Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.631
Filtrer
1.
J Acquir Immune Defic Syndr ; 97(2): 156-164, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39250649

RÉSUMÉ

BACKGROUND: Cocaine-one of the most frequently abused illicit drugs among persons living with HIV [people living with HIV (PLWH)]-slows the decline of viral production after antiretroviral therapy and is associated with higher HIV viral load, more rapid HIV progression, and increased mortality. SETTING: We examined the impact of cocaine use on the CD4+ T-cell HIV latent reservoir (HLR) in virally suppressed PLWH participating in a national, longitudinal cohort study of the natural and treated history of HIV in the United States. METHODS: CD4+ T-cell genomic DNA from 434 women of diverse ancestry (ie, 75% Black, 14% Hispanic, 12% White) who self-reported cocaine use (ie, 160 cocaine users, 59 prior users, 215 non-users) was analyzed using the Intact Proviral HIV DNA Assay, measuring intact provirus per 106 CD4+ T cells. FINDINGS: HIV latent reservoir size differed by cocaine use (ie, median [interquartile range]: 72 [14-193] for never users, 165 [63-387] for prior users, 184 [28-502] for current users), which was statistically significantly larger in both prior (P = 0.023) and current (P = 0.001) cocaine users compared with never users. CONCLUSIONS: Cocaine use may contribute to a larger replication competent HLR in CD4+ T cells among virologically suppressed women living with HIV. Our findings are important because women are underrepresented in HIV reservoir studies and in studies of the impact of cocaine use on outcomes among PLWH.


Sujet(s)
Lymphocytes T CD4+ , Troubles liés à la cocaïne , Infections à VIH , Charge virale , Latence virale , Humains , Femelle , Infections à VIH/traitement médicamenteux , Adulte , Adulte d'âge moyen , Troubles liés à la cocaïne/épidémiologie , Études longitudinales , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , États-Unis/épidémiologie , ADN viral , Cocaïne
3.
Signal Transduct Target Ther ; 9(1): 231, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39245675

RÉSUMÉ

The combination of ASC22, an anti-PD-L1 antibody potentially enhancing HIV-specific immunity and chidamide, a HIV latency reversal agent, may serve as a strategy for antiretroviral therapy-free virological control for HIV. People living with HIV, having achieved virological suppression, were enrolled to receive ASC22 and chidamide treatment in addition to their antiretroviral therapy. Participants were monitored over 24 weeks to measure changes in viral dynamics and the function of HIV-specific CD8+ T cells (NCT05129189). 15 participants completed the study. At week 8, CA HIV RNA levels showed a significant increase from baseline, and the values returned to baseline after discontinuing ASC22 and chidamide. The total HIV DNA was only transiently increased at week 4 (P = 0.014). In contrast, integrated HIV DNA did not significantly differ from baseline. Increases in the proportions of effector memory CD4+ and CD8+ T cells (TEM) were observed from baseline to week 24 (P = 0.034 and P = 0.002, respectively). The combination treatment did not succeed in enhancing the function of HIV Gag/Pol- specific CD8+ T cells. Nevertheless, at week 8, a negative correlation was identified between the proportions of HIV Gag-specific TEM cells and alterations in integrated DNA in the T cell function improved group (P = 0.042 and P = 0.034, respectively). Nine adverse events were solicited, all of which were graded 1 and resolved spontaneously. The combined treatment of ASC22 and chidamide was demonstrated to be well-tolerated and effective in activating latent HIV reservoirs. Further investigations are warranted in the context of analytic treatment interruption.


Sujet(s)
Aminopyridines , Benzamides , Lymphocytes T CD8+ , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Inhibiteurs de désacétylase d'histone , Humains , Mâle , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Infections à VIH/immunologie , Infections à VIH/génétique , Aminopyridines/pharmacologie , Femelle , Adulte , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Benzamides/pharmacologie , Benzamides/usage thérapeutique , Benzamides/administration et posologie , Adulte d'âge moyen , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Antigène CD274/immunologie , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/génétique , Latence virale/effets des médicaments et des substances chimiques , Charge virale/effets des médicaments et des substances chimiques
4.
Viruses ; 16(9)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39339855

RÉSUMÉ

CD80 is the best-known costimulatory molecule for effective T cell functions. Many different reports have summarized the role of CD80 in HSV-1 and its functions in maintaining adaptive immunity, which is the main player in causing herpes stromal keratitis (HSK). To determine the effects of absence or overexpression of CD80 in HSV-1 infection, we infected CD80-/- and WT mice with a recombinant HSV-1 expressing murine CD80 (HSV-CD80) in place of the latency associated transcript (LAT). Parental dLAT2903 virus lacking LAT was used as a control. After infection, critical components of infection like virus replication, eye disease, early cellular infiltrates into the corneas and trigeminal ganglia (TG), latency-reactivation in the infected mice were determined. Our findings reveal that the absence of CD80 in the CD80-/- mice infected with both viruses did not affect the viral titers in the mice eyes or eye disease, but it played a significant role in critical components of HSV-induced immunopathology. The WT mice infected with dLAT2903 virus had significantly higher levels of latency compared with the CD80-/- mice infected with dLAT2903 virus, while levels of latency as determined by gB DNA expression were similar between the WT and CD80-/- mice infected with HSV-CD80 virus. In contrast to the differences in the levels of latency between the infected groups, the absence of CD80 expression in the CD80-/- mice or its overexpression by HSV-CD80 virus did not have any effect on the time of reactivation. Furthermore, the absence of CD80 expression contributed to more inflammation in the CD80-/--infected mice. Overall, this study suggests that in the absence of CD80, inflammation increases, latency is reduced, but reactivation is not affected. Altogether, our study suggests that reduced latency correlated with reduced levels of inflammatory molecules and blocking or reducing expression of CD80 could be used to mitigate the immune responses, therefore controlling HSV-induced infection.


Sujet(s)
Antigène CD80 , Cornée , Herpèsvirus humain de type 1 , Kératite herpétique , Souris knockout , Ganglion trigéminal , Latence virale , Animaux , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/physiologie , Herpèsvirus humain de type 1/génétique , Souris , Antigène CD80/génétique , Antigène CD80/métabolisme , Cornée/virologie , Cornée/anatomopathologie , Cornée/immunologie , Kératite herpétique/virologie , Kératite herpétique/immunologie , Kératite herpétique/anatomopathologie , Ganglion trigéminal/virologie , Ganglion trigéminal/immunologie , Réplication virale , Souris de lignée C57BL , Femelle , Activation virale , Modèles animaux de maladie humaine , Herpès/immunologie , Herpès/virologie , microARN
5.
Viruses ; 16(9)2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39339894

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) is a cancer-causing virus that establishes life-long infection. KSHV is implicated in the etiology of Kaposi's sarcoma, and a number of rare hematopoietic malignancies. The present study focuses on the KSHV open reading frame 20 (ORF20), a member of the conserved herpesvirus UL24 protein family containing five conserved homology domains and a conserved PD-(D/E)XK putative endonuclease motif, whose nuclease function has not been established to date. ORF20 encodes three co-linear protein isoforms, full length, intermediate, and short, though their differential functions are unknown. In an effort to determine the role of ORF20 during KSHV infection, we generated a recombinant ORF20-Null KSHV genome, which fails to express all three ORF20 isoforms. This genome was reconstituted in iSLK cells to establish a latent infection, which resulted in an accelerated transcription of viral mRNAs, an earlier accumulation of viral lytic proteins, an increase in the quantity of viral DNA copies, and a significant decrease in viral yield upon lytic reactivation. This was accompanied by early cell death of cells infected with the ORF20-Null virus. Functional complementation of the ORF20-Null mutant with the short ORF20 isoform rescued KSHV production, whereas its endonuclease mutant form failed to enhance lytic reactivation. Complementation with the short isoform further revealed a decrease in cell death as compared with ORF20-Null virus. Finally, expression of IL6 and CXCL8, previously shown to be affected by the hCMV UL24 homolog, was relatively low upon reactivation of cells infected with the ORF20-Null virus. These findings suggest that ORF20 protein, with its putative endonuclease motif, promotes coordinated lytic reactivation for increased infectious particle production.


Sujet(s)
Herpèsvirus humain de type 8 , Cadres ouverts de lecture , Protéines virales , Activation virale , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/physiologie , Humains , Protéines virales/génétique , Protéines virales/métabolisme , Lignée cellulaire , Virion/métabolisme , Virion/génétique , Réplication virale , Latence virale/génétique , ADN viral/génétique , Régulation de l'expression des gènes viraux , Génome viral
6.
Viruses ; 16(9)2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39339948

RÉSUMÉ

Equid alphaherpesvirus 1 (EHV-1) has been linked to the emergence of neurological disorders, with the horse racing industry experiencing significant impacts from outbreaks of equine herpesvirus myeloencephalopathy (EHM). Building robust immune memory before pathogen exposure enables rapid recognition and elimination, preventing infection. This is crucial for effectively managing EHV-1. Removing neuropathogenic factors and immune evasion genes to develop live attenuated vaccines appears to be a successful strategy for EHV-1 vaccines. We created mutant viruses without ORF38 and ORF37/38 and validated their neuropathogenicity and immunogenicity in hamsters. The ∆ORF38 strain caused brain tissue damage at high doses, whereas the ∆ORF37/38 strain did not. Dexamethasone was used to confirm latent herpesvirus infection and reactivation. Dexamethasone injection increased viral DNA load in the brains of hamsters infected with the parental and ∆ORF38 strains, but not in those infected with the ∆ORF37/38 strain. Immunizing hamsters intranasally with the ∆ORF37/38 strain as a live vaccine produced a stronger immune response compared to the ∆ORF38 strain at the same dose. The hamsters demonstrated effective protection against a lethal challenge with the parental strain. This suggests that the deletion of ORF37/38 may effectively inhibit latent viral infection, reduce the neuropathogenicity of EHV-1, and induce a protective immune response.


Sujet(s)
Infections à Herpesviridae , Herpèsvirus équin de type 1 , Vaccins atténués , Animaux , Herpèsvirus équin de type 1/génétique , Herpèsvirus équin de type 1/immunologie , Herpèsvirus équin de type 1/pathogénicité , Infections à Herpesviridae/prévention et contrôle , Infections à Herpesviridae/virologie , Infections à Herpesviridae/immunologie , Cricetinae , Vaccins atténués/immunologie , Vaccins atténués/génétique , Vaccins atténués/administration et posologie , Protéines virales/génétique , Protéines virales/immunologie , Infection latente/immunologie , Infection latente/virologie , Equus caballus , Encéphale/virologie , Encéphale/anatomopathologie , Latence virale , Mesocricetus , Délétion de séquence , Cadres ouverts de lecture , Femelle , Maladies des chevaux/virologie , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/immunologie , Charge virale
7.
Viruses ; 16(9)2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39339960

RÉSUMÉ

More than 80 million people worldwide have been infected with the human immunodeficiency virus (HIV). There are now approximately 39 million individuals living with HIV/acquired immunodeficiency syndrome (AIDS). Although treatments against HIV infection are available, AIDS remains a serious disease. Combination antiretroviral therapy (cART), also known as highly active antiretroviral therapy (HAART), consists of treatment with a combination of several antiretroviral drugs that block multiple stages in the virus replication cycle. However, the increasing usage of cART is inevitably associated with the emergence of HIV drug resistance. In addition, the development of persistent cellular reservoirs of latent HIV is a critical obstacle to viral eradication since viral rebound takes place once anti-retroviral therapy (ART) is interrupted. Thus, several efforts are being applied to new generations of drugs, vaccines and new types of cART. In this review, we summarize the antiviral therapies used for the treatment of HIV/AIDS, both as individual agents and as combination therapies, and highlight the role of both macrophages and HIV cellular reservoirs and the most recent clinical studies related to this disease.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Macrophages , Latence virale , Humains , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Macrophages/virologie , Macrophages/effets des médicaments et des substances chimiques , Latence virale/effets des médicaments et des substances chimiques , 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 , Agents antiVIH/usage thérapeutique , Agents antiVIH/pharmacologie , Thérapie antirétrovirale hautement active , Réservoirs de maladies/virologie , Réplication virale/effets des médicaments et des substances chimiques , Animaux
8.
Viruses ; 16(9)2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39339966

RÉSUMÉ

Primary Effusion Lymphoma (PEL) cells carry Kaposi's sarcoma-associated herpesvirus (KSHV) in a latent state, except for a small number of cells in which the virus replicates to ensure its persistence into the infected host. However, the lytic cycle can be reactivated in vitro by exposing these lymphoma cells to various treatments, leading to cell lysis. To restrict viral antigen expression, KSHV induces repressive epigenetic changes, including DNA methylation and histone modifications. Among the latter, histone deacetylation and tri-methylation of Histone H3 lisyne-27 (H3K27me3) have been reported to play a role. Here, we found that the inhibition of H3K27 tri-methylation by valemetostat DS3201 (DS), a small molecule that inhibits Enhancer of Zeste Homolog 2 (EZH2) methyltransferase, induced the KSHV lytic cycle in PEL cells, and that this effect involved the activation of the wtp53-p21 axis and autophagic dysregulation. DS also potentiated the lytic cycle activation mediated by the Histone deacetylases (HDAC) inhibitor Suberoylanilide hydroxamic acid (SAHA) and reinforced its cytotoxic effect, suggesting that such a combination could be used to unbalance the latent/lytic cycle and further impair the survival of PEL cells.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste , Herpèsvirus humain de type 8 , Inhibiteurs de désacétylase d'histone , Lymphome primitif des séreuses , Vorinostat , Herpèsvirus humain de type 8/effets des médicaments et des substances chimiques , Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/génétique , Humains , Lymphome primitif des séreuses/virologie , Lymphome primitif des séreuses/traitement médicamenteux , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Vorinostat/pharmacologie , Lignée cellulaire tumorale , Latence virale/effets des médicaments et des substances chimiques , Activation virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Histone/métabolisme
9.
Viruses ; 16(9)2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39339973

RÉSUMÉ

Bovine herpesvirus type 1 (BoHV-1) establishes lifelong latency in trigeminal ganglionic (TG) neurons following intranasal and ocular infection in cattle. Periodically, the latent virus reactivates in the TG due to stress and is transported anterogradely to nerve endings in the nasal epithelium, where the virus replicates and sheds. Consequently, BoHV-1 is transmitted to susceptible animals and maintained in the cattle population. Modified live BoHV-1 vaccine strains (BoHV-1 MLV) also have a similar latency reactivation. Therefore, they circulate and are maintained in cattle herds. Additionally, they can regain virulence and cause vaccine outbreaks because they mutate and recombine with other circulating field wild-type (wt) strains. Recently, we constructed a BoHV-1 quadruple mutant virus (BoHV-1qmv) that lacks immune evasive properties due to UL49.5 and glycoprotein G (gG) deletions. In addition, it also lacks the gE cytoplasmic tail (gE CT) and Us9 gene sequences designed to make it safe, increase its vaccine efficacy against BoHV-1, and restrict its anterograde neuronal transport noted above. Further, we engineered the BoHV-1qmv-vector to serve as a subunit vaccine against the Rift Valley fever virus (BoHV-1qmv Sub-RVFV) (doi: 10.3390/v15112183). In this study, we determined the latency reactivation and nasal virus shedding properties of BoHV-1qmv (vector) and BoHV-1qmv-vectored subunit RVFV (BoHV-1qmv sub-RVFV) vaccine virus in calves in comparison to the BoHV-1 wild-type (wt) following intranasal inoculation. The real-time PCR results showed that BoHV-1 wt- but not the BoHV-1qmv vector- and BoHV-1qmv Sub-RVFV-inoculated calves shed virus in the nose following dexamethasone-induced latency reactivation; however, like the BoHV-1 wt, both the BoHV-1qmv vector and BoHV-1qmv Sub-RVFV viruses established latency, were reactivated, and replicated in the TG neurons. These results are consistent with the anterograde neurotransport function of the gE CT and Us9 sequences, which are deleted in the BoHV-1qmv and BoHV-1qmv Sub-RVFV.


Sujet(s)
Herpèsvirus bovin de type 1 , Muqueuse nasale , Neurones , Ganglion trigéminal , Activation virale , Latence virale , Excrétion virale , Animaux , Herpèsvirus bovin de type 1/génétique , Herpèsvirus bovin de type 1/physiologie , Herpèsvirus bovin de type 1/immunologie , Bovins , Muqueuse nasale/virologie , Ganglion trigéminal/virologie , Neurones/virologie , Délétion de gène , Vaccins atténués/immunologie , Vaccins atténués/génétique , Vaccins atténués/administration et posologie , Réplication virale , Maladies des bovins/virologie , Maladies des bovins/prévention et contrôle , Maladies des bovins/immunologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/génétique , Infections à Herpesviridae/médecine vétérinaire , Infections à Herpesviridae/virologie , Infections à Herpesviridae/prévention et contrôle , Infections à Herpesviridae/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Vecteurs génétiques/génétique , Rhinotrachéite infectieuse bovine/virologie , Rhinotrachéite infectieuse bovine/prévention et contrôle , Rhinotrachéite infectieuse bovine/immunologie , Vaccins contre les herpèsvirus/génétique , Vaccins contre les herpèsvirus/immunologie
10.
PLoS Pathog ; 20(9): e1012525, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39241017

RÉSUMÉ

Epstein-Barr virus (EBV) uses latency programs to colonize the memory B-cell reservoir, and each program is associated with human malignancies. However, knowledge remains incomplete of epigenetic mechanisms that maintain the highly restricted latency I program, present in memory and Burkitt lymphoma cells, in which EBNA1 is the only EBV-encoded protein expressed. Given increasing appreciation that higher order chromatin architecture is an important determinant of viral and host gene expression, we investigated roles of Wings Apart-Like Protein Homolog (WAPL), a host factor that unloads cohesin to control DNA loop size and that was discovered as an EBNA2-associated protein. WAPL knockout (KO) in Burkitt cells de-repressed LMP1 and LMP2A expression, but not other EBV oncogenes, to yield a viral program reminiscent of EBV latency II, which is rarely observed in B-cells. WAPL KO also increased LMP1/2A levels in latency III lymphoblastoid cells. WAPL KO altered EBV genome architecture, triggering formation of DNA loops between the LMP promoter region and the EBV origins of lytic replication (oriLyt). Hi-C analysis further demonstrated that WAPL KO reprogrammed EBV genomic DNA looping. LMP1 and LMP2A de-repression correlated with decreased histone repressive marks at their promoters. We propose that EBV coopts WAPL to negatively regulate latent membrane protein expression to maintain Burkitt latency I.


Sujet(s)
Infections à virus Epstein-Barr , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 4 , Protéines de la matrice virale , Latence virale , Humains , Herpèsvirus humain de type 4/génétique , Latence virale/physiologie , Protéines de la matrice virale/métabolisme , Protéines de la matrice virale/génétique , Infections à virus Epstein-Barr/virologie , Infections à virus Epstein-Barr/métabolisme , Infections à virus Epstein-Barr/génétique , Lymphocytes B/virologie , Lymphocytes B/métabolisme , Lymphome de Burkitt/virologie , Lymphome de Burkitt/génétique , Lymphome de Burkitt/métabolisme , Lignée cellulaire tumorale
11.
PLoS Pathog ; 20(9): e1012083, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39259751

RÉSUMÉ

The persistence of HIV-1 in long-lived latent reservoirs during suppressive antiretroviral therapy (ART) remains one of the principal barriers to a functional cure. Blocks to transcriptional elongation play a central role in maintaining the latent state, and several latency reversal strategies focus on the release of positive transcription elongation factor b (P-TEFb) from sequestration by negative regulatory complexes, such as the 7SK complex and BRD4. Another major cellular reservoir of P-TEFb is in Super Elongation Complexes (SECs), which play broad regulatory roles in host gene expression. Still, it is unknown if the release of P-TEFb from SECs is a viable latency reversal strategy. Here, we demonstrate that the SEC is not required for HIV-1 replication in primary CD4+ T cells and that a small molecular inhibitor of the P-TEFb/SEC interaction (termed KL-2) increases viral transcription. KL-2 acts synergistically with other latency reversing agents (LRAs) to reactivate viral transcription in several cell line models of latency in a manner that is, at least in part, dependent on the viral Tat protein. Finally, we demonstrate that KL-2 enhances viral reactivation in peripheral blood mononuclear cells (PBMCs) from people living with HIV (PLWH) on suppressive ART, most notably in combination with inhibitor of apoptosis protein antagonists (IAPi). Taken together, these results suggest that the release of P-TEFb from cellular SECs may be a novel route for HIV-1 latency reactivation.


Sujet(s)
Lymphocytes T CD4+ , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Facteur B d'élongation transcriptionnelle positive , Latence virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Latence virale/physiologie , Latence virale/effets des médicaments et des substances chimiques , Facteur B d'élongation transcriptionnelle positive/métabolisme , Infections à VIH/virologie , Infections à VIH/métabolisme , Infections à VIH/traitement médicamenteux , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/métabolisme , Activation virale/effets des médicaments et des substances chimiques , Réplication virale , Régulation de l'expression des gènes viraux
13.
Viruses ; 16(8)2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39205284

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) might impact disease progression in people living with HIV (PLWH), including those on effective combination antiretroviral therapy (cART). These individuals often experience chronic conditions characterized by proviral latency or low-level viral replication in CD4+ memory T cells and tissue macrophages. Pro-inflammatory cytokines, such as TNF-α, IL-1ß, IL-6, and IFN-γ, can reactivate provirus expression in both primary cells and cell lines. These cytokines are often elevated in individuals infected with SARS-CoV-2, the virus causing COVID-19. However, it is still unknown whether SARS-CoV-2 can modulate HIV reactivation in infected cells. Here, we report that exposure of the chronically HIV-1-infected myeloid cell line U1 to two different SARS-CoV-2 viral isolates (ancestral and BA.5) reversed its latent state after 24 h. We also observed that SARS-CoV-2 exposure of human primary monocyte-derived macrophages (MDM) initially drove their polarization towards an M1 phenotype, which shifted towards M2 over time. This effect was associated with soluble factors released during the initial M1 polarization phase that reactivated HIV production in U1 cells, like MDM stimulated with the TLR agonist resiquimod. Our study suggests that SARS-CoV-2-induced systemic inflammation and interaction with macrophages could influence proviral HIV-1 latency in myeloid cells in PLWH.


Sujet(s)
COVID-19 , Cytokines , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Macrophages , Cellules myéloïdes , SARS-CoV-2 , Latence virale , Humains , SARS-CoV-2/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , COVID-19/virologie , COVID-19/immunologie , Macrophages/virologie , Macrophages/immunologie , Cellules myéloïdes/virologie , Cytokines/métabolisme , Infections à VIH/virologie , Infections à VIH/immunologie , Infections à VIH/traitement médicamenteux , Lignée cellulaire , Effet bystander , Activation virale , Réplication virale/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/immunologie
14.
Microb Pathog ; 195: 106896, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39208957

RÉSUMÉ

BACKGROUND: Bovine herpesvirus 1 (BoHV-1) is a major pathogen that affects the global bovine population, primarily inducing respiratory and reproductive disorders. Its ability to establish latent infections in neuronal cells and to reactivate under certain conditions poses a continual threat to uninfected hosts. In this study, we aimed to analyze the replication characteristics of BoHV-1 in neuronal cells, as well as the effects of viral replication on host cell immunity and physiology. METHODS: Using the Neuro-2a neuronal-origin cell line as a model, we explored the dynamics of BoHV-1 replication and analyzed differential gene expression profiles post-BoHV-1 infection using high-throughput RNA sequencing. RESULTS: BoHV-1 demonstrated restricted replication in Neuro-2a cells. BoHV-1 induced apoptotic pathways and enhanced the transcription of interferon-stimulated genes and interferon regulatory factors while suppressing the complement cascade in Neuro-2a cells. CONCLUSIONS: Different from BoHV-1 infection in other non-highly differentiated somatic cells result in viral dominance, BoHV-1 regulated the innate immune response in neuronal cells formed a "virus-nerve cell" relative equilibrium state, which may account for the restricted replication of BoHV-1 in neuronal cells, leading to a latent infection. These findings provide a foundation for further research into the mechanism underlying BoHV-1-induced latent infection in nerve cells.


Sujet(s)
Analyse de profil d'expression de gènes , Herpèsvirus bovin de type 1 , Immunité innée , Neurones , Réplication virale , Herpèsvirus bovin de type 1/immunologie , Herpèsvirus bovin de type 1/génétique , Herpèsvirus bovin de type 1/physiologie , Animaux , Bovins , Neurones/virologie , Neurones/immunologie , Lignée cellulaire , Souris , Infections à Herpesviridae/virologie , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/médecine vétérinaire , Apoptose , Transcriptome , Latence virale , Interactions hôte-pathogène/immunologie , Maladies des bovins/virologie , Maladies des bovins/immunologie , Facteurs de régulation d'interféron/génétique , Facteurs de régulation d'interféron/métabolisme , Séquençage nucléotidique à haut débit
15.
J Immunol ; 213(7): 965-970, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39150241

RÉSUMÉ

CMV drives the accumulation of virus-specific, highly differentiated CD8 memory T cells (memory inflation [MI]). In mice, MI was shown to directly correlate with the CMV infection dose, yet the CMV-associated CD8 MI plateaus over time. It is unclear how MI is regulated with aging. We infected young mice with 102, 104, and 106 PFU of murine CMV and confirmed that MI magnitude was directly proportional to the infectious dose, reaching a setpoint by midlife. By old age, MI subsided, most prominently in mice infected with 106 PFU, and reached statistical parity between groups in 26-mo-old mice. This corresponded to an age-related loss in lymphatic endothelial cells in lymph nodes, recently shown to be sufficient to drive MI in mice. We propose that MI size and persistence over the lifespan is controlled by the size of the lymphatic endothelial cell niche, whose shrinking leads to reduced MI with aging.


Sujet(s)
Lymphocytes T CD8+ , Mémoire immunologique , Muromegalovirus , Latence virale , Animaux , Souris , Lymphocytes T CD8+/immunologie , Latence virale/immunologie , Mémoire immunologique/immunologie , Muromegalovirus/immunologie , Vieillissement/immunologie , Souris de lignée C57BL , Cellules T mémoire/immunologie , Infections à cytomégalovirus/immunologie , Infections à Herpesviridae/immunologie , Cellules endothéliales/immunologie , Cellules endothéliales/virologie , Cytomegalovirus/immunologie , Cytomegalovirus/physiologie , Noeuds lymphatiques/immunologie
16.
mBio ; 15(9): e0163224, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39136440

RÉSUMÉ

The HIV reservoir is more dynamic than previously thought with around 70% of the latent reservoir originating from viruses circulating within 1 year of the initiation of antiretroviral therapy (ART). In an ex vivo model system of HIV latency, it was reported that early exposure to class I histone deacetylase (HDAC) inhibitors might prevent these more recently infected cells from entering a state of stable viral latency. This finding raises the possibility that co-administration of HDAC inhibitors at the time of ART initiation may prevent the establishment of much of the HIV reservoir. Here, we tested the effects of the HDAC inhibitors suberoylanilide hydroxamic acid (SAHA) and panobinostat co-administered at the time of ART initiation on the formation of the viral reservoir in HIV-infected humanized mice. As previously shown, SAHA and panobinostat were well tolerated in humanized mice. Unexpectedly, co-administration of SAHA resulted in an increase in the frequency of CD4+ cells carrying HIV DNA but did not alter the frequency of cell-associated HIV RNA in HIV-infected, ART-treated humanized mice. Co-administration of panobinostat did not alter levels of cell-associated HIV DNA or RNA. Our in vivo findings indicate that co-administration of HDAC inhibitors initiated at the same time of ART treatment does not prevent recently infected cells from entering latency.IMPORTANCECurrent antiretroviral therapy (ART) does not eradicate cells harboring replication-competent HIV reservoir. Withdrawal of ART inevitably results in a rapid viremia rebound. The HIV reservoir is more dynamic than previously thought. Early exposure to class I histone deacetylase (HDAC) inhibitors inhibit these more recently infected cells from entering a state of stable viral latency in an ex vivo model of latency, raising the possibility that co-administration of HDAC inhibitors at the time of ART initiation may reduce much of the HIV reservoir. Here, we tested the effects of the HDAC inhibitors suberoylanilide hydroxamic acid or panobinostat during ART initiation on the formation of the viral reservoir in HIV-infected humanized mice. Our in vivo study indicates that in contrast to in vitro observations, the co-administration of HDAC inhibitors at the same time of ART initiation does not prevent recently infected cells from entering latency.


Sujet(s)
Lymphocytes T CD4+ , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Inhibiteurs de désacétylase d'histone , Panobinostat , Latence virale , Vorinostat , Inhibiteurs de désacétylase d'histone/pharmacologie , Animaux , Latence virale/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Souris , Panobinostat/pharmacologie , Humains , Vorinostat/pharmacologie , 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 , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Acides hydroxamiques/pharmacologie , Modèles animaux de maladie humaine , Charge virale/effets des médicaments et des substances chimiques , ARN viral , ADN viral
17.
JCI Insight ; 9(18)2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115957

RÉSUMÉ

Nonreceptor tyrosine phosphatases (NTPs) play an important role in regulating protein phosphorylation and have been proposed as attractive therapeutic targets for cancer and metabolic diseases. We have previously identified that 3-Hydroxy-1,2,3-benzotriazin-4(3H)-one (HODHBt) enhanced STAT activation upon cytokine stimulation, leading to increased reactivation of latent HIV and effector functions of NK and CD8 T cells. Here, we demonstrate that HODHBt interacted with and inhibited the NTPs PTPN1 and PTPN2 through a mixed inhibition mechanism. We also confirm that PTPN1 and PTPN2 specifically controlled the phosphorylation of different STATs. The small molecule ABBV-CLS-484 (AC-484) is an active site inhibitor of PTPN1 and PTPN2 currently in clinical trials for advanced solid tumors. We compared AC-484 and HODHBt and found similar effects on STAT5 and immune activation, albeit with different mechanisms of action leading to varying effects on latency reversal. Our studies provide the first specific evidence to our knowledge that enhancing STAT phosphorylation via inhibition of PTPN1 and PTPN2 is an effective tool against HIV.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Latence virale , Protein Tyrosine Phosphatase, Non-Receptor Type 2/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 2/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 1/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonistes et inhibiteurs , Humains , Latence virale/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Triazines
18.
J Virol ; 98(9): e0060424, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39194241

RÉSUMÉ

Viruses normally reprogram the host cell metabolic pathways as well as metabolic sensors to facilitate their persistence. The serine-threonine liver kinase B1 (LKB1) is a master upstream kinase of 5'-AMP-activated protein kinase (AMPK) that senses the energy status and therefore regulates the intracellular metabolic homeostasis. Previous studies showed that AMPK restricts Kaposi's sarcoma-associated herpesvirus (KSHV) lytic replication in endothelial cells during primary infection and promotes primary effusion lymphoma (PEL) cell survival. However, the role of LKB1 in KSHV lytic reactivation and KSHV-associated malignancies is unclear. In this study, we found that LKB1 is phosphorylated or activated in KSHV-positive PEL cells. Mechanistically, KSHV-encoded vCyclin mediated LKB1 activation in PEL cells, as vCyclin knockout ablated, while vCyclin overexpression enhanced LKB1 activation. Furthermore, knockdown of LKB1 inactivated AMPK and induced KSHV reactivation, as indicated by the increased expression of viral lytic genes and the increased virions in supernatants. Accordingly, AMPK inhibition by functional knockdown or a pharmacologic inhibitor, Compound C, promoted KSHV reactivation in PEL cells. Furthermore, inhibition of either LKB1 or AMPKα1 efficiently induced cell death by apoptosis of PEL cells both in vitro and in vivo. Together, these results identify LKB1 as a vulnerable target for PEL, which could be potentially exploited for treating other virus-associated diseases.IMPORTANCEKaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic virus associated with several human cancers, such as primary effusion lymphoma (PEL). Here, we showed that serine-threonine liver kinase B1 (LKB1), upstream of 5' AMP-activated protein kinase (AMPK), is activated by KSHV-encoded vCyclin and maintains KSHV latency in PEL cells. Inhibition of either LKB1 or AMPK enhances KSHV lytic replication from latency, which at least partially accounts for PEL cell death by apoptosis. Compound C, a potent AMPK inhibitor, induced KSHV reactivation and efficiently inhibited PEL progression in vivo. Thus, our work revealed that LKB1 is a potential therapeutic target for KSHV-associated cancers.


Sujet(s)
AMP-activated protein kinase kinases , AMP-Activated Protein Kinases , Herpèsvirus humain de type 8 , Lymphome primitif des séreuses , Protein-Serine-Threonine Kinases , Activation virale , Herpèsvirus humain de type 8/physiologie , Lymphome primitif des séreuses/virologie , Lymphome primitif des séreuses/métabolisme , Lymphome primitif des séreuses/anatomopathologie , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Animaux , AMP-Activated Protein Kinases/métabolisme , AMP-Activated Protein Kinases/génétique , Souris , Lignée cellulaire tumorale , Apoptose , Réplication virale , Latence virale , Évolution de la maladie , Phosphorylation
19.
J Exp Med ; 221(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39141127

RÉSUMÉ

HIV-1 antiretroviral therapy is highly effective but fails to eliminate a reservoir of latent proviruses, leading to a requirement for life-long treatment. How the site of integration of authentic intact latent proviruses might impact their own or neighboring gene expression or reservoir dynamics is poorly understood. Here, we report on proviral and neighboring gene transcription at sites of intact latent HIV-1 integration in cultured T cells obtained directly from people living with HIV, as well as engineered primary T cells and cell lines. Proviral gene expression was correlated to the level of endogenous gene expression under resting but not activated conditions. Notably, latent proviral promoters were 100-10,000× less active than in productively infected cells and had little or no measurable impact on neighboring gene expression under resting or activated conditions. Thus, the site of integration has a dominant effect on the transcriptional activity of intact HIV-1 proviruses in the latent reservoir, thereby influencing cytopathic effects and proviral immune evasion.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Provirus , Transcription génétique , Intégration virale , Latence virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Provirus/génétique , Latence virale/génétique , Intégration virale/génétique , Infections à VIH/virologie , Infections à VIH/génétique , Régulation de l'expression des gènes viraux , Régions promotrices (génétique)/génétique , Lymphocytes T CD4+/virologie , Lymphocytes T/virologie , Lymphocytes T/immunologie , Lignée cellulaire
20.
PLoS Pathog ; 20(8): e1012081, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39186813

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) establishes persistent infection in the host by encoding a vast network of proteins that aid immune evasion. One of these targeted innate immunity pathways is the cGAS-STING pathway, which inhibits the reactivation of KSHV from latency. Previously, we identified multiple cGAS/STING inhibitors encoded by KSHV, suggesting that the counteractions of this pathway by viral proteins are critical for maintaining a successful KSHV life cycle. However, the detailed mechanisms of how these viral proteins block innate immunity and facilitate KSHV lytic replication remain largely unknown. In this study, we report that ORF48, a previously identified negative regulator of the cGAS/STING pathway, is required for optimal KSHV lytic replication. We used both siRNA and deletion-based systems to evaluate the importance of intact ORF48 in the KSHV lytic cycle. In both systems, loss of ORF48 resulted in defects in lytic gene transcription, lytic protein expression, viral genome replication and infectious virion production. ORF48 genome deletion caused more robust and global repression of the KSHV transcriptome, possibly due to the disruption of RTA promoter activity. Mechanistically, overexpressed ORF48 was found to colocalize and interact with endogenous STING in HEK293 cells. Endogenous ORF48 and STING interactions were also detected in reactivated iSLK.219 cells. Compared with the control cell line, HUVEC cells stably expressing ORF48 exhibited repressed STING-dependent innate immune signaling upon ISD or diABZI treatment. However, the loss of ORF48 in our iSLK-based lytic system failed to induce IFNß production, suggesting a redundant role of ORF48 on STING signaling during the KSHV lytic phase. Thus, ORF48 is required for optimal KSHV lytic replication through additional mechanisms that need to be further explored.


Sujet(s)
Herpèsvirus humain de type 8 , Protéines virales , Réplication virale , Herpèsvirus humain de type 8/physiologie , Humains , Réplication virale/physiologie , Protéines virales/métabolisme , Protéines virales/génétique , Immunité innée , Cellules HEK293 , Sarcome de Kaposi/virologie , Sarcome de Kaposi/métabolisme , Régulation de l'expression des gènes viraux , Latence virale/physiologie , Infections à Herpesviridae/métabolisme , Infections à Herpesviridae/virologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE