Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000308

ABSTRACT

Viral infection generally induces polyclonal neutralizing antibody responses. However, how many lineages of antibody responses can fully represent the neutralization activities in sera has not been well studied. Using the newly designed stable HIV-1 Env trimer as hook, we isolated two distinct broadly neutralizing antibodies (bnAbs) from Chinese rhesus macaques infected with SHIV1157ipd3N4 for 5 years. One lineage of neutralizing antibodies (JT15 and JT16) targeted the V2-apex in the Env trimers, similar to the J038 lineage bnAbs identified in our previous study. The other lineage neutralizing antibody (JT18) targeted the V3 crown region in the Env, which strongly competed with human 447-52D. Each lineage antibody neutralized a different set of viruses. Interestingly, when the two neutralizing antibodies from different lineages isolated from the same macaque were combined, the mixture had a neutralization breath very similar to that from the cognate sera. Our study demonstrated that a minimum of two different neutralizing antibodies can fully recapitulate the serum neutralization breadth. This observation can have important implications in AIDS vaccine design.


Subject(s)
Antibodies, Neutralizing , HIV Antibodies , HIV-1 , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome , Macaca mulatta/immunology , Animals , HIV-1/immunology , HIV Antibodies/immunology , HIV Antibodies/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Humans , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/virology , HIV Infections/immunology , HIV Infections/virology , HIV Infections/blood , Simian Immunodeficiency Virus/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Neutralization Tests
2.
Sci Rep ; 14(1): 16993, 2024 07 23.
Article in English | MEDLINE | ID: mdl-39043848

ABSTRACT

The BCG vaccine is given to millions of children globally but efficacy wanes over time and differences in the immune systems between infants and adults can influence vaccine efficacy. To this end, 34 rhesus macaques were vaccinated with BCG within seven days of birth and blood samples were collected over 88 weeks for quantification of blood cell populations. Overall, the composition of cell populations did not change significantly between BCG vaccinated and unvaccinated groups, and that BCG vaccination did not perturb normal development. In comparison to adult macaques, higher numbers of CD4+ T-cells, Tregs and NK cells were measured in the infant age group, suggesting a potential bias towards immunosuppressive and innate immune populations. Antigen-specific IFNγ secreting cell frequencies in infant BCG vaccinated animals were detectable in peripheral blood samples for 36 weeks after vaccination but declined following this. To evaluate the long-term impact of infant BCG vaccination on subsequent revaccination with BCG, a pilot study of three adult macaques received an aerosol BCG revaccination approximately 3 years after their initial BCG vaccination as infants. This induced an increase in PPD-specific IFNγ secreting cells, and increased secretion of the cytokines IFNγ and IL-1ß, following stimulation with other microorganisms, which are signals associated with trained innate immunity.


Subject(s)
Animals, Newborn , BCG Vaccine , Immunization, Secondary , Macaca mulatta , Macaca mulatta/immunology , BCG Vaccine/immunology , Animals, Newborn/immunology , Lymphocytes/immunology , Cytokines/blood , Cytokines/immunology , Animals , Male , Female
3.
J Virol ; 98(6): e0028324, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38780248

ABSTRACT

Human and simian immunodeficiency viruses (HIV and SIV) are lentiviruses that reverse transcribe their RNA genome with subsequent integration into the genome of the target cell. How progressive infection and administration of antiretrovirals (ARVs) longitudinally influence the transcriptomic and epigenetic landscape of particular T cell subsets, and how these may influence the genetic location of integration are unclear. Here, we use RNAseq and ATACseq to study the transcriptomics and epigenetic landscape of longitudinally sampled naïve and memory CD4+ and CD8+ T cells in two species of non-human primates prior to SIV infection, during chronic SIV infection, and after administration of ARVs. We find that SIV infection leads to significant alteration to the transcriptomic profile of all T cell subsets that are only partially reversed by administration of ARVs. Epigenetic changes were more apparent in animals with longer periods of untreated SIV infection and correlated well with changes in corresponding gene expression. Known SIV integration sites did not vary due to SIV status but did contain more open chromatin in rhesus macaque memory T cells, and the expression of proteasome-related genes at the pre-SIV timepoint correlated with subsequent viremia.IMPORTANCEChronic inflammation during progressive human and simian immunodeficiency virus (HIV and SIV) infections leads to significant co-morbidities in infected individuals with significant consequences. Antiretroviral (ARV)-treated individuals also manifest increased levels of inflammation which are associated with increased mortalities. These data will help guide rational development of modalities to reduce inflammation observed in people living with HIV and suggest mechanisms underlying lentiviral integration site preferences.


Subject(s)
Anti-Retroviral Agents , Epigenesis, Genetic , Memory T Cells , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Transcriptome , Animals , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epigenesis, Genetic/drug effects , Macaca mulatta/genetics , Macaca mulatta/immunology , Macaca mulatta/virology , Macaca nemestrina/genetics , Macaca nemestrina/immunology , Macaca nemestrina/virology , Memory T Cells/drug effects , Memory T Cells/immunology , Memory T Cells/metabolism , Memory T Cells/virology , Proteasome Endopeptidase Complex/genetics , RNA-Seq , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/drug effects , Transcriptome/drug effects , Viremia/drug therapy , Viremia/genetics , Viremia/immunology , Viremia/virology
4.
J Virol ; 98(2): e0165223, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38299866

ABSTRACT

CCR5-tropic simian/human immunodeficiency viruses (SHIV) with clade C transmitted/founder envelopes represent a critical tool for the investigation of HIV experimental vaccines and microbicides in nonhuman primates, although many such isolates lead to spontaneous viral control post infection. Here, we generated a high-titer stock of pathogenic SHIV-C109p5 by serial passage in two rhesus macaques (RM) and tested its virulence in aged monkeys. The co-receptor usage was confirmed before infecting five geriatric rhesus macaques (four female and one male). Plasma viral loads were monitored by reverse transcriptase-quantitative PCR (RT-qPCR), cytokines by multiplex analysis, and biomarkers of gastrointestinal damage by enzyme-linked immunosorbent assay. Antibodies and cell-mediated responses were also measured. Viral dissemination into tissues was determined by RNAscope. Intravenous SHIV-C109p5 infection of aged RMs leads to high plasma viremia and rapid disease progression; rapid decrease in CD4+ T cells, CD4+CD8+ T cells, and plasmacytoid dendritic cells; and wasting necessitating euthanasia between 3 and 12 weeks post infection. Virus-specific cellular immune responses were detected only in the two monkeys that survived 4 weeks post infection. These were Gag-specific TNFα+CD8+, MIP1ß+CD4+, Env-specific IFN-γ+CD4+, and CD107a+ T cell responses. Four out of five monkeys had elevated intestinal fatty acid binding protein levels at the viral peak, while regenerating islet-derived protein 3α showed marked increases at later time points in the three animals surviving the longest, suggesting gut antimicrobial peptide production in response to microbial translocation post infection. Plasma levels of monocyte chemoattractant protein-1, interleukin-15, and interleukin-12/23 were also elevated. Viral replication in gut and secondary lymphoid tissues was extensive.IMPORTANCESimian/human immunodeficiency viruses (SHIV) are important reagents to study prevention of virus acquisition in nonhuman primate models of HIV infection, especially those representing transmitted/founder (T/F) viruses. However, many R5-tropic SHIV have limited fitness in vivo leading to many monkeys spontaneously controlling the virus post acute infection. Here, we report the generation of a pathogenic SHIV clade C T/F stock by in vivo passage leading to sustained viral load set points, a necessity to study pathogenicity. Unexpectedly, administration of this SHIV to elderly rhesus macaques led to extensive viral replication and fast disease progression, despite maintenance of a strict R5 tropism. Such age-dependent rapid disease progression had previously been reported for simian immunodeficiency virus but not for R5-tropic SHIV infections.


Subject(s)
HIV Infections , HIV , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Virus Replication , Animals , Female , Male , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Aging , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Progression , HIV/classification , HIV/growth & development , HIV/pathogenicity , HIV/physiology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukins/immunology , Interleukins/metabolism , Intestines/virology , Lymphoid Tissue/virology , Macaca mulatta/immunology , Macaca mulatta/metabolism , Serial Passage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/pathogenicity , Simian Immunodeficiency Virus/physiology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Viral Load , Viral Tropism , Virulence , Receptors, CCR5/metabolism
5.
Nature ; 626(7998): 385-391, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38096903

ABSTRACT

A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunity, Mucosal , Immunization, Secondary , Macaca mulatta , SARS-CoV-2 , Animals , Humans , Administration, Intranasal , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/immunology , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Cytokines/immunology , Immunity, Mucosal/immunology , Immunization, Secondary/methods , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Injections, Intramuscular , Killer Cells, Natural/immunology , Lung/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , mRNA Vaccines/administration & dosage , mRNA Vaccines/immunology , SARS-CoV-2/classification , SARS-CoV-2/immunology , Trachea/immunology , Trachea/virology
6.
Int. j. lepr. other mycobact. dis ; 68(1): 27-39, Mar., 2000. graf
Article in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1226926

ABSTRACT

Groups of rhesus monkeys were vaccinated and boosted with Mycobacterium bovis bacillus Calmette Guerin (BCG) or BCG plus low-dose (LD) or high-dose (HD) heat-killed M. leprae (HKML), or were unvaccinated. Prior to and following vaccination-boosting and subsequent M. leprae (ML) challenge, these and unvaccinated, unchallenged control monkeys were observed longitudinally for approximately 3 years. Vaccination with BCG plus HKML initially stimulated significant in vitro blood mononuclear cell blastogenic responses to lepromin, which returned to baseline post-boosting and post-live-ML-challenge, minimally reappearing significantly 2 years post-ML-challenge. Vaccination with BCG failed to stimulated positive blastogenic responses to lepromin before ML-challenge but small, marginally positive, intermittent responses were seen post-ML-challenge. Compared to the unvaccinated ML-challenged group, significant increases in the numbers of blood CD4+ and CD8+ T-cell subsets and an increased CD4+:CD8+ ratio were observed in both BCG plus HKML-vaccinated, ML-challenged groups, but not in the BCG-only-vaccinated, ML-challenged group. CD4+CD29+ and CD4+CD45RA+ subset numbers increased significantly over time in only the BCG plus LD HKML-vaccinated, ML-challenged group. Compared to unvaccinated, ML-challenged groups, vaccination with BCG or BCG plus HKML followed by ML-challenge produced lower IgM:IgG antiphenolic glycolipid-I (PGL-I) serum antibody ratios and protected rhesus monkeys from clinical leprosy, consistent with prior observations that low IgM:IgG anti-PGL-I responses correlated with resistance to and protection from leprosy.


Subject(s)
Animals , Macaca mulatta/genetics , Macaca mulatta/immunology , Mycobacterium leprae/immunology , BCG Vaccine/immunology , BCG Vaccine/therapeutic use
7.
P. R. health sci. j ; 5(1): 13-8, Apr. 1986. tab
Article in English | LILACS | ID: lil-96437

ABSTRACT

Experimental autoimmune myasthenia gravis (EAMG) was induced in thesus monkeys using purified acetylcholine recptor (AChR) from Torpedo california. A single dose of 80 µg induced antibody formation two weeks after injection. Two subsequent doses at two-week intervals caused clinical signs (anorexia, fatigability, weight loss, ptosis and dysphagia) which initially responded to treatment with neostigmine. Histologic examination of post-mortem tissues revealed lesions characteristic of myasthenia gravis in man: musuclar atrophy, fibrous degeneration and lymphocytic infiltration. Antibodies were quantitated in the sera of three other monkeys which received only 60 µg of purified AChR. Abnormally high titers persisted for two years (60-200µg/ml versus 0-10µg/ml for controls). A monkey injected with 60µgAChR as part of reconstituted membrane vesicles had lower titers (30-50µg/ml) than those which received purified receptor. Only those monkeys with antibody titers exceeding 800 µg/ml developed overt disease. These titers were 4-100 times higher than those reported for myasthenic humans. The antibody-antigen molar ratios were higher for monkeys with disease than for asymptomatic animals. These data suggest that the diversity of antibody molecules synthesized by the sensitized monkeys determined the appearance of clinical signs, and that the cross reaction of tanti-torpedo antivodies with monkey receptor was primarily responsible for the development of EAMG


Subject(s)
Animals , Myasthenia Gravis/immunology , Agglutination Tests , Antibody Formation , Macaca mulatta/immunology , Receptors, Cholinergic/immunology , Torpedo/immunology
SELECTION OF CITATIONS
SEARCH DETAIL