival (>1 year) after aerosol challenge with virulent M. tuberculosis comparable to bacillus Calmette-Guérin immunization. Mtb72F in AS02A formulation is currently in phase I clinical trial, making it the first recombinant tuberculosis vaccine to be tested in humans.


Subject(s)
Immunization/methods , Mycobacterium tuberculosis , Tuberculosis Vaccines/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemical synthesis , Base Sequence , DNA, Bacterial/administration & dosage , Female , Guinea Pigs , Immunity , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Polyproteins/chemical synthesis , Polyproteins/immunology , Polyproteins/therapeutic use , Survival Rate , Tuberculosis Vaccines/chemistry , Tuberculosis, Pulmonary/prevention & control , Tuberculosis, Pulmonary/therapy , Vaccines, DNA , Vaccines, Synthetic
16.
Methods Mol Med ; 94: 91-106, 2004.
Article in English | MEDLINE | ID: mdl-14959824

ABSTRACT

Expression cloning involves the selection of specific polypeptides, generated from a cDNA or genomic DNA library, based on certain characteristics of the expressed proteins, such as antibody or ligand binding, recognition by T-cells, function, or complementation of cell defects. Here we describe the detailed construction of a genomic, random shear lambda expression library, adsorption of anti Escherichia coli antibody from antiserum, the screening of an expression library with specific antisera, and the cloning of genes with potential use in the diagnosis of infectious disease. This approach has been used successfully by our laboratory for the discovery of antigenic components of diagnostics and vaccines for several infectious agents including: Mycobacterium tuberculosis, Anaplasma phagocytophila (formerly Ehrlichia spp. or E. phagocytophila), Babesia microti, Trypanosoma cruzi, Leishmania chagasi, and Chlamydia spp.


Subject(s)
Cloning, Molecular/methods , Gene Expression , Proteins/genetics , Proteins/immunology , Animals , Antibodies , Antibodies, Bacterial , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Genomic Library , Humans , Immunosorbent Techniques , Peptide Library , Serologic Tests
17.
Infect Immun ; 72(1): 381-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14688119

ABSTRACT

Recombinant immunodominant mycobacterial antigens are needed for the development of new vaccines and immunodiagnostic tools for use against tuberculosis. Ubiquitous exposure to mycobacteria in tropical countries could influence vaccine-induced immunity and the specificity of tuberculosis immunodiagnosis. For this study conducted in The Gambia, cellular immune responses to recombinant mycobacterial antigens were characterized in Mycobacterium bovis BCG-vaccinated and nonvaccinated infants, adult community controls, household contacts, health care workers, and tuberculosis patients. Neonatal BCG vaccination induced gamma interferon (IFN-gamma) responses to Mtb8.4, Mtb32-C, Mtb39A, Mtb9.9A, and Mtb32-N, but not CFP-10 (Mtb11) and alpha-crystallin (Mtb16). Exposure to Mycobacterium tuberculosis in household contacts and health care workers was associated with high responses to CFP-10 and alpha-crystallin. Generally, low IFN-gamma responses were found in tuberculosis patients. These results suggest that Mtb8.4, Mtb32-C, Mtb39A, Mtb9.9A, and Mtb32-N may be used in a subunit vaccine to boost BCG-induced immunity. While CFP-10 and alpha-crystallin are promising candidates for the immunodiagnosis of M. tuberculosis infection or for vaccine use, disease-associated immunosuppression may prevent IFN-gamma immunodiagnosis of more advanced tuberculosis.


Subject(s)
Antigens, Bacterial/immunology , BCG Vaccine/immunology , Endemic Diseases , Mycobacterium/immunology , Tuberculosis, Pulmonary/epidemiology , Tuberculosis, Pulmonary/prevention & control , Adult , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Gambia/epidemiology , Humans , Infant , Infant, Newborn , Interferon-gamma/biosynthesis , Mycobacterium tuberculosis/immunology , Recombinant Proteins/immunology , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/immunology , Vaccination
18.
Infect Immun ; 71(9): 5065-76, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12933849

ABSTRACT

Several members of the Trypanosomatidae family, when freshly isolated from their mammalian hosts, have immunoglobulins adsorbed to their cell surfaces. However, a significant portion of these antibody molecules is not parasite specific, i.e., the immunoglobulins are bound to the parasite's cell surface molecules via noncognitive interactions. It has been proposed that this noncognitive adsorption of immunoglobulins to the parasite is mediated by an Fc-like receptor present in several members of the Trypanosomatidae family. However, the molecular identification of this receptor has never been defined. Here, we describe the cloning of a gene encoding a protein that might represent this molecule. The gene, named Lmsp1, was cloned by screening a Leishmania major cDNA expression library using a rabbit antiserum. Lmsp1 is present in both Leishmania and Trypanosoma and is expressed in all developmental stages of these parasites. The predicted protein has a molecular mass of 16.6 kDa and contains an RGD sequence starting at residue 104 and three cysteine residues at positions 55, 74, and 116. The purified recombinant protein strongly binds to normal immunoglobulins of various animal species (humans, rabbits, sheep, goats, guinea pigs, donkeys, rats, and mice) and the binding to human immunoglobulins appears to be immunoglobulin G (IgG) and IgM isotype specific. Moreover, Lmsp1 binds to both purified Fc and Fab fragments of IgG from both humans and rabbits. The mapping of the Lmsp1 epitopes that bind human IgG revealed that different sequences of the molecule bind to Fc or Fab. In addition, fluorescence-activated cell sorter analyses with a specific rabbit anti-Lmsp1 antiserum showed that Lmsp1 is associated with the parasite's cell surface. Finally, inhibition experiments point to an active role of this molecule in the immunoglobulin-mediated attachment and penetration of Trypanosoma cruzi in its macrophage host cells, thus suggesting that Lmsp1 is a putative Trypanosomatidae immunoglobulin receptor.


Subject(s)
Genes, Protozoan , Immunoglobulins/metabolism , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Trypanosomatina/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cloning, Molecular , DNA, Complementary/genetics , DNA, Protozoan/genetics , Humans , In Vitro Techniques , Leishmania major/genetics , Leishmania major/immunology , Leishmania major/metabolism , Mice , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Rabbits , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Sequence Homology, Amino Acid , Trypanosoma/genetics , Trypanosoma/immunology , Trypanosoma/metabolism , Trypanosomatina/immunology , Trypanosomatina/metabolism
19.
Protein Expr Purif ; 30(1): 124-33, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821330

ABSTRACT

Expression of recombinant proteins is an important step towards elucidating the functions of many genes discovered through genomic sequencing projects. It is also critical for validating gene targets and for developing effective therapies for many diseases. Here we describe a novel method to express recombinant proteins that are extremely difficult to produce otherwise. The increased protein expression level is achieved by using a fusion partner, MTB32-C, which is the carboxyl terminal fragment of the Mycobacterium tuberculosis antigen, MTB32 (Rv0125). By fusing MTB32-C to the N-termini of target genes, we have demonstrated significant enhancement of recombinant protein expression level in Escherichia coli. The inclusion of a 6xHis tag and the 128-amino acid of MTB32-C will add 13.5 kDa to the fusion molecule. Comparison of the mRNA levels of the fusion and non-fusion proteins indicated that the increased fusion protein expression may be regulated at translational or post-translational steps. There are many potential applications for the generated fusion proteins. For example, MTB32-C fusion proteins have been used successfully as immunogens to generate both polyclonal and monoclonal antibodies. These antibodies have been used to characterize cellular localization of the proteins and to validate gene targets at protein level. In addition, these antibodies may be useful in diagnostic and therapeutic applications for many diseases. If desired, the MTB32-C portion in the fusion protein can be removed after protein expression, making it possible to study protein structure and function as well as to screen for potential drugs. Thus, this novel fusion expression system has become a powerful tool for many applications.


Subject(s)
Biotechnology/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Base Sequence , Cell Line , Escherichia coli , Flow Cytometry , Gene Expression , Genetic Engineering/methods , Genetic Vectors , Humans , Immunohistochemistry , Molecular Sequence Data , Mycobacterium tuberculosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification
20.
J Immunol ; 170(6): 3171-9, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626575

ABSTRACT

Upon loading with microbial Ag and adoptive transfer, dendritic cells (DC) are able to induce immunity to infections. This offers encouragement for the development of DC-based vaccination strategies. However, the mechanisms underlying the adjuvant effect of DC are not fully understood, and there is a need to identify Ag with which to arm DC. In the present study, we analyzed the role of DC-derived IL-12 in the induction of resistance to Leishmania major, and we evaluated the protective efficacy of DC loaded with individual Leishmania Ag. Using Ag-pulsed Langerhans cells (LC) from IL-12-deficient or wild-type mice for immunization of susceptible animals, we showed that the inability to release IL-12 completely abrogated the capacity of LC to mediate protection against leishmaniasis. This suggests that the availability of donor LC-derived IL-12 is a requirement for the development of protective immunity. In addition, we tested the protective effect of LC loaded with Leishmania homolog of receptor for activated C kinase, gp63, promastigote surface Ag, kinetoplastid membrane protein-11, or Leishmania homolog of eukaryotic ribosomal elongation and initiation factor 4a. The results show that mice vaccinated with LC that had been pulsed with selected molecularly defined parasite proteins are capable of controlling infection with L. major. Moreover, the protective potential of DC pulsed with a given Leishmania Ag correlated with the level of their IL-12 expression. Analysis of the cytokine profile of mice after DC-based vaccination revealed that protection was associated with a shift toward a Th1-type response. Together, these findings emphasize the critical role of IL-12 produced by the sensitizing DC and suggest that the development of a DC-based subunit vaccine is feasible.


Subject(s)
Antigens, Protozoan/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/parasitology , Interleukin-12/physiology , Intracellular Fluid/immunology , Intracellular Fluid/parasitology , Leishmania major/immunology , Animals , Antigens, Protozoan/immunology , Cytokines/biosynthesis , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Host-Parasite Interactions/immunology , Injections, Intravenous , Interleukin-12/deficiency , Interleukin-12/genetics , Intracellular Fluid/metabolism , Langerhans Cells/immunology , Langerhans Cells/parasitology , Langerhans Cells/transplantation , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Membrane Glycoproteins/immunology , Membrane Glycoproteins/therapeutic use , Metalloendopeptidases/immunology , Metalloendopeptidases/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Knockout , Peptide Initiation Factors/immunology , Peptide Initiation Factors/therapeutic use , Protozoan Proteins/immunology , Protozoan Proteins/therapeutic use , Protozoan Vaccines/immunology , Protozoan Vaccines/therapeutic use , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...