Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 623
Filtrer
1.
mBio ; : e0163924, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39258922

RÉSUMÉ

Sooty mangabeys (SMs) are natural hosts of simian immunodeficiency virus (SIV) and do not progress to AIDS despite high viral replication. The main factors involved in the benign nature of this infection are (i) low level of immune activation, (ii) relative preservation of specific CD4+ T-cell subsets from direct virus infection, and (iii) absence of microbial translocation from the gut to the systemic circulation. To determine the impact of SIV infection on underlying cause of death, we retrospectively analyzed data from 307 SMs (219 SIV infected and 88 uninfected) housed at the Emory Primate Center that have died between 1986 and 2022. Interestingly, we found that SIV-infected SMs live ~4 years longer than SIV-uninfected SMs, although this result is hard to interpret due to differences in how animals were housed and assigned to specific experimental studies. While the causes of death were not different between SIV-infected and uninfected SMs that died before age 15 (i.e., adult), we found significant differences in the relative frequency of specific causes of death in the elderly population (≥15 years old). Specifically, we observed that SIV-infected SMs were more likely to die from infections but less likely to die from cardiovascular disease (and diabetes in female animals) as compared to uninfected SMs. While confirming the non-pathogenic nature of SIV infection in SMs, these data reveal, for the first time, a qualitative impact of SIV infection on the host physiology that induces a significant change in the mortality pattern in these natural SIV hosts. IMPORTANCE: In this study, we demonstrate, for the first time, that the natural, non-pathogenic SIV infection of the African monkey SM has a clinical impact which is revealed in terms of main causes of mortality, which are significantly different in the infected animals as compared to the uninfected ones. Indeed, SIV-infected SMs are at higher risk of dying of infectious diseases but appear to be somewhat protected from cardiovascular causes of death. The identification of a specific pattern of mortality associated with the infection suggests that the host-pathogen interaction between SIV and the SM immune system, while non-pathogenic in nature, has a detectable impact on the overall health status of the animals.

2.
Int J Mol Sci ; 25(16)2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39201388

RÉSUMÉ

HIV-induced persistent immune activation is a key mediator of inflammatory comorbidities such as cardiovascular disease (CVD) and neurocognitive disorders. While a preponderance of data indicate that gut barrier disruption and microbial translocation are drivers of chronic immune activation, the molecular mechanisms of this persistent inflammatory state remain poorly understood. Here, utilizing the nonhuman primate model of Human Immunodeficiency Virus (HIV) infection with suppressive antiretroviral therapy (ART), we investigated activation of inflammasome pathways and their association with intestinal epithelial barrier disruption (IEBD). Longitudinal blood samples obtained from rhesus macaques with chronic SIV infection and long-term suppressive ART were evaluated for IEBD biomarkers, inflammasome activation (IL-1ß and IL-18), inflammatory cytokines, and triglyceride (TG) levels. Activated monocyte subpopulations and glycolytic potential were investigated in peripheral blood mononuclear cells (PBMCs). During the chronic phase of treated SIV infection, elevated levels of plasma IL-1ß and IL-18 were observed following the hallmark increase in IEBD biomarkers, intestinal fatty acid-binding protein (IFABP) and LPS-binding protein (LBP). Further, significant correlations of plasma IFABP levels with IL-1ß and IL-18 were observed between 10 and 12 months of ART. Higher levels of sCD14, IL-6, and GM-CSF, among other inflammatory mediators, were also observed only during the long-term SIV + ART phase along with a trend of increase in the frequencies of activated CD14+CD16+ intermediate monocyte subpopulations. Lastly, we found elevated levels of blood TG and higher glycolytic capacity in PBMCs of chronic SIV-infected macaques with long-term ART. The increase in circulating IL-18 and IL-1ß following IEBD and their significant positive correlation with IFABP suggest a connection between gut barrier disruption and inflammasome activation during chronic SIV infection, despite viral suppression with ART. Additionally, the increase in markers of monocyte activation, along with elevated TG and enhanced glycolytic pathway activity, indicates metabolic remodeling that could fuel metabolic syndrome. Further research is needed to understand the mechanisms by which gut dysfunction and inflammasome activation contribute to HIV-associated metabolic complications, enabling targeted interventions in people with HIV.


Sujet(s)
Interleukine-18 , Interleukine-1 bêta , Muqueuse intestinale , Macaca mulatta , Monocytes , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/sang , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Interleukine-18/sang , Interleukine-18/métabolisme , Monocytes/métabolisme , Monocytes/immunologie , Interleukine-1 bêta/sang , Interleukine-1 bêta/métabolisme , Muqueuse intestinale/métabolisme , Antirétroviraux/usage thérapeutique , Inflammasomes/métabolisme , Marqueurs biologiques/sang , Mâle , Agranulocytes/métabolisme , Maladie chronique
3.
Front Immunol ; 15: 1444621, 2024.
Article de Anglais | MEDLINE | ID: mdl-39170621

RÉSUMÉ

Simian immunodeficiency virus (SIV) vaccines based upon 68-1 Rhesus Cytomegalovirus (RhCMV) vectors show remarkable protection against pathogenic SIVmac239 challenge. Across multiple independent rhesus macaque (RM) challenge studies, nearly 60% of vaccinated RM show early, complete arrest of SIVmac239 replication after effective challenge, whereas the remainder show progressive infection similar to controls. Here, we performed viral sequencing to determine whether the failure to control viral replication in non-protected RMs is associated with the acquisition of viral escape mutations. While low level viral mutations accumulated in all animals by 28 days-post-challenge, which is after the establishment of viral control in protected animals, the dominant circulating virus in virtually all unprotected RMs was nearly identical to the challenge stock, and there was no difference in mutation patterns between this cohort and unvaccinated controls. These data definitively demonstrate that viral mutation does not explain lack of viral control in RMs not protected by RhCMV/SIV vaccination. We further demonstrate that during chronic infection RhCMV/SIV vaccinated RMs do not acquire escape mutation in epitopes targeted by RhCMV/SIV, but instead display mutation in canonical MHC-Ia epitopes similar to unvaccinated RMs. This suggests that after the initial failure of viral control, unconventional T cell responses induced by 68-1 RhCMV/SIV vaccination do not exert strong selective pressure on systemically replicating SIV.


Sujet(s)
Macaca mulatta , Mutation , Vaccins contre le SIDA simien , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Virus de l'immunodéficience simienne/immunologie , Virus de l'immunodéficience simienne/génétique , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Vaccins contre le SIDA simien/immunologie , Vaccins contre le SIDA simien/génétique , Cytomegalovirus/immunologie , Cytomegalovirus/génétique , Réplication virale/immunologie , Vaccination , Échappement immunitaire/génétique
4.
Article de Anglais | MEDLINE | ID: mdl-39041622

RÉSUMÉ

Multifaceted natural killer (NK) cell activities are indispensable for controlling human immunodeficiency virus (HIV)-1 transmission and pathogenesis. Among the diverse functions of NK cells, antibody-dependent cellular cytotoxicity (ADCC) has been shown to predict better HIV-1 protection. ADCC is initiated by the engagement of an Fc γ receptor CD16 with an Fc portion of the antibody, leading to phosphorylation of the CD3 ζ chain (CD3ζ) and Fc receptor γ chain (FcRγ) as well as downstream signaling activation. Though CD3ζ and FcRγ were thought to have overlapping roles in NK cell ADCC, several groups have reported that CD3ζ-mediated signals trigger a more robust ADCC. However, few studies have illustrated the direct contribution of CD3ζ in HIV-1-specific ADCC. To further understand the roles played by CD3ζ in HIV-1-specific ADCC, we developed a CD3ζ knockdown system in primary human NK cells. We observed that HIV-1-specific ADCC was inhibited by CD3ζ perturbation. In summary, we demonstrated that CD3ζ is important for eliciting HIV-1-specific ADCC, and this dynamic can be utilized for NK cell immunotherapeutics against HIV-1 infection and other diseases.

5.
Brain ; 147(9): 3059-3069, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39049445

RÉSUMÉ

Perivascular macrophages (PVMs) and, to a lesser degree, microglia are targets and reservoirs of HIV and simian immunodeficiency virus (SIV) in the brain. Previously, we demonstrated that colony-stimulating factor 1 receptor (CSF1R) in PVMs was upregulated and activated in chronically SIV-infected rhesus macaques with encephalitis, correlating with SIV infection of PVMs. Herein, we investigated the role of CSF1R in the brain during acute SIV infection using BLZ945, a brain-penetrant CSF1R kinase inhibitor. Apart from three uninfected historic controls, nine Indian rhesus macaques were infected acutely with SIVmac251 and divided into three groups (n = 3 each): an untreated control and two groups treated for 20-30 days with low- (10 mg/kg/day) or high- (30 mg/kg/day) dose BLZ945. With the high-dose BLZ945 treatment, there was a significant reduction in cells expressing CD163 and CD206 across all four brain areas examined, compared with the low-dose treatment and control groups. In 9 of 11 tested regions, tissue viral DNA (vDNA) loads were reduced by 95%-99% following at least one of the two doses, and even to undetectable levels in some instances. Decreased numbers of CD163+ and CD206+ cells correlated significantly with lower levels of vDNA in all four corresponding brain areas. In contrast, BLZ945 treatment did not significantly affect the number of microglia. Our results indicate that doses as low as 10 mg/kg/day of BLZ945 are sufficient to reduce the tissue vDNA loads in the brain with no apparent adverse effect. This study provides evidence that infected PVMs are highly sensitive to CSF1R inhibition, opening new possibilities to achieve viral clearance.


Sujet(s)
Encéphale , Macaca mulatta , Macrophages , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/virologie , Récepteur de facteur de croissance granulocyte-macrophage/antagonistes et inhibiteurs , Récepteur de facteur de croissance granulocyte-macrophage/métabolisme , Charge virale/effets des médicaments et des substances chimiques , Pyrimidines/pharmacologie , Pyrimidines/usage thérapeutique , Antigènes CD/métabolisme , Mâle , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Microglie/virologie , Antigènes de différenciation des myélomonocytes/métabolisme , Récepteurs de surface cellulaire/métabolisme , Anisoles
6.
Front Cell Infect Microbiol ; 14: 1408245, 2024.
Article de Anglais | MEDLINE | ID: mdl-39006742

RÉSUMÉ

While simian immunodeficiency virus (SIV) infection is non-pathogenic in naturally infected African nonhuman primate hosts, experimental or accidental infection in rhesus macaques often leads to AIDS. Baboons, widely distributed throughout Africa, do not naturally harbor SIV, and experimental infection of baboons with SIVmac results in transient low-level viral replication. Elucidation of mechanisms of natural immunity in baboons could uncover new targets of antiviral intervention. We tested the hypothesis that an SIVmac adapted to replicate in baboon primary cells will gain the capacity to establish chronic infections in vivo. Here, we generated SIVmac variants in baboon cells through serial passage in PBMC from different donors (SIVbn-PBMC s1), in PBMC from the same donors (SIVbn-PBMC s2), or in isolated CD4 cells from the same donors used for series 2 (SIVbn-CD4). While SIVbn-PBMC s1 and SIVbn-CD4 demonstrated increased replication capacity, SIVbn-PBMC s2 did not. Pharmacological blockade of CCR5 revealed SIVbn-PBMC s1 could more efficiently use available CCR5 than SIVmac, a trait we hypothesize arose to circumvent receptor occupation by chemokines. Sequencing analysis showed that all three viruses accumulated different types of mutations, and that more non-synonymous mutations became fixed in SIVbn-PBMC s1 than SIVbn-PBMC s2 and SIVbn-CD4, supporting the notion of stronger fitness pressure in PBMC from different genetic backgrounds. Testing the individual contribution of several newly fixed SIV mutations suggested that is the additive effect of these mutations in SIVbn-PBMC s1 that contributed to its enhanced fitness, as recombinant single mutant viruses showed no difference in replication capacity over the parental SIVmac239 strain. The replicative capacity of SIVbn-PBMC passage 4 (P4) s1 was tested in vivo by infecting baboons intravenously with SIVbn-PBMC P4 s1 or SIVmac251. While animals infected with SIVmac251 showed the known pattern of transient low-level viremia, animals infected with SIVbn-PBMC P4 s1 had undetectable viremia or viral DNA in lymphoid tissue. These studies suggest that adaptation of SIV to grow in baboon primary cells results in mutations that confer increased replicative capacity in the artificial environment of cell culture but make the virus unable to avoid the restrictive factors generated by a complex multicellular organism.


Sujet(s)
Papio , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Réplication virale , Animaux , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/physiologie , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/immunologie , Agranulocytes/virologie , Agranulocytes/immunologie , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/immunologie , Cellules cultivées , Passages en série
7.
Viruses ; 16(7)2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39066335

RÉSUMÉ

The effects of immunodeficiency associated with chronic HIV infection on COVID-19 disease and viral persistence have not been directly addressed in a controlled setting. In this pilot study, we exposed two pigtail macaques (PTMs) chronically infected with SIVmac239, exhibiting from very low to no CD4 T cells across all compartments, to SARS-CoV-2. We monitored the disease progression, viral replication, and evolution, and compared these outcomes with SIV-naïve PTMs infected with SARS-CoV-2. No overt signs of COVID-19 disease were observed in either animal, and the SARS-CoV-2 viral kinetics and evolution in the SIVmac239 PTMs were indistinguishable from those in the SIV-naïve PTMs in all sampled mucosal sites. However, the single-cell RNA sequencing of bronchoalveolar lavage cells revealed an infiltration of functionally inert monocytes after SARS-CoV-2 infection. Critically, neither of the SIV-infected PTMs mounted detectable anti-SARS-CoV-2 T-cell responses nor anti-SARS-CoV-2 binding or neutralizing antibodies. Thus, HIV-induced immunodeficiency alone may not be sufficient to drive the emergence of novel viral variants but may remove the ability of infected individuals to mount adaptive immune responses against SARS-CoV-2.


Sujet(s)
COVID-19 , Co-infection , Modèles animaux de maladie humaine , SARS-CoV-2 , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Virus de l'immunodéficience simienne/immunologie , COVID-19/immunologie , COVID-19/virologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , SARS-CoV-2/immunologie , Co-infection/immunologie , Co-infection/virologie , Réplication virale , Macaca nemestrina , Projets pilotes , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Charge virale , Lymphocytes T CD4+/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang
8.
J Virol ; 98(7): e0029424, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38829139

RÉSUMÉ

Arenaviral vaccine vectors encoding simian immunodeficiency virus (SIV) immunogens are capable of inducing efficacious humoral and cellular immune responses in nonhuman primates. Several studies have evaluated the use of immune modulators to further enhance vaccine-induced T-cell responses. The hematopoietic growth factor Flt3L drives the expansion of various bone marrow progenitor populations, and administration of Flt3L was shown to promote expansion of dendritic cell populations in spleen and blood, which are targets of arenaviral vectors. Therefore, we evaluated the potential of Flt3 signaling to enhance the immunogenicity of arenaviral vaccines encoding SIV immunogens (SIVSME543 Gag, Env, and Pol) in rhesus macaques, with a rhesus-specific engineered Flt3L-Fc fusion protein. In healthy animals, administration of Flt3L-Fc led to a 10- to 100-fold increase in type 1 dendritic cells 7 days after dosing, with no antidrug antibody (ADA) generation after repeated dosing. We observed that administration of Flt3L-Fc fusion protein 7 days before arenaviral vaccine increased the frequency and activation of innate immune cells and enhanced T-cell activation with no treatment-related adverse events. Flt3L-Fc administration induced early innate immune activation, leading to a significant enhancement in magnitude, breadth, and polyfunctionality of vaccine-induced T-cell responses. The Flt3L-Fc enhancement in vaccine immunogenicity was comparable to a combination with αCTLA-4 and supports the use of safe and effective variants of Flt3L to augment therapeutic vaccine-induced T-cell responses.IMPORTANCEInduction of a robust human immunodeficiency virus (HIV)-specific CD4+ and CD8+ T-cell response through therapeutic vaccination is considered essential for HIV cure. Arenaviral vaccine vectors encoding simian immunodeficiency virus (SIV) immunogens have demonstrated strong immunogenicity and efficacy in nonhuman primates. Here, we demonstrate that the immunogenicity of arenaviral vectors encoding SIV immunogens can be enhanced by administration of Flt3L-Fc fusion protein 7 days before vaccination. Flt3L-Fc-mediated increase in dendritic cells led to robust improvements in vaccine-induced T- and B-cell responses compared with vaccine alone, and Flt3L-Fc dosing was not associated with any treatment-related adverse events. Importantly, immune modulation by either Flt3L-Fc or αCTLA-4 led to comparable enhancement in vaccine response. These results indicate that the addition of Flt3L-Fc fusion protein before vaccine administration can significantly enhance vaccine immunogenicity. Thus, safe and effective Flt3L variants could be utilized as part of a combination therapy for HIV cure.


Sujet(s)
Cellules dendritiques , Macaca mulatta , Vaccins contre le SIDA simien , Virus de l'immunodéficience simienne , Animaux , Virus de l'immunodéficience simienne/immunologie , Cellules dendritiques/immunologie , Vaccins contre le SIDA simien/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Protéines membranaires/immunologie , Protéines membranaires/génétique , Tyrosine kinase-3 de type fms/immunologie , Tyrosine kinase-3 de type fms/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Vecteurs génétiques , Immunogénicité des vaccins , Lymphocytes T CD8+/immunologie
9.
J Virol ; 98(6): e0028324, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38780248

RÉSUMÉ

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.


Sujet(s)
Antirétroviraux , Épigenèse génétique , Cellules T mémoire , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Transcriptome , Animaux , Antirétroviraux/usage thérapeutique , Antirétroviraux/pharmacologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Épigenèse génétique/effets des médicaments et des substances chimiques , Macaca mulatta/génétique , Macaca mulatta/immunologie , Macaca mulatta/virologie , Macaca nemestrina/génétique , Macaca nemestrina/immunologie , Macaca nemestrina/virologie , Cellules T mémoire/effets des médicaments et des substances chimiques , Cellules T mémoire/immunologie , Cellules T mémoire/métabolisme , Cellules T mémoire/virologie , Proteasome endopeptidase complex/génétique , RNA-Seq , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/génétique , Virus de l'immunodéficience simienne/immunologie , Virus de l'immunodéficience simienne/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Virémie/traitement médicamenteux , Virémie/génétique , Virémie/immunologie , Virémie/virologie
10.
J Virol ; 98(6): e0027324, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38775481

RÉSUMÉ

TIGIT is a negative immune checkpoint receptor associated with T cell exhaustion in cancer and HIV. TIGIT upregulation in virus-specific CD8+ T cells and NK cells during HIV/SIV infection results in dysfunctional effector capabilities. In vitro studies targeting TIGIT on CD8+ T cells suggest TIGIT blockade as a viable strategy to restore SIV-specific T cell responses. Here, we extend these studies in vivo using TIGIT blockage in nonhuman primates in an effort to reverse T cell and NK cell exhaustion in the setting of SIV infection. We demonstrate that in vivo administration of a humanized anti-TIGIT monoclonal antibody (mAb) is well tolerated in both cynomolgus macaques and rhesus macaques. Despite sustained plasma concentrations of anti-TIGIT mAb, we observed no consistent improvement in NK or T cell cytolytic capacity. TIGIT blockade minimally enhanced T cell proliferation and virus-specific T cell responses in both magnitude and breadth though plasma viral loads in treated animals remained stable indicating that anti-TIGIT mAb treatment alone was insufficient to increase anti-SIV CD8+ T cell function. The enhancement of virus-specific T cell proliferative responses observed in vitro with single or dual blockade of TIGIT and/or PD-1 highlights TIGIT as a potential target to reverse T cell dysfunction. Our studies, however, reveal that targeting the TIGIT pathway alone may be insufficient in the setting of viremia and that combining immune checkpoint blockade with other immunotherapeutics may be a future path forward for improved viral control or elimination of HIV.IMPORTANCEUpregulation of the immune checkpoint receptor TIGIT is associated with HIV-mediated T cell dysfunction and correlates with HIV disease progression. Compelling evidence exists for targeting immune checkpoint receptor pathways that would potentially enhance immunity and refocus effector cell efforts toward viral clearance. In this report, we investigate TIGIT blockade as an immunotherapeutic approach to reverse immune exhaustion during chronic SIV/SHIV infection in a nonhuman primate model of HIV infection. We show that interfering with the TIGIT signaling axis alone is insufficient to improve viral control despite modest improvement in T cell immunity. Our data substantiate the use of targeting multiple immune checkpoint receptors to promote synergy and ultimately eliminate HIV-infected cells.


Sujet(s)
Lymphocytes T CD8+ , Cellules tueuses naturelles , Macaca fascicularis , Macaca mulatta , Récepteurs immunologiques , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Charge virale , Animaux , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/immunologie , Récepteurs immunologiques/antagonistes et inhibiteurs , Lymphocytes T CD8+/immunologie , Charge virale/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie
11.
Viruses ; 16(4)2024 03 27.
Article de Anglais | MEDLINE | ID: mdl-38675857

RÉSUMÉ

The persistence of the latent viral reservoir is the main hurdle to curing HIV-1 infection. SIV infection of non-human primates (NHPs), namely Indian-origin rhesus macaques, is the most relevant and widely used animal model to evaluate therapies that seek to eradicate HIV-1. The utility of a model ultimately rests on how accurately it can recapitulate human disease, and while reservoirs in the NHP model behave quantitatively very similar to those of long-term suppressed persons with HIV-1 (PWH) in the most salient aspects, recent studies have uncovered key nuances at the clonotypic level that differentiate the two in qualitative terms. In this review, we will highlight differences relating to proviral intactness, clonotypic structure, and decay rate during ART between HIV-1 and SIV reservoirs and discuss the relevance of these distinctions in the interpretation of HIV-1 cure strategies. While these, to some degree, may reflect a unique biology of the virus or host, distinctions among the proviral landscape in SIV are likely to be shaped significantly by the condensed timeframe of NHP studies. ART is generally initiated earlier in the disease course, and animals are virologically suppressed for shorter periods before receiving interventions. Because these are experimental variables dictated by the investigator, we offer guidance on study design for cure-related studies performed in the NHP model. Finally, we highlight the case of GS-9620 (Vesatolimod), an antiviral TLR7 agonist tested in multiple independent pre-clinical studies in which virological outcomes may have been influenced by study-related variables.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Latence virale , Animaux , Humains , Modèles animaux de maladie humaine , Réservoirs de maladies/virologie , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Macaca mulatta , Provirus/génétique , Provirus/physiologie , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/physiologie , Charge virale , Latence virale/effets des médicaments et des substances chimiques
12.
Pathogens ; 13(4)2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38668296

RÉSUMÉ

Porcine respiratory coronavirus (PRCoV), porcine reproductive and respiratory syndrome virus (PRRSV), swine influenza virus (SIV), and pseudorabies virus (PRV) are significant viruses causing respiratory diseases in pigs. Sick pigs exhibit similar clinical symptoms such as fever, cough, runny nose, and dyspnea, making it very difficult to accurately differentially diagnose these diseases on site. In this study, a quadruplex one-step reverse-transcription real-time quantitative PCR (RT-qPCR) for the detection of PRCoV, PRRSV, SIV, and PRV was established. The assay showed strong specificity, high sensitivity, and good repeatability. It could detect only PRCoV, PRRSV, SIV, and PRV, without cross-reactions with TGEV, PEDV, PRoV, ASFV, FMDV, PCV2, PDCoV, and CSFV. The limits of detection (LODs) for PRCoV, PRRSV, SIV, and PRV were 129.594, 133.205, 139.791, and 136.600 copies/reaction, respectively. The intra-assay and inter-assay coefficients of variation (CVs) ranged from 0.29% to 1.89%. The established quadruplex RT-qPCR was used to test 4909 clinical specimens, which were collected in Guangxi Province, China, from July 2022 to September 2023. PRCoV, PRRSV, SIV, and PRV showed positivity rates of 1.36%, 10.17%, 4.87%, and 0.84%, respectively. In addition, the previously reported RT-qPCR was also used to test these specimens, and the agreement between these methods was higher than 99.43%. The established quadruplex RT-qPCR can accurately detect these four porcine respiratory viruses simultaneously, providing an accurate and reliable detection technique for clinical diagnosis.

13.
bioRxiv ; 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38617282

RÉSUMÉ

Human Immunodeficiency Virus (HIV) is widely acknowledged for its profound impact on the immune system. Although HIV primarily affects peripheral CD4 T cells, its influence on the central nervous system (CNS) cannot be overlooked. Within the brain, microglia and CNS-associated macrophages (CAMs) serve as the primary targets for HIV, as well as for the simian immunodeficiency virus (SIV) in nonhuman primates. This infection can lead to neurological effects and the establishment of a viral reservoir. Given the gaps in our understanding of how these cells respond in vivo to acute CNS infection, we conducted single-cell RNA sequencing (scRNA-seq) on myeloid cells from the brains of three rhesus macaques 12-days after SIV infection, along with three uninfected controls. Our analysis revealed six distinct microglial clusters including homeostatic microglia, preactivated microglia, and activated microglia expressing high levels of inflammatory and disease-related molecules. In response to acute SIV infection, the population of homeostatic and preactivated microglia decreased, while the activated and disease-related microglia increased. All microglial clusters exhibited upregulation of MHC class I molecules and interferon-related genes, indicating their crucial roles in defending against SIV during the acute phase. All microglia clusters also upregulated genes linked to cellular senescence. Additionally, we identified two distinct CAM populations: CD14lowCD16hi and CD14hiCD16low CAMs. Interestingly, during acute SIV infection, the dominant CAM population changed to one with an inflammatory phenotype. Notably, specific upregulated genes within one microglia and one macrophage cluster were associated with neurodegenerative pathways, suggesting potential links to neurocognitive disorders. This research sheds light on the intricate interactions between viral infection, innate immune responses, and the CNS, providing valuable insights for future investigations.

14.
AIDS Res Hum Retroviruses ; 40(9): 521-530, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38535626

RÉSUMÉ

HIV/AIDS cannot be cured because of the persistence of the viral reservoir. Because of the complexity of the cellular composition and structure of the human organs, HIV reservoirs of anatomical site are also complex. Recently, although a variety of molecules have been reported to be involved in the establishment and maintenance of the viral reservoirs, or as marker of latent cells, the research mainly focuses on blood and lymph nodes. Now, the characteristics of the viral reservoir in tissue are not yet fully understood. In this study, various tissues were collected from SIVmac239-infected monkeys, and the level of total SIV DNA, SIV 2-LTR DNA, and cell-associated virus RNA in them were compared with character of the anatomical viral reservoir under early treatment. The results showed that short-term combination antiretroviral therapy (cART) starting from 3 days after infection could significantly inhibit viremia and reduce the size of the anatomical viral reservoir, but it could not eradicate de novo infections and ongoing replication of virus. Moreover, the effects of early cART on the level of total SIV DNA, SIV 2-LTR DNA, and cell-associated virus RNA in different tissues were different, which changed the size distribution of viral reservoir in anatomical site. Finally, the contribution of nonlymphoid tissues, especially liver and lung, to the viral reservoir increased after treatment, while the contribution of intestinal lymphoid to the viral reservoir significantly reduced. These results suggested that early treatment effectively decreased the size of viral reservoir, and that the effects of cART on the tissue viral reservoir varied greatly by tissue type. The results implied that persistent existence of virus in nonlymphoid tissues after short-term treatment suggested that the role of nonlymphoid tissues cannot be ignored in development strategies for AIDS therapy.


Sujet(s)
Antirétroviraux , Macaca mulatta , ARN viral , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Charge virale , Animaux , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/isolement et purification , Antirétroviraux/usage thérapeutique , ADN viral , Virémie/virologie , Virémie/traitement médicamenteux , Latence virale , Réservoirs de maladies/virologie , Réplication virale
15.
BMC Bioinformatics ; 25(1): 125, 2024 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-38519883

RÉSUMÉ

In the battle of the host against lentiviral pathogenesis, the immune response is crucial. However, several questions remain unanswered about the interaction with different viruses and their influence on disease progression. The simian immunodeficiency virus (SIV) infecting nonhuman primates (NHP) is widely used as a model for the study of the human immunodeficiency virus (HIV) both because they are evolutionarily linked and because they share physiological and anatomical similarities that are largely explored to understand the disease progression. The HIHISIV database was developed to support researchers to integrate and evaluate the large number of transcriptional data associated with the presence/absence of the pathogen (SIV or HIV) and the host response (NHP and human). The datasets are composed of microarray and RNA-Seq gene expression data that were selected, curated, analyzed, enriched, and stored in a relational database. Six query templates comprise the main data analysis functions and the resulting information can be downloaded. The HIHISIV database, available at  https://hihisiv.github.io , provides accurate resources for browsing and visualizing results and for more robust analyses of pre-existing data in transcriptome repositories.


Sujet(s)
Infections à VIH , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Humains , Virus de l'immunodéficience simienne/génétique , VIH (Virus de l'Immunodéficience Humaine) , Syndrome d'immunodéficience acquise du singe/génétique , Évolution de la maladie , Immunité , Expression des gènes
16.
Cell ; 187(5): 1223-1237.e16, 2024 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-38428396

RÉSUMÉ

While CD4+ T cell depletion is key to disease progression in people living with HIV and SIV-infected macaques, the mechanisms underlying this depletion remain incompletely understood, with most cell death involving uninfected cells. In contrast, SIV infection of "natural" hosts such as sooty mangabeys does not cause CD4+ depletion and AIDS despite high-level viremia. Here, we report that the CARD8 inflammasome is activated immediately after HIV entry by the viral protease encapsulated in incoming virions. Sensing of HIV protease activity by CARD8 leads to rapid pyroptosis of quiescent cells without productive infection, while T cell activation abolishes CARD8 function and increases permissiveness to infection. In humanized mice reconstituted with CARD8-deficient cells, CD4+ depletion is delayed despite high viremia. Finally, we discovered loss-of-function mutations in CARD8 from "natural hosts," which may explain the peculiarly non-pathogenic nature of these infections. Our study suggests that CARD8 drives CD4+ T cell depletion during pathogenic HIV/SIV infections.


Sujet(s)
Infections à VIH , Inflammasomes , Syndrome d'immunodéficience acquise du singe , Animaux , Humains , Souris , Protéines adaptatrices de signalisation CARD/génétique , Protéines adaptatrices de signalisation CARD/métabolisme , Lymphocytes T CD4+/métabolisme , Évolution de la maladie , Infections à VIH/anatomopathologie , Inflammasomes/métabolisme , Protéines tumorales/métabolisme , Syndrome d'immunodéficience acquise du singe/anatomopathologie , Virus de l'immunodéficience simienne/physiologie , Virémie , VIH (Virus de l'Immunodéficience Humaine)/physiologie
17.
J Med Virol ; 96(3): e29520, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38528837

RÉSUMÉ

The evolution of SARS-CoV-2 paired with immune imprinting by prototype messenger RNA (mRNA) vaccine has challenged the current vaccination efficacy against newly emerged Omicron subvariants. In our study, we investigated a cohort of macaques infected by SIV and vaccinated with two doses of bivalent Pfizer mRNA vaccine containing wildtype and BA.5 spikes. Using a pseudotyped lentivirus neutralization assay, we determined neutralizing antibody (nAb) titers against new XBB variants, i.e., XBB.1.5, XBB.1.16, and XBB.2.3, alongside D614G and BA.4/5. We found that compared to humans vaccinated with three doses of monovalent mRNA vaccine plus a bivalent booster, the monkeys vaccinated with two doses of bivalent mRNA vaccines exhibited relatively increased titers against XBB subvariants. Of note, SIV-positive dam macaques had reduced nAb titers relative to SIV-negative dams. Additionally, SIV positive dams that received antiretroviral therapy had lower nAb titers than untreated dams. Our study underscores the importance of reformulating the COVID-19 vaccine to better protect against newly emerged XBB subvariants as well as the need for further investigation of vaccine efficacy in individuals living with HIV-1.


Sujet(s)
COVID-19 , Vaccins à ARNm , Humains , Animaux , Macaca mulatta , Vaccins combinés , SARS-CoV-2/génétique , Vaccins contre la COVID-19 , COVID-19/prévention et contrôle , Vaccination , Anticorps neutralisants , ARN messager , Anticorps antiviraux
18.
Cell Rep ; 43(4): 113994, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38530856

RÉSUMÉ

Distinct dendritic cell (DC) subsets play important roles in shaping immune responses. Circulating DC precursors (pre-DCs) are more susceptible to HIV infection in vitro, which may explain the inefficiency of immune responses against HIV. However, the interplay between HIV and pre-DC is not defined in vivo. We identify human pre-DC equivalents in the cynomolgus macaque and then analyze their dynamics during simian immunodeficiency virus (SIV) infection to illustrate a sharp decrease of blood pre-DCs in early SIV infection and accumulation in lymph nodes (LNs), where they neglect to upregulate CD83/CD86 or MHC-II. Additionally, SIV infection attenuates the capacity of stimulated LN pre-DCs to produce IL-12p40. Analysis of HIV cohorts provides correlation between costimulatory molecule expression on pre-DCs and T cell activation in spontaneous HIV controllers. These findings pinpoint certain dynamics and functional changes of pre-DCs during SIV infection, providing a deeper understanding of immune dysregulation mechanisms elicited in people living with HIV.


Sujet(s)
Cellules dendritiques , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Syndrome d'immunodéficience acquise du singe/sang , Syndrome d'immunodéficience acquise du singe/anatomopathologie , Cellules dendritiques/immunologie , Virus de l'immunodéficience simienne/immunologie , Humains , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/sang , Infections à VIH/anatomopathologie , Macaca fascicularis , Activation des lymphocytes/immunologie
19.
Curr HIV/AIDS Rep ; 21(2): 62-74, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38411842

RÉSUMÉ

PURPOSE OF REVIEW: HIV reservoirs are the main barrier to cure. CD4+ T cells have been extensively studied as the primary HIV-1 reservoir. However, there is substantial evidence that HIV-1-infected myeloid cells (monocytes/macrophages) also contribute to viral persistence and pathogenesis. RECENT FINDINGS: Recent studies in animal models and people with HIV-1 demonstrate that myeloid cells are cellular reservoirs of HIV-1. HIV-1 genomes and viral RNA have been reported in circulating monocytes and tissue-resident macrophages from the brain, urethra, gut, liver, and spleen. Importantly, viral outgrowth assays have quantified persistent infectious virus from monocyte-derived macrophages and tissue-resident macrophages. The myeloid cell compartment represents an important target of HIV-1 infection. While myeloid reservoirs may be more difficult to measure than CD4+ T cell reservoirs, they are long-lived, contribute to viral persistence, and, unless specifically targeted, will prevent an HIV-1 cure.


Sujet(s)
Infections à VIH , Séropositivité VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Humains , Infections à VIH/anatomopathologie , Syndrome d'immunodéficience acquise du singe/anatomopathologie , Macrophages , Lymphocytes T CD4+ , Latence virale , Charge virale
20.
Viruses ; 16(2)2024 02 15.
Article de Anglais | MEDLINE | ID: mdl-38400071

RÉSUMÉ

Metabolic-dysfunction-associated fatty liver disease (MAFLD) is a major cause of morbidity and mortality in HIV-infected individuals, even those receiving optimal antiretroviral therapy. Here, we utilized the SIV rhesus macaque model and advanced laparoscopic techniques for longitudinal collection of liver tissue to elucidate the timing of pathologic changes. The livers of both SIV-infected (N = 9) and SIV-naïve uninfected (N = 8) macaques were biopsied and evaluated at four time points (weeks -4, 2, 6, and 16-20 post-infection) and at necropsy (week 32). SIV DNA within the macaques' livers varied by over 4 logs at necropsy, and liver SIV DNA significantly correlated with SIV RNA in the plasma throughout the study. Acute phase liver pathology (2 weeks post-infection) was characterized by evidence for fat accumulation (microvesicular steatosis), a transient elevation in both AST and cholesterol levels within the serum, and increased hepatic expression of the PPARA gene associated with cholesterol metabolism and beta oxidation. By contrast, the chronic phase of the SIV infection (32 weeks post-infection) was associated with sinusoidal dilatation, while steatosis resolved and concentrations of AST and cholesterol remained similar to those in uninfected macaques. These findings suggest differential liver pathologies associated with the acute and chronic phases of infection and the possibility that therapeutic interventions targeting metabolic function may benefit liver health in people newly diagnosed with HIV.


Sujet(s)
Stéatose hépatique , Infections à VIH , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Humains , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Macaca mulatta , Infections à VIH/complications , Hépatocytes/métabolisme , ADN , Cholestérol
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE