Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38464164

ABSTRACT

The COVID-19 pandemic persists despite the availability of vaccines, and it is therefore crucial to develop new therapeutic and preventive approaches. In this study, we investigated the potential role of the oral microbiome in SARS-CoV-2 infection. Using an in vitro SARS-CoV-2 pseudovirus infection assay, we found a potent inhibitory effect exerted by Porphyromonas gingivalis on SARS-CoV-2 infection mediated by known P. gingivalis compounds such as phosphoglycerol dihydroceramide (PGDHC) and gingipains as well as by unknown bacterial factors. We found that the gingipain-mediated inhibition of infection is likely due to cytotoxicity, while PGDHC inhibited virus infection by an unknown mechanism. Unidentified factors present in P. gingivalis supernatant inhibited SARS-CoV-2 likely via the fusion step of the virus life cycle. We addressed the role of other oral bacteria and found certain periodontal pathogens capable of inhibiting SARS-CoV-2 pseudovirus infection by inducing cytotoxicity on target cells. In the human oral cavity, we observed the modulatory activity of oral microbial communities varied among individuals in that some saliva-based cultures were capable of inhibiting while others were enhancing infection. These findings contribute to our understanding of the complex relationship between the oral microbiome and viral infections, offering potential avenues for innovative therapeutic strategies in combating COVID-19.

2.
Eur J Oral Sci ; 131(1): e12906, 2023 02.
Article in English | MEDLINE | ID: mdl-36412995

ABSTRACT

SARS-CoV-2, the causative agent of the debilitating COVID-19, is mainly transmitted by first infecting nose and lung epithelial cells. The mouth is also believed to be a viral portal site since certain types of oral epithelial cells were shown to express ACE2 receptor. However, it is unclear whether oral epithelial cells are directly infected by SARS-CoV-2. In this study, we addressed whether epithelial cells of the oral gingiva were susceptible to infection. Interestingly, we found that KRT5+ and KRT18+ gingival epithelial cells do not express ACE2 but highly express TMPRSS2 and Furin as well as CD147, which was proposed to be an alternative receptor for SARS-CoV-2. However, using SARS-CoV-2 pseudoviruses containing the spike protein, we observed that gingival epithelial cells were not susceptible to infection due to the lack of ACE2 expression and the inability of CD147 to mediate viral entry. These results strongly suggest that epithelial cells from the gingiva are not susceptible to SARS-CoV-2 and CD147 is not a receptor for the SARS-CoV-2 virus. The susceptibility of oral cells from other oral structures under healthy and pathological conditions still needs to be confirmed to better understand the role of the oral cavity in COVID-19 infection and transmission.


Subject(s)
Basigin , Receptors, Coronavirus , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , Disease Susceptibility , Epithelial Cells/metabolism , Gingiva , SARS-CoV-2/metabolism , Basigin/metabolism , Receptors, Coronavirus/metabolism
3.
AIDS Res Hum Retroviruses ; 36(9): 762-770, 2020 09.
Article in English | MEDLINE | ID: mdl-32056466

ABSTRACT

A tremendous loss of financial and human resources from seven large-scale HIV vaccine efficacy trials suggest a need for a systematic approach to vaccine selection. We conducted a systematic analysis of three important envelope glycoprotein (Env) vaccine candidates: BG505 SOSIP.664, 1086.C gp140, and 1086.C gp120 to determine the most promising by comparing their structure and antigenicity. We found that the BG505 SOSIP trimer and 1086.C gp140 clearly outperformed the 1086.C gp120 monomer. BG505 SOSIP.664 bound the strongest to the most potent and broadest broadly neutralizing antibodies (bnAbs) PG9, PGT145, VRC01, and PGT121. Of interest, although BG505 SOSIP.664 did not bind to the CH58 mAb, 1086.C gp140 bound strongly to this mAb, which belongs to a class of non-neutralizing antibodies that may be protective based on correlates of protection studies of the RV144 HIV vaccine trial. The 1086.C gp120 monomer was the least antigenic of the three vaccine immunogens, binding the weakest to bnAbs and CH58 mAb. Taken together, the evidence provided here combined with previous preclinical immunogenicity and efficacy data strongly argue that the BG505 SOSIP.664 trimer and 1086.C gp140 are likely to be better vaccine immunogens than the monomeric 1086.C gp120, which was just recently tested and shown to be nonefficacious in a phase IIb/III trial. Thus, to best utilize our financial and valuable human resources, we propose a systematic approach by not only comparing structure and antigenicity, but also immunogenicity and efficacy of Env vaccine candidates in the preclinical phase to the selection of only the most promising vaccine candidates for clinical testing.


Subject(s)
AIDS Vaccines , HIV Infections , HIV-1 , Antibodies, Neutralizing , HIV Antibodies , HIV Envelope Protein gp120/genetics , HIV Infections/prevention & control , HIV-1/genetics , Humans , Protein Multimerization , env Gene Products, Human Immunodeficiency Virus/genetics
5.
PLoS One ; 10(11): e0143422, 2015.
Article in English | MEDLINE | ID: mdl-26618634

ABSTRACT

The pioneer human oral commensal bacterium Streptococcus mitis has unique biologic features that make it an attractive mucosal vaccine or therapeutic delivery vector. S. mitis is safe as a natural persistent colonizer of the mouth, throat and nasopharynx and the oral commensal bacterium is capable of inducing mucosal antibody responses. A recombinant S. mitis (rS. mitis) that stably expresses HIV envelope protein was generated and tested in the germ-free mouse model to evaluate the potential usefulness of this vector as a mucosal vaccine against HIV. Oral vaccination led to the efficient and persistent bacterial colonization of the mouth and the induction of both salivary and systemic antibody responses. Interestingly, persistently colonized animals developed antigen-specific systemic T cell tolerance. Based on these findings we propose the use of rS. mitis vaccine vector for the induction of mucosal antibodies that will prevent the penetration of the mucosa by pathogens such as HIV. Moreover, the first demonstration of rS. mitis having the ability to elicit T cell tolerance suggest the potential use of rS. mitis as an immunotherapeutic vector to treat inflammatory, allergic and autoimmune diseases.


Subject(s)
AIDS Vaccines/immunology , Genetic Vectors/genetics , Immune Tolerance , Mouth Mucosa/immunology , Streptococcus mitis/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , AIDS Vaccines/genetics , Animals , Antibodies, Viral/immunology , Female , Genetic Vectors/administration & dosage , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , Mice , Mice, Inbred BALB C , Streptococcus mitis/immunology , Vaccines, Synthetic/genetics
6.
Microbes Infect ; 17(3): 237-42, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522856

ABSTRACT

The development of vaccine approaches that induce mucosal and systemic immune responses is critical for the effective prevention of several infections. Here, we report on the use of the abundant human oral commensal bacterium Streptococcus mitis as a delivery vehicle for mucosal immunization. Using homologous recombination we generated a stable rS. mitis expressing a Mycobacterium tuberculosis protein (Ag85b). Oral administration of rS. mitis in gnotobiotic piglets resulted in efficient oral colonization and production of oral and systemic anti-Ag85b specific IgA and IgG antibodies. These results support that the commensal S. mitis is potentially a useful vector for mucosal vaccination.


Subject(s)
Bacterial Vaccines/immunology , Oral Mucosal Absorption/immunology , Streptococcus mitis/immunology , Vaccination/methods , Administration, Oral , Animals , Bacterial Proteins/genetics , Genetic Vectors/immunology , Mucous Membrane , Mycobacterium tuberculosis , Streptococcus mitis/genetics , Swine/immunology
7.
Vaccine ; 31(42): 4834-40, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23933335

ABSTRACT

Accruing data strongly support the possible role of CD8+ T cells in immunity against tuberculosis (TB). Multivalent vaccines against Mycobacterium tuberculosis (Mtb) that incorporate CD8+ T cell antigens with those that elicit CD4+ T cells are therefore highly desirable. To screen for potential CD8+ T cell antigens that are produced by Mtb during infection, we isolated pathogen-derived peptides that bound to MHC Class I molecules expressed in adherent splenocytes obtained from Mtb-infected mice. Mass spectroscopy analysis revealed the following four nonamer peptides that had 100% homology with Mtb proteins: DGYVGAPAH (MT_0401), TTMPLFAD (MT_1164), RSGAATPVR (MT_2160.1) and LAAVVGVVL (MT_0078). The gene MT_0401 codes the protein 5'-phosphoribosylglycinamide transformylase 2 and the other three genes code for hypothetical proteins with unknown function. The NCBI/Blast analysis showed that among the four peptides DGYVGAPAH had the highest maximum alignment score and lowest E value (number of alignments expected by chance). Therefore, we assessed whether MT_0401 expressed in two genetic vaccine formulations was capable of stimulating CD8+ T cell response that is specific to DGYVGAPAH peptide. When mice were immunized with a recombinant plasmid DNA and an E1/E3-deleted Adenovirus 5 expressing MT0401 protein, using both homologous and heterologous prime-boost protocols, they developed strong DGYVGAPAH-specific CD8+ T cell response as well as antibody and CD4+ specific T cell response to the full length MT0401 protein. Equally important was the observation that mice infected with Mtb developed DGYVGAPAH-specific CD8+ T cell responses in both spleen and lungs. These results demonstrate that Mtb antigens that are processed and presented via MHC Class I machinery can be readily identified by the described approach and may be useful candidate antigens to stimulate specific CD8+ T cell responses in vaccine development programs.


Subject(s)
Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Peptides/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/chemistry , Antigens, Bacterial/isolation & purification , CD4-Positive T-Lymphocytes/immunology , Female , Lung/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Peptides/chemistry , Peptides/isolation & purification , Spleen/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
8.
Article in English | MEDLINE | ID: mdl-23230563

ABSTRACT

Antibiotics and vaccines are the two most successful medical countermeasures that humans have created against a number of pathogens. However a select few e.g., Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB) have evaded eradication by vaccines and therapeutic approaches. TB is a global public health problem that kills 1.4 million people per year. The past decade has seen significant progress in developing new vaccine candidates, but the most fundamental questions in understanding disease progression and protective host responses that are responsible for controlling Mtb infection still remain poorly resolved. Current TB treatment requires intense chemotherapy with several antimicrobials, while the only approved vaccine is the classical viable whole-cell based Bacille-Calmette-Guerin (BCG) that protects children from severe forms of TB, but fails to protect adults. Taken together, there is a growing need to conduct basic and applied research to develop novel vaccine strategies against TB. This review is focused on the discussion surrounding current strategies and innovations being explored to discover new protective antigens, adjuvants, and delivery systems in the hopes of creating an efficacious TB vaccine.


Subject(s)
Drug Discovery/trends , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/isolation & purification , Tuberculosis/prevention & control , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/isolation & purification , Global Health , Humans , Tuberculosis/drug therapy , Tuberculosis/epidemiology , Tuberculosis/immunology , Vaccination/methods
9.
Immunology ; 135(3): 216-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22043824

ABSTRACT

An efficacious tuberculosis (TB) vaccine will probably need to induce both CD4 and CD8 T-cell responses specific to a protective Mycobacterium tuberculosis antigen(s). To achieve this broad cellular immune response we tested a heterologous DNA/protein combination vaccine strategy. We used a purified recombinant protein preparation of a unique M. tuberculosis antigen (rMT1721) found in the urine of TB patients, an optimized plasmid DNA expressing this protein (DNA-MT1721), and a Toll-like receptor 4 agonist adjuvant. We found that priming mice with DNA-MT1721 and subsequently boosting with rMT1721 elicited high titres of specific IgG1 and IgG2a antibodies as well as high magnitude and polyfunctional CD4(+) T-cell responses. However, no detectable CD8(+) T-cell response was observed using this regimen of immunization. In contrast, both CD4(+) and CD8(+) T-cell responses were detected after a prime/boost vaccination regimen using rMT1721 as the priming antigen and DNA-MT1721 as the boosting immunogen. These findings support the exploration of heterologous DNA/protein immunization strategies in vaccine development against TB and possibly other infectious diseases.


Subject(s)
Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA, Bacterial/administration & dosage , DNA, Bacterial/genetics , Female , Immunization, Secondary , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists , Tuberculosis Vaccines/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage
10.
J Virol ; 83(11): 5505-13, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19297477

ABSTRACT

While mycobacteria have been proposed as vaccine vectors because of their persistence and safety, little has been done systematically to optimize their immunogenicity in nonhuman primates. We successfully generated recombinant Mycobacterium bovis BCG (rBCG) expressing simian immunodeficiency virus (SIV) Gag and Pol as multigenic, nonintegrating vectors, but rBCG-expressing SIV Env was unstable. A dose and route determination study in rhesus monkeys revealed that intramuscular administration of rBCG was associated with local reactogenicity, whereas intravenous and intradermal administration of 10(6) to 10(8) CFU of rBCG was well tolerated. After single or repeat rBCG inoculations, monkeys developed high-frequency gamma interferon enzyme-linked immunospot responses against BCG purified protein derivative. However, the same animals developed only modest SIV-specific CD8(+) T-cell responses. Nevertheless, high-frequency SIV-specific cellular responses were observed in the rBCG-primed monkeys after boosting with recombinant adenovirus 5 (rAd5) expressing the SIV antigens. These cellular responses were of greater magnitude and more persistent than those generated after vaccination with rAd5 alone. The vaccine-elicited cellular responses were predominantly polyfunctional CD8(+) T cells. These findings support the further exploration of mycobacteria as priming vaccine vectors.


Subject(s)
Adenoviridae/genetics , BCG Vaccine/immunology , Immunization, Secondary/methods , Macaca mulatta/immunology , Mycobacterium bovis/immunology , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/immunology , Animals , BCG Vaccine/genetics , BCG Vaccine/metabolism , Cells, Cultured , Gene Products, env/genetics , Gene Products, env/immunology , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/immunology , Gene Products, gag/metabolism , Gene Products, pol/genetics , Gene Products, pol/immunology , Gene Products, pol/metabolism , Genetic Vectors/genetics , Mycobacterium bovis/genetics , Mycobacterium bovis/metabolism , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/metabolism , Titrimetry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism
11.
J Immunol ; 179(10): 6725-33, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982062

ABSTRACT

The duration of Ag expression in vivo has been reported to have a minimal impact on both the magnitude and kinetics of contraction of a pathogen-induced CD8(+) T cell response. In this study, we controlled the duration of Ag expression by excising the ear pinnae following intradermal ear pinnae DNA immunization. This resulted in decreased magnitude, accelerated contraction and differentiation, and surprisingly greater secondary CD8(+) T cell responses. Furthermore, we found delayed and prolonged Ag presentation in the immunized mice; however, this presentation was considerably decreased when the depot Ag was eliminated. These findings suggest that the magnitude and the contraction phase of the CD8(+) T cell response following intradermal DNA immunization is regulated by the duration rather than the initial exposure to Ag.


Subject(s)
Antigen Presentation , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Antigen Presentation/genetics , Antigens/biosynthesis , Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Immunization , Injections, Intradermal , Mice , Mice, Inbred BALB C , Time Factors , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
12.
J Virol ; 81(23): 12793-802, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881444

ABSTRACT

While recent studies have demonstrated that secondary CD8+ T cells develop into effector-memory cells, the impact of particular vaccine regimens on the elicitation of these cells remains poorly defined. In the present study we evaluated the effect of three different immunogens--recombinant vaccinia, recombinant adenovirus, and plasmid DNA--on the generation of memory cellular immune responses. We found that vectors that induce the rapid movement of CD8+ T cells into the memory compartment during a primary immune response also drive a rapid differentiation of these cells into effector-memory CD8+ T cells following a secondary immunization. In contrast, the functional profiles of both CD8+ and CD4+ T cells, assessed by measuring antigen-stimulated gamma interferon and interleukin-2 production, were not predominantly shaped by the boosting immunogen. We also demonstrated that the in vivo expression of antigen by recombinant vectors was brief following boosting immunization, suggesting that antigen persistence has a minimal impact on the differentiation of secondary CD8+ T cells. When used in heterologous or in homologous prime-boost combinations, these three vectors generated antigen-specific CD8+ T cells with different phenotypic profiles. Expression of the memory-associated molecule CD27 on effector CD8+ T cells decreased following heterologous but not homologous boosting, resulting in a phenotypic profile similar to that seen on primary CD8+ T cells. These data therefore suggest that the phenotype of secondary CD8+ T cells is determined predominantly by the boosting immunogen whereas the cytokine profile of these cells is shaped by both the priming and boosting immunogens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization, Secondary , Immunologic Memory , T-Lymphocyte Subsets/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred BALB C , Plasmids/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/biosynthesis , Vaccines, DNA/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
13.
J Virol ; 81(19): 10606-13, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652387

ABSTRACT

While administration of granulocyte-macrophage colony-stimulating factor (GM-CSF) can induce the local recruitment of activated antigen-presenting cells at the site of vaccine inoculation, this cellular recruitment is associated with a paradoxical decrease in local vaccine antigen expression and vaccine-elicited CD8+ T-cell responses. To clarify why this cytokine administration does not potentiate immunization, we examined the recruited cells and expressed inflammatory mediators in muscles following intramuscular administration of plasmid GM-CSF in mice. While large numbers of dendritic cells and macrophages were attracted to the site of plasmid GM-CSF inoculation, high concentrations of type I interferons were also detected in the muscles. As type I interferons have been reported to damp foreign gene expression in vivo, we examined the possibility that these local innate mediators might decrease plasmid DNA expression and therefore the immunogenicity of plasmid DNA vaccines. In fact, we found that coadministration of an anti-beta interferon monoclonal antibody with the plasmid DNA immunogen and plasmid GM-CSF restored both the local antigen expression and the CD8+ T-cell immunogenicity of the vaccine. These data demonstrate that local innate immune responses can change the ability of vaccines to generate robust adaptive immunity.


Subject(s)
Antigen Presentation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Interferon-beta/metabolism , Vaccines, DNA/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunity, Innate/drug effects , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Interferon-beta/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Muscle, Skeletal/immunology , Plasmids/administration & dosage , Plasmids/genetics , Th1 Cells/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
14.
J Virol ; 81(1): 74-83, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050608

ABSTRACT

The most promising vaccine strategies for the induction of cytotoxic-T-lymphocyte responses have been heterologous prime/boost regimens employing a plasmid DNA prime and a live recombinant-vector boost. The priming immunogen in these regimens must elicit antigen-specific memory CD8+ T lymphocytes that will expand following the boosting immunization. Because plasmid DNA immunogens are expensive and their immunogenicity has proven disappointing in human clinical trials, we have been exploring novel priming immunogens that might be used in heterologous immunization regimens. Here we show that priming with a prototype recombinant Mycobacterium smegmatis strain expressing human immunodeficiency virus type 1 (HIV-1) gp120-elicited CD4+ T lymphocytes with a functional profile of helper cells as well as a CD8+ T-lymphocyte population. These CD8+ T lymphocytes rapidly differentiated to memory cells, defined on the basis of their cytokine profile and expression of CD62L and CD27. Moreover, these recombinant-mycobacterium-induced T lymphocytes rapidly expanded following boosting with a recombinant adenovirus expressing HIV-1 Env to gp120-specific CD8+ T lymphocytes. This work demonstrates a remarkable skewing of recombinant-mycobacterium-induced T lymphocytes to durable antigen-specific memory CD8+ T cells and suggests that such immunogens might be used as priming vectors in prime/boost vaccination regimens for the induction of cellular immune responses.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Mycobacterium smegmatis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Gene Products, gag/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Immunization, Secondary , Kinetics , Mice , Mice, Inbred BALB C , Mycobacterium smegmatis/genetics , Plasmids/immunology , gag Gene Products, Human Immunodeficiency Virus
15.
Clin Vaccine Immunol ; 13(11): 1204-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16943347

ABSTRACT

A successful vaccine vector for human immunodeficiency virus type 1 (HIV-1) should induce anti-HIV-1 immune responses at mucosal sites. We have generated recombinant Mycobacterium smegmatis vectors that express the HIV-1 group M consensus envelope protein (Env) as a surface, intracellular, or secreted protein and have tested them in animals for induction of both anti-HIV-1 T-cell and antibody responses. Recombinant M. smegmatis engineered for expression of secreted protein induced optimal T-cell gamma interferon enzyme-linked immunospot assay responses to HIV-1 envelope in the spleen, female reproductive tract, and lungs. Unlike with the induction of T-cell responses, priming and boosting with recombinant M. smegmatis did not induce anti-HIV-1 envelope antibody responses, due primarily to insufficient protein expression of the insert. However, immunization with recombinant M. smegmatis expressing HIV-1 Env was able to prime for an HIV-1 Env protein boost for the induction of anti-HIV-1 antibody responses.


Subject(s)
HIV-1/immunology , Immunity, Mucosal , Mycobacterium smegmatis/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Animals , Cloning, Molecular , Epitopes, T-Lymphocyte/immunology , Female , Gene Products, env/biosynthesis , Gene Products, env/genetics , Gene Products, env/physiology , Genetic Vectors , HIV Envelope Protein gp120/biosynthesis , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/physiology , HIV-1/genetics , Humans , Mice , Mice, Inbred BALB C , Mycobacterium smegmatis/metabolism , T-Lymphocytes/immunology , Transformation, Genetic , env Gene Products, Human Immunodeficiency Virus
16.
J Virol ; 80(4): 1645-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16439521

ABSTRACT

Because the vaccine vectors currently being evaluated in human populations all have significant limitations in their immunogenicity, novel vaccine strategies are needed for the elicitation of cell-mediated immunity. The nonpathogenic, rapidly growing mycobacterium Mycobacterium smegmatis was engineered as a vector expressing full-length human immunodeficiency virus type 1 (HIV-1) HXBc2 envelope protein. Immunization of mice with recombinant M. smegmatis led to the expansion of major histocompatibility complex class I-restricted HIV-1 epitope-specific CD8(+) T cells that were cytolytic and secreted gamma interferon. Effector and memory T lymphocytes were elicited, and repeated immunization generated a stable central memory pool of virus-specific cells. Importantly, preexisting immunity to Mycobacterium bovis BCG had only a marginal effect on the immunogenicity of recombinant M. smegmatis. This mycobacterium may therefore be a useful vaccine vector.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, env , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Mycobacterium smegmatis/genetics , Animals , Bacterial Vaccines/immunology , Cytotoxicity Tests, Immunologic , Female , Immunization , Immunologic Memory , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mycobacterium bovis , Mycobacterium smegmatis/immunology , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...