Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 106(7): 2301-6, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19188599

RESUMO

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.


Assuntos
Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Adjuvantes Imunológicos/química , Animais , Citocinas/metabolismo , Progressão da Doença , Haplorrinos , Sistema Imunitário , Interferon gama/metabolismo , Interleucina-6/metabolismo , Macaca fascicularis , Mycobacterium tuberculosis/metabolismo , Fatores de Tempo , Resultado do Tratamento , Tuberculose/microbiologia , Vacinas contra a Tuberculose/química
2.
Immunology ; 129(4): 496-505, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20002212

RESUMO

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.


Assuntos
Alelos , Linfócitos T CD8-Positivos/imunologia , Antígenos HLA-B/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Mycobacterium tuberculosis/imunologia , Peptídeos/imunologia , Sítios de Ligação , Antígeno HLA-B7 , Antígeno HLA-B8 , Humanos , Tuberculose Pulmonar/diagnóstico , Tuberculose Pulmonar/imunologia
3.
Immunology ; 131(1): 128-40, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20465573

RESUMO

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.


Assuntos
Imunidade Celular , Macaca mulatta/imunologia , Subpopulações de Linfócitos T/imunologia , Adulto , Animais , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Humanos , Interleucina-7/imunologia , Fosforilação , Fator de Transcrição STAT5/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
4.
Nat Rev Microbiol ; 4(6): 469-76, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16710326

RESUMO

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.


Assuntos
Vacinas contra a Tuberculose/imunologia , Vacinas contra a Tuberculose/normas , Tuberculose/prevenção & controle , Vacinação , Vacina BCG/imunologia , Humanos , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Tempo , Tuberculose/imunologia , Tuberculose/microbiologia , Vacinação/normas , Vacinação/tendências , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/imunologia
5.
Methods Mol Med ; 94: 91-106, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14959824

RESUMO

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.


Assuntos
Clonagem Molecular/métodos , Expressão Gênica , Proteínas/genética , Proteínas/imunologia , Animais , Anticorpos , Anticorpos Antibacterianos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Biblioteca Genômica , Humanos , Técnicas de Imunoadsorção , Biblioteca de Peptídeos , Testes Sorológicos
6.
Vaccine ; 28(4): 1084-93, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19896449

RESUMO

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.


Assuntos
Imunização Secundária/métodos , Oligodesoxirribonucleotídeos/efeitos adversos , Oligopeptídeos/efeitos adversos , Vacinas contra a Tuberculose/efeitos adversos , Vacinas contra a Tuberculose/imunologia , Tuberculose Pulmonar/prevenção & controle , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Combinação de Medicamentos , Feminino , Cobaias , Pulmão/imunologia , Pulmão/patologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligopeptídeos/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Análise de Sobrevida , Vacinas contra a Tuberculose/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
7.
Vaccine ; 27(2): 223-33, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19000730

RESUMO

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.


Assuntos
Antígenos de Bactérias/genética , Linfócitos T CD4-Positivos/metabolismo , Clonagem Molecular/métodos , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Linhagem Celular , Mapeamento de Epitopos , Escherichia coli/genética , Escherichia coli/metabolismo , Cobaias , Humanos , Ativação Linfocitária , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Tuberculose/imunologia , Tuberculose/microbiologia , Tuberculose/prevenção & controle , Vacinas contra a Tuberculose/administração & dosagem , Vacinas contra a Tuberculose/genética
8.
Vaccine ; 27(33): 4412-23, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19500523

RESUMO

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.


Assuntos
Antígenos de Bactérias/imunologia , Vacina BCG/imunologia , Toxinas Bacterianas/imunologia , Proteínas Hemolisinas/imunologia , Epitopos Imunodominantes/imunologia , Tuberculose/prevenção & controle , Animais , Vacina BCG/administração & dosagem , Vacina BCG/genética , Toxinas Bacterianas/genética , Linhagem Celular , Eritrócitos/microbiologia , Feminino , Genes Bacterianos , Cobaias , Proteínas Hemolisinas/genética , Hemólise , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Mycobacterium bovis/genética , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/imunologia , Plasmídeos , Ovinos , Tuberculose/imunologia
9.
PLoS One ; 3(11): e3790, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19023426

RESUMO

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.


Assuntos
Vacina BCG/administração & dosagem , Linfócitos T/imunologia , Adenoviridae/genética , Animais , Antígenos de Bactérias/genética , Vacina BCG/genética , Toxinas Bacterianas/genética , Feminino , Vetores Genéticos , Proteínas Hemolisinas/genética , Imunidade Celular , Imunização Secundária , Interferon gama/biossíntese , Ativação Linfocitária , Macaca mulatta , Vacinas contra a Tuberculose/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética
10.
Infect Immun ; 75(8): 4105-15, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17526747

RESUMO

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.


Assuntos
Epitopos de Linfócito T/imunologia , Interferon gama/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Vacinas Virais/imunologia , Aciltransferases/genética , Aciltransferases/imunologia , Adenoviridae/genética , Administração Intranasal , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Vetores Genéticos , Injeções Intramusculares , Fígado/microbiologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Baço/microbiologia , Vacinas contra a Tuberculose/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Virais/genética
11.
Microbiology (Reading) ; 152(Pt 12): 3667-3681, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17159220

RESUMO

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.


Assuntos
Acne Vulgar/imunologia , Antígenos de Bactérias/biossíntese , Antígenos de Bactérias/imunologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/imunologia , Propionibacterium acnes/imunologia , Propionibacterium acnes/metabolismo , Sequência de Aminoácidos , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/química , Antígenos de Bactérias/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Transporte/genética , Clonagem Molecular , Sequência Conservada , Corynebacterium diphtheriae/genética , Dermatan Sulfato/metabolismo , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito T/imunologia , Mutação da Fase de Leitura , Expressão Gênica , Variação Genética , Humanos , Immunoblotting , Imunoglobulina G/imunologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Dados de Sequência Molecular , Propionibacterium acnes/genética , Ligação Proteica , Sinais Direcionadores de Proteínas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Homologia de Sequência de Aminoácidos , Streptococcus equi/genética
12.
Infect Immun ; 74(4): 2392-401, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16552069

RESUMO

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.


Assuntos
Proteínas de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose Meníngea/imunologia , Tuberculose Meníngea/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Proteínas de Bactérias/administração & dosagem , Peso Corporal/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Combinação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Imunidade Ativa , Imunização Secundária , Lipídeo A/administração & dosagem , Lipídeo A/análogos & derivados , Lipídeo A/imunologia , Pulmão/imunologia , Pulmão/patologia , Mycobacterium bovis/imunologia , Coelhos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Saponinas/administração & dosagem , Saponinas/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Tuberculose Meníngea/patologia
13.
Vaccine ; 23(30): 3937-45, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15917115

RESUMO

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).


Assuntos
Antígenos de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Adjuvantes Imunológicos , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Contagem de Colônia Microbiana , Citocinas/análise , Citocinas/imunologia , Epitopos/imunologia , Cobaias , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Baço/citologia , Tuberculose/imunologia , Tuberculose/microbiologia , Vacinas Sintéticas/imunologia
14.
J Immunol ; 170(6): 3171-9, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12626575

RESUMO

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.


Assuntos
Antígenos de Protozoários/uso terapêutico , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Interleucina-12/fisiologia , Líquido Intracelular/imunologia , Líquido Intracelular/parasitologia , Leishmania major/imunologia , Animais , Antígenos de Protozoários/imunologia , Citocinas/biossíntese , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Feminino , Interações Hospedeiro-Parasita/imunologia , Injeções Intravenosas , Interleucina-12/deficiência , Interleucina-12/genética , Líquido Intracelular/metabolismo , Células de Langerhans/imunologia , Células de Langerhans/parasitologia , Células de Langerhans/transplante , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/prevenção & controle , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/uso terapêutico , Metaloendopeptidases/imunologia , Metaloendopeptidases/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fatores de Iniciação de Peptídeos/imunologia , Fatores de Iniciação de Peptídeos/uso terapêutico , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/uso terapêutico , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/uso terapêutico , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
15.
J Infect Dis ; 187(3): 513-7, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12552438

RESUMO

The ability of 17 recombinant mycobacterial proteins, native antigen 85 complex, lipoarabinomannan (LAM), and Mycobacterium tuberculosis lysate to detect antibody responses induced by bacille Calmette-Guérin (BCG) vaccination and active tuberculosis infection were studied in enzyme-linked immunosorbent assays. Only LAM-reactive serum immunoglobulin G responses were significantly increased in both BCG-vaccinated patients and patients with active tuberculosis (P<.05), and oral BCG vaccination also induced significant increases in LAM-reactive secretory immunoglobulin A (P<.05). LAM-reactive antibody assays can serve as markers of humoral and mucosal immunity in future trials of BCG and newer attenuated mycobacterial vaccines.


Assuntos
Vacina BCG/imunologia , Imunoglobulina A/imunologia , Lipopolissacarídeos/imunologia , Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Vacinação
16.
Infect Immun ; 71(9): 5065-76, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12933849

RESUMO

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.


Assuntos
Genes de Protozoários , Imunoglobulinas/metabolismo , Proteínas de Protozoários/genética , Receptores de Superfície Celular/genética , Trypanosomatina/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Clonagem Molecular , DNA Complementar/genética , DNA de Protozoário/genética , Humanos , Técnicas In Vitro , Leishmania major/genética , Leishmania major/imunologia , Leishmania major/metabolismo , Camundongos , Dados de Sequência Molecular , Peso Molecular , Mapeamento de Peptídeos , Ligação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Coelhos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/química , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Homologia de Sequência de Aminoácidos , Trypanosoma/genética , Trypanosoma/imunologia , Trypanosoma/metabolismo , Trypanosomatina/imunologia , Trypanosomatina/metabolismo
17.
Protein Expr Purif ; 30(1): 124-33, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12821330

RESUMO

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.


Assuntos
Biotecnologia/métodos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Sequência de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Sequência de Bases , Linhagem Celular , Escherichia coli , Citometria de Fluxo , Expressão Gênica , Engenharia Genética/métodos , Vetores Genéticos , Humanos , Imuno-Histoquímica , Dados de Sequência Molecular , Mycobacterium tuberculosis , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação
18.
Vaccine ; 20(27-28): 3292-303, 2002 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12213399

RESUMO

Three immunodominant leishmanial antigens (TSA, LmSTI1 and LeIF) previously identified in the context of host response to infection in infected donors and BALB/c mice, as well as their ability to elicit at least partial protection against Leishmania major infection in the BALB/c mouse model, were selected for inclusion into a subunit based vaccine. This is based on the premise that an effective vaccine against leishmaniasis (a complex parasitic infection) would require a multivalent cocktail of several antigens containing a broader range of protective epitopes that would cover a wide range of MHC types in a heterogeneous population. For practical considerations of vaccine development, we report on the generation of a single recombinant polyprotein comprising the sequences of all three open reading frames genetically linked in tandem. The resulting molecule, Leish-111f, comprises an open reading frame that codes for a 111kDa polypeptide. Evaluation of the immunogenicity and protective efficacy of Leish-111f formulated with IL-12 revealed that the immune responses to the individual components were maintained and as well, rLeish-111f protected BALB/c mice against L. major infection to a magnitude equal or superior to those seen with any of the individual components of the vaccine construct or SLA, a soluble Leishmania lysate. But because rIL-12 is expensive and difficult to manufacture and its efficacy and safety as an adjuvant for human use is questionable, we screened for other adjuvants that could potentially substitute for IL-12. We report that monophosphoryl lipid A (MPL) plus squalene (MPL-SE) formulated with rLeish-111f elicited protective immunity against L. major infection. The demonstrated feasibility to manufacture a single recombinant vaccine comprising multiple protective open reading frames and the potential use of MPL-SE as a substitute for IL-12, takes us closer to the realization of an affordable and safe Leishmania vaccine.


Assuntos
Leishmania major/imunologia , Leishmaniose Cutânea/prevenção & controle , Lipídeo A/análogos & derivados , Vacinas Protozoárias/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/sangue , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , DNA de Protozoário/genética , Feminino , Humanos , Leishmania major/genética , Leishmaniose Cutânea/imunologia , Lipídeo A/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fases de Leitura Aberta , Vacinas Protozoárias/genética , Esqualeno/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética
19.
Infect Immun ; 70(8): 4215-25, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12117930

RESUMO

Development of an effective vaccine against Leishmania infection is a priority of tropical disease research. We have recently demonstrated protection against Leishmania major in the murine and nonhuman primate models with individual or combinations of purified leishmanial recombinant antigens delivered as plasmid DNA constructs or formulated with recombinant interleukin-12 (IL-12) as adjuvant. In the present study, we immunized BALB/c mice with a recombinant polyprotein comprising a tandem fusion of the leishmanial antigens thiol-specific antioxidant, L. major stress-inducible protein 1 (LmSTI1), and Leishmania elongation initiation factor (LeIF) delivered with adjuvants suitable for human use. Aspects of the safety, immunogenicity, and vaccine efficacy of formulations with each individual component, as well as the polyprotein referred to as Leish-111f, were assessed by using the L. major challenge model with BALB/c mice. No adverse reactions were observed when three subcutaneous injections of the Leish-111f polyprotein formulated with either MPL-squalene (SE) or Ribi 529-SE were given to BALB/c mice. A predominant Th1 immune response characterized by in vitro lymphocyte proliferation, gamma interferon production, and immunoglobulin G2A antibodies was observed with little, if any, IL-4. Moreover, Leish-111f formulated with MPL-SE conferred immunity to leishmaniasis for at least 3 months. These data demonstrate success at designing and developing a prophylactic leishmaniasis vaccine that proved effective in a preclinical model using multiple leishmanial antigens produced as a single protein delivered with a powerful Th1 adjuvant suitable for human use.


Assuntos
Antígenos de Protozoários/imunologia , Antioxidantes , Proteínas de Choque Térmico/imunologia , Leishmania major/imunologia , Leishmania mexicana/imunologia , Leishmaniose Cutânea/prevenção & controle , Lipídeo A/análogos & derivados , Chaperonas Moleculares/imunologia , Fatores de Iniciação de Peptídeos/imunologia , Peroxidases/imunologia , Poliproteínas/imunologia , Proteínas de Protozoários , Vacinas Protozoárias/imunologia , Vacinas Sintéticas/imunologia , Adjuvantes Imunológicos , Animais , Antígenos de Protozoários/genética , Esqueleto da Parede Celular , Fatores Corda , Modelos Animais de Doenças , Feminino , Proteínas de Choque Térmico/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Chaperonas Moleculares/genética , Fatores de Iniciação de Peptídeos/genética , Peroxidases/genética , Peroxirredoxinas , Poliproteínas/genética , Vacinas Protozoárias/genética , Linfócitos T/imunologia , Vacinação , Vacinas Sintéticas/genética
20.
Infect Immun ; 72(11): 6622-32, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15501795

RESUMO

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.


Assuntos
Vacina BCG/imunologia , Proteínas de Bactérias/imunologia , Imunização Secundária , Mycobacterium tuberculosis/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose Pulmonar/prevenção & controle , Animais , Vacina BCG/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Cobaias , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Poliproteínas/administração & dosagem , Poliproteínas/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Tuberculose Pulmonar/mortalidade , Tuberculose Pulmonar/patologia , Vacinação , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA