Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
1.
Front Immunol ; 15: 1386243, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835757

RÉSUMÉ

Introduction: Current vaccines against COVID-19 administered via parenteral route have limited ability to induce mucosal immunity. There is a need for an effective mucosal vaccine to combat SARS-CoV-2 virus replication in the respiratory mucosa. Moreover, sex differences are known to affect systemic antibody responses against vaccines. However, their role in mucosal cellular responses against a vaccine remains unclear and is underappreciated. Methods: We evaluated the mucosal immunogenicity of a booster vaccine regimen that is recombinant protein-based and administered intranasally in mice to explore sex differences in mucosal humoral and cellular responses. Results: Our results showed that vaccinated mice elicited strong systemic antibody (Ab), nasal, and bronchiole alveolar lavage (BAL) IgA responses, and local T cell immune responses in the lung in a sex-biased manner irrespective of mouse genetic background. Monocytes, alveolar macrophages, and CD103+ resident dendritic cells (DCs) in the lungs are correlated with robust mucosal Ab and T cell responses induced by the mucosal vaccine. Discussion: Our findings provide novel insights into optimizing next-generation booster vaccines against SARS-CoV-2 by inducing spike-specific lung T cell responses, as well as optimizing mucosal immunity for other respiratory infections, and a rationale for considering sex differences in future vaccine research and vaccination practice.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Immunité muqueuse , Immunogénicité des vaccins , SARS-CoV-2 , Vaccins sous-unitaires , Animaux , Femelle , Souris , SARS-CoV-2/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/virologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/administration et posologie , Mâle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Poumon/immunologie , Poumon/virologie , Lymphocytes T/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Souris de lignée C57BL , Administration par voie nasale , Facteurs sexuels , Immunoglobuline A/immunologie , Cellules dendritiques/immunologie , Rappel de vaccin , Immunité humorale
2.
bioRxiv ; 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38798555

RÉSUMÉ

Most COVID-19 vaccine trials have focused on recipient protection, not protection of their contacts, a critical need. As a subunit intranasal COVID-19 vaccine reduced nasopharyngeal virus more than did an intramuscular (IM) vaccine, we hypothesized that this vaccine might reduce onward transmission to others. We vaccinated hamsters with either the IM-administrated Moderna mRNA vaccine twice or one dose of mRNA IM followed by adjuvanted subunit intranasal vaccine. 24 hours after SARS-CoV-2 challenge, these animals were housed with naïve recipients in a contactless chamber that allows airborne transmission. Onward airborne transmission was profoundly blocked: the donor and recipients of the intranasal vaccine-boosted group had lower oral and lung viral loads (VL), which correlated with mucosal ACE2 inhibition activity. These data strongly support the use of the intranasal vaccine as a boost to protect not only the vaccinated person, but also people exposed to the vaccinated person, a key public health goal. Author summary: Natural transmission of SARS-CoV-2 is primarily airborne, through the respiratory mucosal route. However, current licensed COVID-19 vaccines are all intramuscular and induce more systemic than mucosal immunity. Here, we did a head-to-head comparison of COVID-19 booster vaccines on SARS-CoV-2 onward transmission. We found that compared to boosting with a Moderna mRNA systemic vaccine, a nanoparticle intranasal COVID-19 vaccine much more effectively prevents onward airborne transmission to naïve recipient hamsters. The protection was correlated with local mucosal antibody. Thus, a mucosal nanoparticle vaccine should be considered for preventing onward airborne transmission, a key public health necessity that has not been adequately studied.

3.
Vaccine ; 42(2): 339-351, 2024 Jan 12.
Article de Anglais | MEDLINE | ID: mdl-38071106

RÉSUMÉ

Intranasal mucosal vaccines can more effectively induce mucosal immune responses against SARS-CoV-2. Here, we show in hamsters that an intranasal subunit mucosal vaccine boost with the beta variant S1 can prevent weight loss, in addition to reducing viral load, which cannot be studied in macaques that don't develop COVID-like disease. Protective efficacy against both viral load and weight loss correlated with serum antibody titers. A sex bias was detected in that immune responses and protection against viral load were greater in females than males. We also found that priming with S1 from the Wuhan strain elicited lower humoral immune responses against beta variant and led to less protection against beta viral challenge, suggesting the importance of matched antigens. The greater efficacy of mucosal vaccines in the upper respiratory tract and the need to consider sex differences in vaccine protection are important in the development of future improved COVID-19 vaccines.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Femelle , Mâle , Animaux , Cricetinae , Humains , Sexisme , SARS-CoV-2 , COVID-19/prévention et contrôle , Macaca , Perte de poids , Anticorps antiviraux , Anticorps neutralisants , Glycoprotéine de spicule des coronavirus
4.
Front Immunol ; 14: 1154496, 2023.
Article de Anglais | MEDLINE | ID: mdl-37020550

RÉSUMÉ

Introduction: Adjuvant plays an important role in directing the immune responses induced by vaccines. In previous studies, we have shown that a mucosal SARS-CoV-2 S1 subunit vaccine adjuvanted with a combination of CpG, Poly I:C and IL-15 (named CP15) induced effective mucosal and systemic immunity and conferred nearly sterile protection against SARS-CoV-2 viral replication in macaque models. Methods: In this study, we used a hamster model, which mimics the human scenario and reliably exhibits severe SARS-CoV-2 disease similar to hospitalized patients, to investigate the protection efficacy of the vaccines against COVID-19 disease. We compared the weight loss, viral loads (VLs), and clinical observation scores of three different vaccine regimens. All three regimens consisted of priming/boosting with S1 subunit vaccines, but adjuvanted with alum and/or CP15 administrated by either intramuscular (IM) or intranasal (IN) routes: Group 1 was adjuvanted with alum/alum administrated IM/IM; Group 2 was alum-IM/CP15-IN; and Group 3 was CP15-IM/CP15-IN. Results: After challenge with SARS-CoV-2 WA strain, we found that the alum/CP15 group showed best protection against weight loss, while the CP15 group demonstrated best reduction of oral SARS-CoV-2 VLs, suggesting that the protection profiles were different. Sex differences for VL and clinical scores were observed. Humoral immunity was induced but not correlated with protection. Moreover, S1-specific binding antibody titers against beta, omicron BA.1, and BA.2 variants showed 2.6-, 4.9- and 2.8- fold reduction, respectively, compared to the Wuhan strain. Discussion: Overall, the data suggested that adjuvants in subunit vaccines determine the protection profiles after SARS-CoV-2 infection and that nasal/oral mucosal immunization can protect against systemic COVID-19 disease.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Mâle , Cricetinae , Animaux , Humains , Femelle , SARS-CoV-2 , Adjuvants immunologiques , Adjuvants pharmaceutiques , Vaccins sous-unitaires
5.
PNAS Nexus ; 1(3): pgac091, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35873792

RÉSUMÉ

Emergence of SARS-CoV-2 variants and waning of vaccine/infection-induced immunity pose threats to curbing the COVID-19 pandemic. Effective, safe, and convenient booster vaccines are in need. We hypothesized that a variant-modified mucosal booster vaccine might induce local immunity to prevent SARS-CoV-2 infection at the port of entry. The beta-variant is one of the hardest to cross-neutralize. Herein, we assessed the protective efficacy of an intranasal booster composed of beta variant-spike protein S1 with IL-15 and TLR agonists in previously immunized macaques. The macaques were first vaccinated with Wuhan strain S1 with the same adjuvant. A total of 1 year later, negligibly detectable SARS-CoV-2-specific antibody remained. Nevertheless, the booster induced vigorous humoral immunity including serum- and bronchoalveolar lavage (BAL)-IgG, secretory nasal- and BAL-IgA, and neutralizing antibody against the original strain and/or beta variant. Beta-variant S1-specific CD4+ and CD8+ T cell responses were also elicited in PBMC and BAL. Following SARS-CoV-2 beta variant challenge, the vaccinated group demonstrated significant protection against viral replication in the upper and lower respiratory tracts, with almost full protection in the nasal cavity. The fact that one intranasal beta-variant booster administrated 1 year after the first vaccination provoked protective immunity against beta variant infections may inform future SARS-CoV-2 booster design and administration timing.

6.
Front Immunol ; 12: 737406, 2021.
Article de Anglais | MEDLINE | ID: mdl-34603318

RÉSUMÉ

IL-7/IL-7R signaling is critical for development, maturation, maintenance and survival of many lymphocytes in the thymus and periphery. IL-7 has been used as immunotherapy in pre-clinical and clinical studies to treat cancer, HIV infection and sepsis. Here, we discuss the critical function of IL-7 in diagnosis, prognosis and treatment of COVID-19 patients. We also summarize a promising role of IL-7 as a vaccine adjuvant. It could potentially enhance the immune responses to vaccines especially against SARS-CoV-2 or other new vaccines.


Sujet(s)
Adjuvants immunologiques , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Interleukine-7/immunologie , SARS-CoV-2/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , COVID-19/immunologie , Humains , Immunogénicité des vaccins/immunologie , Interleukine-7/métabolisme , Récepteurs à l'interleukine-7/métabolisme
7.
Front Immunol ; 12: 658428, 2021.
Article de Anglais | MEDLINE | ID: mdl-34149696

RÉSUMÉ

SARS-CoV-2 virus causes upper and lower respiratory diseases including pneumonia, and in some cases, leads to lethal pulmonary failure. Angiotensin converting enzyme-2 (ACE2), the receptor for cellular entry of SARS-CoV-2 virus, has been shown to protect against severe acute lung failure. Here, we provide evidence that SARS-CoV-2 spike protein S1 reduced the mRNA expression of ACE2 and type I interferons in primary cells of lung bronchoalveolar lavage (BAL) from naïve rhesus macaques. The expression levels of ACE2 and type I interferons were also found to be correlated with each other, consistent with the recent finding that ACE2 is an interferon-inducible gene. Furthermore, induction of ACE2 and type I interferons by poly I:C, an interferon inducer, was suppressed by S1 protein in primary cells of BAL. These observations suggest that the downregulation of ACE2 and type I interferons induced by S1 protein may directly contribute to SARS-CoV-2-associated lung diseases.


Sujet(s)
Angiotensin-converting enzyme 2/métabolisme , COVID-19 , Interféron de type I/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Animaux , Liquide de lavage bronchoalvéolaire/cytologie , Macaca mulatta , SARS-CoV-2
8.
JCI Insight ; 6(10)2021 04 28.
Article de Anglais | MEDLINE | ID: mdl-33908897

RÉSUMÉ

Effective SARS-CoV-2 vaccines are urgently needed. Although most vaccine strategies have focused on systemic immunization, here we compared the protective efficacy of 2 adjuvanted subunit vaccines with spike protein S1: an intramuscularly primed/boosted vaccine and an intramuscularly primed/intranasally boosted mucosal vaccine in rhesus macaques. The intramuscular-alum-only vaccine induced robust binding and neutralizing antibody and persistent cellular immunity systemically and mucosally, whereas intranasal boosting with nanoparticles, including IL-15 and TLR agonists, elicited weaker T cell and Ab responses but higher dimeric IgA and IFN-α. Nevertheless, following SARS-CoV-2 challenge, neither group showed detectable subgenomic RNA in upper or lower respiratory tracts versus naive controls, indicating full protection against viral replication. Although mucosal and systemic protective mechanisms may differ, results demonstrate both vaccines can protect against respiratory SARS-CoV-2 exposure. In summary, we have demonstrated that the mucosal vaccine was safe after multiple doses and cleared the input virus more efficiently in the nasal cavity and thus may act as a potent complementary reinforcing boost for conventional systemic vaccines to provide overall better protection.


Sujet(s)
Vaccins contre la COVID-19/usage thérapeutique , COVID-19/médecine vétérinaire , Macaca mulatta/immunologie , SARS-CoV-2/immunologie , Immunité acquise , Animaux , Anticorps neutralisants/immunologie , COVID-19/immunologie , COVID-19/anatomopathologie , COVID-19/prévention et contrôle , Humains , Immunité cellulaire , Immunité humorale , Vaccins sous-unitaires/usage thérapeutique
9.
Front Immunol ; 12: 638872, 2021.
Article de Anglais | MEDLINE | ID: mdl-33732259

RÉSUMÉ

CXCL13 signals through the G protein-coupled chemokine receptor CXCR5 to drive development of secondary lymphoid tissue as well as B cell and Tfh cell trafficking to germinal centers (GC), which leads to the differentiation of B cells to plasma cells and memory B cells. CXCL13 has been proposed as a general plasma biomarker for GC activities. In HIV-1 infected individuals, plasma CXCL13 levels have been associated with the rate of disease progression to AIDS. Moreover, CXCL13 production has been reported to be increased in HIV-1-infected lymph nodes, which may drive increased downregulation of CXCR5. In this review, we address the role of CXCL13 in HIV-1 infected individuals with regard to GC formation, generation of broadly neutralizing antibodies after infection and vaccination, and AIDS-related B cell lymphoma.


Sujet(s)
Vaccins contre le SIDA/immunologie , Chimiokine CXCL13/immunologie , Anticorps anti-VIH/immunologie , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Production d'anticorps/immunologie , Anticorps neutralisants à large spectre/immunologie , Humains
10.
Pathogens ; 10(2)2021 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-33573221

RÉSUMÉ

Both SARS-CoV-2 infections and vaccines induce robust immune responses. Current data suggested that high neutralizing antibody titers with sustained Th1 responses might correlate with protection against viral transmission and disease development and severity. In addition, genetic and innate immune factors, including higher levels of type I interferons, as well as the induction of trained immunity and local mucosal immunity also contribute to lower risk of infection and amelioration of disease severity. The identification of immune correlates of protection will facilitate the development of effective vaccines and therapeutics strategies.

11.
J Virol ; 94(24)2020 11 23.
Article de Anglais | MEDLINE | ID: mdl-32967951

RÉSUMÉ

An efficacious human immunodeficiency virus (HIV) vaccine will likely require induction of both mucosal and systemic immune responses. We compared the immunogenicity and protective efficacy of two mucosal/systemic vaccine regimens and investigated their effects on the rectal microbiome. Rhesus macaques were primed twice mucosally with replication-competent adenovirus type 5 host range mutant (Ad5hr)-simian immunodeficiency virus (SIV) recombinants and boosted twice intramuscularly with ALVAC-SIV recombinant plus SIV gp120 protein or with DNA for SIV genes and rhesus interleukin-12 plus SIV gp120 protein. Controls received empty Ad5hr vector and alum adjuvant only. Both regimens elicited strong, comparable mucosal and systemic cellular and humoral immunity. Prevaccination rectal microbiomes of males and females differed and significantly changed over the course of immunization, most strongly in females after Ad5hr immunizations. Following repeated low-dose intrarectal SIV challenges, both vaccine groups exhibited modestly but significantly reduced acute viremia. Male and female controls exhibited similar acute viral loads; however, vaccinated females, but not males, exhibited lower levels of acute viremia, compared to same-sex controls. Few differences in adaptive immune responses were observed between the sexes. Striking differences in correlations of the rectal microbiome of males and females with acute viremia and immune responses associated with protection were seen and point to effects of the microbiome on vaccine-induced immunity and viremia control. Our study clearly demonstrates direct effects of a mucosal SIV vaccine regimen on the rectal microbiome and validates our previously reported SIV vaccine-induced sex bias. Sex and the microbiome are critical factors that should not be overlooked in vaccine design and evaluation.IMPORTANCE Differences in HIV pathogenesis between males and females, including immunity postinfection, have been well documented, as have steroid hormone effects on the microbiome, which is known to influence mucosal immune responses. Few studies have applied this knowledge to vaccine trials. We investigated two SIV vaccine regimens combining mucosal priming immunizations and systemic protein boosting. We again report a vaccine-induced sex bias, with female rhesus macaques but not males displaying significantly reduced acute viremia. The vaccine regimens, especially the mucosal primes, significantly altered the rectal microbiome. The greatest effects were in females. Striking differences between female and male macaques in correlations of prevalent rectal bacteria with viral loads and potentially protective immune responses were observed. Effects of the microbiome on vaccine-induced immunity and viremia control require further study by microbiome transfer. However, the findings presented highlight the critical importance of considering effects of sex and the microbiome in vaccine design and evaluation.


Sujet(s)
Rappel de vaccin/méthodes , Macaca mulatta/immunologie , Microbiote/effets des médicaments et des substances chimiques , Rectum/microbiologie , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Virémie/immunologie , Vaccins contre le SIDA/immunologie , Adenoviridae/génétique , Animaux , Femelle , Immunité humorale , Immunité muqueuse , Mâle , Microbiote/physiologie , Rectum/immunologie , Vaccins contre le SIDA simien/immunologie
12.
J Clin Invest ; 130(6): 2789-2799, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32420917

RÉSUMÉ

The critical role of suppressive myeloid cells in immune regulation has come to the forefront in cancer research, with myeloid-derived suppressor cells (MDSCs) as a main oncology immunotherapeutic target. Recent improvement and standardization of criteria classifying tumor-induced MDSCs have led to unified descriptions and also promoted MDSC research in tuberculosis (TB) and AIDS. Despite convincing evidence on the induction of MDSCs by pathogen-derived molecules and inflammatory mediators in TB and AIDS, very little attention has been given to their therapeutic modulation or roles in vaccination in these diseases. Clinical manifestations in TB are consequences of complex host-pathogen interactions and are substantially affected by HIV infection. Here we summarize the current understanding and knowledge gaps regarding the role of MDSCs in HIV and Mycobacterium tuberculosis (co)infections. We discuss key scientific priorities to enable application of this knowledge to the development of novel strategies to improve vaccine efficacy and/or implementation of enhanced treatment approaches. Building on recent findings and potential for cross-fertilization between oncology and infection biology, we highlight current challenges and untapped opportunities for translating new advances in MDSC research into clinical applications for TB and AIDS.


Sujet(s)
Syndrome d'immunodéficience acquise , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Mycobacterium tuberculosis/immunologie , Cellules myéloïdes suppressives/immunologie , Tuberculose , Syndrome d'immunodéficience acquise/immunologie , Syndrome d'immunodéficience acquise/anatomopathologie , Syndrome d'immunodéficience acquise/thérapie , Humains , Cellules myéloïdes suppressives/anatomopathologie , Tuberculose/immunologie , Tuberculose/anatomopathologie , Tuberculose/thérapie
13.
Front Immunol ; 11: 315, 2020.
Article de Anglais | MEDLINE | ID: mdl-32184782

RÉSUMÉ

Trained innate immunity has recently emerged as a novel concept of innate immune cells, such as myeloid cells, exhibiting immune memory, and nonspecific heterologous immunity to protect against infections. The memory and specificity are mediated by epigenetic, metabolic, and functional reprogramming of the myeloid cells and myeloid progenitors (and/or NK cells) in the bone marrow and peripheral tissues such as gut and lung mucosa. A variety of agents, such as BCG, viruses, and their components, as well as TLR agonists, and cytokines have been shown to be involved in the induction of trained immunity. Since these agents have been widely used in AIDS vaccine development as antigen delivery vectors or adjuvants, myeloid cell mediated trained immunity might also play an important role in protecting against mucosal AIDS virus transmission or in control of virus replication in the major gut mucosal reservoir. Here we review the trained innate immunity induced by these vectors/adjuvants that have been used in AIDS vaccine studies and discuss their role in mucosal vaccine efficacy and possible utilization in AIDS vaccine development. Delineating the protective effect of the trained innate immunity mediated by myeloid cells will guide the design of novel AIDS vaccines.


Sujet(s)
Vaccins contre le SIDA/immunologie , Syndrome d'immunodéficience acquise/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Cellules souches hématopoïétiques/physiologie , Muqueuse intestinale/immunologie , Cellules myéloïdes/immunologie , Immunité acquise , Animaux , Humains , Immunité innée , Mémoire immunologique
14.
Front Immunol ; 10: 1072, 2019.
Article de Anglais | MEDLINE | ID: mdl-31139193

RÉSUMÉ

Vaccination with DNA-SIV + ALVAC-SIV + gp120 alum results in inflammasome activation, high levels of IL-1ß production, emergency myelopoiesis, and the egress of CXCR4+ CD14+ pre-monocytes from bone marrow. Previously we have shown that this vaccine-induced innate monocyte memory is associated with decreased risk of SIVmac251 acquisition. Because IL-1ß also promotes the propagation of monocyte-derived suppressor (M-MDSC)-like cells, here we extended our analysis to this negative regulator subset, characterizing its levels and functions in macaques. Interestingly, we found that DNA prime engages M-MDSC-like cells and their levels are positively associated with the frequency of CD14+ classical monocytes, and negatively with the levels of CD16+ monocytes, correlates of decreased and increased risk of SIV acquisition, respectively. Accordingly, M-MDSC frequency, arginase activity, and NO were all associated with decrease of CD8 T cells responses and worse vaccination outcome. DNA vaccination thus induces innate immunity by engaging three subsets of myeloid cells, M-MDSCs, CD14+ innate monocyte memory, and CD16+ monocytes all playing different role in protection. The full characterization of the immunological space created by myeloid cell crosstalk will likely provide clues to improve the efficacy of HIV vaccine candidates.


Sujet(s)
Protéine d'enveloppe gp120 du VIH/immunologie , Cellules myéloïdes suppressives/physiologie , Vaccins contre le SIDA simien/immunologie , Vaccins à ADN/immunologie , Vaccins antiviraux/immunologie , Animaux , Arginase/sang , Arginase/physiologie , Lymphocytes B/immunologie , Communication cellulaire , Antigènes HLA-DR/analyse , Macaca mulatta , Monoxyde d'azote/physiologie , Espèces réactives de l'oxygène/métabolisme , Lymphocytes T/immunologie
15.
J Clin Invest ; 129(3): 1314-1328, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30776026

RÉSUMÉ

It is widely believed that protection against acquisition of HIV or SIV infection requires anti-envelope (anti-Env) antibodies, and that cellular immunity may affect viral loads but not acquisition, except in special cases. Here we provide evidence to the contrary. Mucosal immunization may enhance HIV vaccine efficacy by eliciting protective responses at portals of exposure. Accordingly, we vaccinated macaques mucosally with HIV/SIV peptides, modified vaccinia Ankara-SIV (MVA-SIV), and HIV-gp120-CD4 fusion protein plus adjuvants, which consistently reduced infection risk against heterologous intrarectal SHIVSF162P4 challenge, both high dose and repeated low dose. Surprisingly, vaccinated animals exhibited no anti-gp120 humoral responses above background and Gag- and Env-specific T cells were induced but failed to correlate with viral acquisition. Instead, vaccine-induced gut microbiome alteration and myeloid cell accumulation in colorectal mucosa correlated with protection. Ex vivo stimulation of the myeloid cell-enriched population with SHIV led to enhanced production of trained immunity markers TNF-α and IL-6, as well as viral coreceptor agonist MIP1α, which correlated with reduced viral Gag expression and in vivo viral acquisition. Overall, our results suggest mechanisms involving trained innate mucosal immunity together with antigen-specific T cells, and also indicate that vaccines can have critical effects on the gut microbiome, which in turn can affect resistance to infection. Strategies to elicit similar responses may be considered for vaccine designs to achieve optimal protective efficacy.


Sujet(s)
Vaccins contre le SIDA/immunologie , Syndrome d'immunodéficience acquise/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Immunité muqueuse , Muqueuse intestinale/immunologie , Vaccins contre le SIDA simien/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Syndrome d'immunodéficience acquise/anatomopathologie , Syndrome d'immunodéficience acquise/prévention et contrôle , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/anatomopathologie , Côlon/immunologie , Côlon/anatomopathologie , Immunité cellulaire , Muqueuse intestinale/anatomopathologie , Macaca mulatta , Rectum/immunologie , Rectum/anatomopathologie , Syndrome d'immunodéficience acquise du singe/anatomopathologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle
16.
Mucosal Immunol ; 11(4): 1219-1229, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29858581

RÉSUMÉ

It is unknown whether the gut microbiome affects HIV transmission. In our recent SHIV vaccine study, we found that the naive rhesus macaques from two different sources had significantly different rates of infection against repeated low-dose intrarectal challenge with SHIVSF162P4 virus. Exploring causes, we found that the more susceptible group of seven macaques had significantly more activated CD4+CCR5+Ki67+ T cells in the rectal mucosa than the more resistant group of 11 macaques from a different source. The prevalence of pre-challenge activated rectal CD4 T cells in the naive macaques correlated inversely with the number of challenges required to infect. Because the two naive groups came from different sources, we hypothesized that their microbiomes may differ and might explain the activation difference. Indeed, after sequencing 16s rRNA, we found differences between the two naive groups that correlated with immune activation status. Distinct gut microbiota induced different levels of immune activation ex vivo. Significantly lower ratios of Bacteroides to Prevotella, and significantly lower levels of Firmicutes were found in the susceptible cohort, which were also inversely correlated with high levels of immune activation in the rectal mucosa. Thus, host-microbiome interactions might influence HIV/SIV mucosal transmission through effects on mucosal immune activation.


Sujet(s)
Bacteroides/physiologie , Lymphocytes T CD4+/immunologie , Firmicutes/physiologie , Microbiome gastro-intestinal/immunologie , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Muqueuse intestinale/immunologie , Prevotella/physiologie , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/physiologie , Animaux , Prédisposition aux maladies , Infections à VIH/microbiologie , Humains , Muqueuse intestinale/microbiologie , Antigène KI-67/métabolisme , Activation des lymphocytes , Macaca , ARN ribosomique 16S/analyse , Récepteurs CCR5/métabolisme , Syndrome d'immunodéficience acquise du singe/microbiologie
17.
J Immunol ; 200(5): 1853-1864, 2018 03 01.
Article de Anglais | MEDLINE | ID: mdl-29374075

RÉSUMÉ

Unlike cytosolic processing and presentation of viral Ags by virus-infected cells, Ags first expressed in infected nonprofessional APCs, such as CD4+ T cells in the case of HIV, are taken up by dendritic cells and cross-presented. This generally requires entry through the endocytic pathway, where endosomal proteases have first access for processing. Thus, understanding virus escape during cross-presentation requires an understanding of resistance to endosomal proteases, such as cathepsin S (CatS). We have modified HIV-1MN gp120 by mutating a key CatS cleavage site (Thr322Thr323) in the V3 loop of the immunodominant epitope IGPGRAFYTT to IGPGRAFYVV to prevent digestion. We found this mutation to facilitate cross-presentation and provide evidence from MHC binding and X-ray crystallographic structural studies that this results from preservation of the epitope rather than an increased epitope affinity for the MHC class I molecule. In contrast, when the protein is expressed by a vaccinia virus in the cytosol, the wild-type protein is immunogenic without this mutation. These proof-of-concept results show that a virus like HIV, infecting predominantly nonprofessional presenting cells, can escape T cell recognition by incorporating a CatS cleavage site that leads to destruction of an immunodominant epitope when the Ag undergoes endosomal cross-presentation.


Sujet(s)
Présentation d'antigène/immunologie , Lymphocytes T CD4+/immunologie , Cross-priming/immunologie , Infections à VIH/immunologie , VIH (Virus de l'Immunodéficience Humaine)/immunologie , Échappement immunitaire/immunologie , Peptides/immunologie , Animaux , Cathepsines/immunologie , Cellules dendritiques/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Cellules HEK293 , Protéine d'enveloppe gp120 du VIH/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Épitopes immunodominants/immunologie , Souris , Souris de lignée BALB C , Virus de la vaccine/immunologie
18.
J Leukoc Biol ; 102(6): 1381-1388, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-28951425

RÉSUMÉ

Mechanisms that imprint T cell homing to the small intestine have been well studied; however, those for homing to the colon are poorly understood. Recently, we found that these are distinct subcompartments of the gut mucosal immune system, which implies differential homing. Here, we show that colonic CD11c+ APCs imprint CD8+ T cell preferential homing to the colon, in contrast to those from the small intestine that imprint CD8+ T cell homing to the small intestine, and that the differences are related to the variable ability of APCs to induce α4ß7-integrin and CCR9 expression on T cells. Colon APCs also expressed lower levels of retinoic acid-producing enzymes that are known to control the mucosal homing of T cells. These findings are the first to our knowledge to directly demonstrate that colon APCs imprint T cells to selectively home to the large bowel, which is critical for the design of successful T cell-based therapies and vaccines, such as colon cancer immunotherapy and HIV vaccines.


Sujet(s)
Cellules présentatrices d'antigène/immunologie , Antigènes CD11c/métabolisme , Côlon/immunologie , Intestin grêle/immunologie , Récepteurs d'écotaxie des lymphocytes/métabolisme , Lymphocytes T/immunologie , Animaux , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Compartimentation cellulaire , Mouvement cellulaire , Cellules cultivées , Cellules dendritiques/cytologie , Immunisation , Intégrines/métabolisme , Muqueuse intestinale/immunologie , Souris de lignée C57BL , Récepteurs CCR/métabolisme , Lymphocytes T/cytologie , Trétinoïne/métabolisme
19.
Eur J Immunol ; 47(12): 2059-2069, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-28741316

RÉSUMÉ

The ability of different CD4+ T cell subsets to help CD8+ T-cell response is not fully understood. Here, we found using the murine system that Th17 cells induced by IL-1ß, unlike Th1, were not effective helpers for antiviral CD8 responses as measured by IFNγ-producing cells or protection against virus infection. However, they skewed CD8 responses to a Tc17 phenotype. Thus, the apparent lack of help was actually immune deviation. This skewing depended on both IL-21 and IL-23. To overcome this effect, we inhibited Th17 induction by blocking TGF-ß. Anti-TGF-ß allowed the IL-1ß adjuvant to enhance CD8+ T-cell responses without skewing the phenotype to Tc17, thereby providing an approach to harness the benefit of common IL-1-inducing adjuvants like alum without immune deviation.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Immunité cellulaire/immunologie , Lymphocytes T auxiliaires/immunologie , Animaux , Anticorps bloquants/immunologie , Anticorps bloquants/pharmacologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/métabolisme , Déterminants antigéniques des lymphocytes T/immunologie , Cytométrie en flux , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukine-1 bêta/immunologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/pharmacologie , Souris de lignée BALB C , Souris de lignée C57BL , Lymphocytes T auxiliaires/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/immunologie , Cellules Th17/métabolisme , Facteur de croissance transformant bêta/immunologie , Facteur de croissance transformant bêta/métabolisme
20.
PLoS Pathog ; 13(5): e1006395, 2017 May.
Article de Anglais | MEDLINE | ID: mdl-28498847

RÉSUMÉ

Myeloid derived suppressor cells (MDSCs), which suppress anti-tumor or anti-viral immune responses, are expanded in the peripheral blood and tissues of patients/animals with cancer or viral infectious diseases. We here show that in chronic SIV infection of Indian rhesus macaques, the frequency of MDSCs in the bone marrow (BM) was paradoxically and unexpectedly decreased, but increased in peripheral blood. Reduction of BM MDSCs was found in both CD14+MDSC and Lin-CD15+MDSC subsets. The reduction of MDSCs correlated with high plasma viral loads and low CD4+ T cell counts, suggesting that depletion of BM MDSCs was associated with SIV/AIDS disease progression. Of note, in SHIVSF162P4-infected macaques, which naturally control viral replication within a few months of infection, the frequency of MDSCs in the bone marrow was unchanged. To investigate the mechanisms by which BM MDSCs were reduced during chronic SIV infection, we tested several hypotheses: depletion due to viral infection, alterations in MDSC trafficking, and/or poor MDSC replenishment. We found that the possible mobilization of MDSCs from BM to peripheral tissues and the slow self-replenishment of MDSCs in the BM, along with the viral infection-induced depletion, all contribute to the observed BM MDSC reduction. We first demonstrate MDSC SIV infection in vivo. Correlation between BM CD14+MDSC reduction and CD8+ T cell activation in tissues is consistent with decreased immune suppression by MDSCs. Thus, depletion of BM MDSCs may contribute to the pathologic immune activation during chronic SIV infection and by extension HIV infection.


Sujet(s)
Lymphocytes T CD4+/immunologie , Macaca mulatta , Cellules myéloïdes suppressives/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Moelle osseuse/immunologie , Moelle osseuse/virologie , Modèles animaux de maladie humaine , Femelle , Humains , Activation des lymphocytes , Mâle , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/physiologie , Charge virale , Réplication virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...