Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.597
Filtrer
1.
Virol J ; 21(1): 236, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39350155

RÉSUMÉ

BACKGROUND: Cytomegalovirus (CMV) reactivation is a serious problem in recipients of allogeneic hematopoietic stem cell transplantation. Long-term latency depends on specific T cell immune reconstitution, which identifies various pathogens by T cell receptors (TCRs). However, the mechanisms underlying the selection of CMV-specific TCRs in recipients after transplantation remain unclear. METHODS: Using high-throughput sequencing and bioinformatics analysis, the T cell immune repertoire of seven CMV reactivated recipients (CRRs) were analyzed and compared to those of seven CMV non-activated recipients (CNRs) at an early stage after transplant. RESULTS: The counts of unique complementarity-determining region 3 (CDR3) were significantly higher in CNRs than in CRRs. The CDR3 clones in the CNRs exhibit higher homogeneity compared to the CRRs. With regard to T cell receptor ß-chain variable region (TRBV) and joint region (TRBJ) genotypes, significant differences were observed in the frequencies of TRBV6, BV23, and BV7-8 between the two groups. In addition to TRBV29-1/BJ1-2, TRBV2/BJ2-2, and TRBV12-4/BJ1-5, 11 V-J combinations had significantly different expression levels between CRRs and CNRs. CONCLUSIONS: The differences in TCR diversity, TRBV segments, and TRBV-BJ combinations observed between CNRs and CRRs might be associated with post-transplant CMV reactivation and could serve as a foundation for further research.


Sujet(s)
Infections à cytomégalovirus , Cytomegalovirus , Transplantation de cellules souches hématopoïétiques , Récepteurs aux antigènes des cellules T , Transplantation homologue , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Cytomegalovirus/immunologie , Cytomegalovirus/génétique , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/immunologie , Mâle , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie , Adulte , Femelle , Adulte d'âge moyen , Transplantation homologue/effets indésirables , Régions déterminant la complémentarité/génétique , Receveurs de transplantation , Séquençage nucléotidique à haut débit , Jeune adulte , Activation virale , Génotype , Lymphocytes T/immunologie , Adolescent , Biologie informatique/méthodes
2.
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
3.
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 , Femelle , Souris , Antigène CD80/génétique , Antigène CD80/métabolisme , Cornée/virologie , Cornée/anatomopathologie , Cornée/immunologie , Modèles animaux de maladie humaine , Herpès/immunologie , Herpès/virologie , Herpèsvirus humain de type 1/immunologie , Herpèsvirus humain de type 1/physiologie , Herpèsvirus humain de type 1/génétique , Kératite herpétique/virologie , Kératite herpétique/immunologie , Kératite herpétique/anatomopathologie , Souris de lignée C57BL , microARN , Ganglion trigéminal/virologie , Ganglion trigéminal/immunologie , Activation virale , Réplication virale , Mâle
4.
Front Immunol ; 15: 1403429, 2024.
Article de Anglais | MEDLINE | ID: mdl-39253091

RÉSUMÉ

Background: Currently, evidence regarding the causal relationship between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome is limited and inconsistent. Therefore, this study employs Mendelian randomization (MR) methodology to investigate the causal relationship between the two. Methods: This study selected 110 single-nucleotide polymorphisms (SNPs) of primary immunodeficiency-related genes as instrumental variables (IVs). Genetic associations of primary immunodeficiency-related genes were derived from recent genome-wide association studies (GWAS) data on human plasma protein levels and circulating immune cells. Data on genes associated with varicella-zoster virus reactivation syndrome were obtained from the GWAS Catalog and FINNGEN database, primarily analyzed using inverse variance weighting (IVW) and sensitivity analysis. Results: Through MR analysis, we identified 9 primary immunodeficiency-related genes causally associated with herpes zoster and its subsequent neuralgia; determined causal associations of 20 primary immunodeficiency-related genes with three vascular lesions (stroke, cerebral aneurysm, giant cell arteritis); revealed causal associations of 10 primary immunodeficiency-related genes with two ocular diseases (retinopathy, keratitis); additionally, three primary immunodeficiency-related genes each were associated with encephalitis, cranial nerve palsy, and gastrointestinal infections. Conclusions: This study discovers a certain association between primary immunodeficiency-related genes and varicella-zoster virus reactivation syndrome, yet further investigations are warranted to explore the specific mechanisms underlying these connections.


Sujet(s)
Étude d'association pangénomique , Herpèsvirus humain de type 3 , Analyse de randomisation mendélienne , Polymorphisme de nucléotide simple , Humains , Herpèsvirus humain de type 3/immunologie , Zona/génétique , Zona/immunologie , Zona/virologie , Activation virale , Maladies d'immunodéficience primaire/génétique , Maladies d'immunodéficience primaire/immunologie , Prédisposition génétique à une maladie , Infection à virus varicelle-zona/génétique , Infection à virus varicelle-zona/immunologie , Déficits immunitaires/génétique
5.
Transplant Proc ; 56(8): 1878-1884, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39232922

RÉSUMÉ

BACKGROUND: Reactivation of cytomegalovirus (CMV) is typically considered harmless as long as the immune system remains unaffected by medications or other factors. CMV reactivation may occur as a result of acute graft-versus-host disease of Grades II to IV. One possible factor contributing to this risk is the rise in the number of donors who lack genetic similarities or relationships. We hypothesized that the anti-CMV IgG level before transplantation could potentially serve as an indicator of the likelihood of CMV reactivation following hematopoietic cell transplantation. METHODS: We examined a cohort of young individuals who underwent allogeneic HCT between 1998 and 2022 to evaluate the occurrence of CMV reactivation. The patients were divided into 2 time periods: 1998 to 2016 (comparison group) and 2017 to 2022 (intervention group). RESULTS: Between 1998 and 2016, 292 patients underwent hematopoietic HCT. Recipients from 2017 to 2022 experienced a slightly higher risk of CMV reactivation than those from 1998 to 2016. The comparison of prophylactic and preemptive medication showed no significant difference between the periods (P = .32). Patients treated from 1998 to 2016 experienced a 23% decrease in the risk of symptomatic CMV reactivation and related illnesses compared to those treated from 2017 to 2022 (P = .08 and .15, respectively). CONCLUSIONS: Our study showed that the intervention group had more symptomatic CMV reactivations. Various factors may contribute to this, including CD19-directed immunotherapy and the CMV status of the recipient before transplantation.


Sujet(s)
Infections à cytomégalovirus , Cytomegalovirus , Transplantation de cellules souches hématopoïétiques , Activation virale , Humains , Transplantation de cellules souches hématopoïétiques/effets indésirables , Infections à cytomégalovirus/immunologie , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/épidémiologie , Enfant , Mâle , Cytomegalovirus/immunologie , Femelle , Enfant d'âge préscolaire , Adolescent , Transplantation homologue , Nourrisson , Maladie du greffon contre l'hôte/étiologie , Études rétrospectives , Antiviraux/usage thérapeutique
6.
Viruses ; 16(9)2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39339846

RÉSUMÉ

The herpes simplex viruses consist of the strains, HSV-1 and HSV-2, which are prevalent worldwide and lack a definitive cure. We aimed to explore the specific characteristics of HSV 1 and 2 infections, such as differences between gender assigned at birth, age at infection, site of infection, comorbidities, and effect of pregnancy, through a data analysis. Between 2011 and 2018, the Israeli Central Virology Laboratory diagnosed 9189 samples using multiplexed real-time PCR. In addition, we extracted all of the medical data for 287 females hospitalized at the Sheba Medical Center with HSV-1 (161) or HSV-2 (126) genital infections. HSV-2 was almost absent in the orofacial samples from both genders, while in other lesion sites, HSV-2 was significantly more abundant in females than in males (p < 0.05,). HSV-2 was initially detected at puberty. In the hospitalized females' malignancies, both HSV-1 and HSV-2 were found with a non-significant difference. Simultaneously, pregnancies were more common in females who were HSV-2-positive compared with those who were HSV-1-positive (27.8% vs. 12.4%, respectively, p < 0.01). Primary infections occur more with HSV-1 than with HSV-2 (15.6% vs. 3.2%, respectively). Our findings demonstrate that genital HSV-2 infection episodes are more frequent during pregnancy, suggesting that pregnancy may serve as a risk factor for HSV-2 reactivation or infection.


Sujet(s)
Herpèsvirus humain de type 1 , Herpèsvirus humain de type 2 , Complications infectieuses de la grossesse , Activation virale , Humains , Femelle , Grossesse , Herpèsvirus humain de type 2/génétique , Herpèsvirus humain de type 2/isolement et purification , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/isolement et purification , Adulte , Mâle , Complications infectieuses de la grossesse/virologie , Herpès/virologie , Jeune adulte , Herpès génital/virologie , Israël/épidémiologie , Adulte d'âge moyen , Adolescent , Sujet âgé
7.
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
8.
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 , Humains , Lignée cellulaire , ADN viral/génétique , Régulation de l'expression des gènes viraux , Génome viral , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/physiologie , Protéines virales/génétique , Protéines virales/métabolisme , Virion/métabolisme , Virion/génétique , Latence virale , Réplication virale
9.
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
11.
J Med Virol ; 96(8): e29849, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39135451

RÉSUMÉ

Kaposi's sarcoma (KS) is an angio-proliferative disease with a viral etiology and a multifactorial pathogenesis that results from immune dysfunction. In patients affected by latent viral infections such as herpesviruses, SARS-CoV-2 infection may result in lytic cycle reactivation in host cells. A robust immune system response is crucial for eliminating pathogens and resolving both latent and non-latent viral infections. We report a case series of KS characterized by tumor progression after SARS-CoV-2 infection. We performed a systematic literature review of the PubMed/MEDLINE and EMBASE databases. The keyword terms included "SARS-CoV-2," "HHV-8," "Kaposi's sarcoma," "IL-6," and "COVID-19." English language restriction was applied. Items not covered by our study were excluded. KS is a complex disease linked to an impaired immune system. Conditions that result in temporary or permanent immunodeficiency can trigger viral reactivation or exacerbate an existing disease. It is feasible that the increase in cytokine levels in COVID-19 patients, coupled with lymphocyte downregulation and treatment that induces herpesvirus lytic reactivation, may contribute to the progression of KS after SARS-CoV-2 infection. These observations suggest that patients with KS should be clinically monitored both during and after SARS-CoV-2 infection. Nevertheless, prospective data should be collected to validate this hypothesis and enhance our understanding of the mechanisms implicated in the onset or progression of KS.


Sujet(s)
COVID-19 , Herpèsvirus humain de type 8 , SARS-CoV-2 , Sarcome de Kaposi , Humains , COVID-19/immunologie , COVID-19/complications , COVID-19/virologie , Sarcome de Kaposi/virologie , Mâle , Adulte d'âge moyen , Femelle , Sujet âgé , Activation virale
12.
World J Gastroenterol ; 30(26): 3193-3197, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39086636

RÉSUMÉ

In this editorial we comment on the article published in the recent issue of the World Journal of Gastroenterology. We focus specifically on the problem of occult hepatitis B virus (HBV) infection, that is a result of previous hepatitis B (PHB) and a source for reactivation of HBV. The prevalence of PHB is underestimated due to the lack of population testing programs. However, this condition not only complicate anticancer treatment, but may be responsible for the development of other diseases, like cancer or autoimmune disorders. Here we unveil possible mechanisms responsible for realization of these processes and suggest practical approaches for diagnosis and treatment.


Sujet(s)
Virus de l'hépatite B , Hépatite B , Activation virale , Humains , Virus de l'hépatite B/immunologie , Virus de l'hépatite B/pathogénicité , Hépatite B/épidémiologie , Hépatite B/virologie , Hépatite B/diagnostic , Antiviraux/usage thérapeutique , Prévalence
15.
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
16.
Viruses ; 16(8)2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39205268

RÉSUMÉ

The acquisition or reactivation of Epstein-Barr virus (EBV) after allogeneic Hematopoietic Stem Cell Transplant (HSCT) can be associated with complications including the development of post-transplant lymphoproliferative disorder (PTLD), which is associated with significant morbidity and mortality. A number of risk factors for PTLD have been defined, including T-cell depletion, and approaches to monitoring EBV, especially in high-risk patients, with the use of preemptive therapy upon viral activation have been described. Newer therapies for the preemption or treatment of PTLD, such as EBV-specific cytotoxic T-cells, hold promise. Further studies to help define risks, diagnosis, and treatment of EBV-related complications are needed in this at-risk population.


Sujet(s)
Infections à virus Epstein-Barr , Transplantation de cellules souches hématopoïétiques , Herpèsvirus humain de type 4 , Syndromes lymphoprolifératifs , Transplantation homologue , Activation virale , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Infections à virus Epstein-Barr/virologie , Transplantation homologue/effets indésirables , Syndromes lymphoprolifératifs/étiologie , Syndromes lymphoprolifératifs/thérapie , Facteurs de risque
17.
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
18.
Int Immunopharmacol ; 141: 112793, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39146777

RÉSUMÉ

Renal transplantation is one of the primary approaches for curing end-stage kidney disease. With advancements in immunosuppressive agents, the short-term and long-term survival rates of transplanted kidneys have significantly improved. However, infections associated with potent immunosuppression have remained a persistent challenge. Among them, BK virus (BKV) reactivation following renal transplantation leading to BK virus-associated nephropathy (BKVAN) is a major cause of graft dysfunction. However, we still face significant challenges in understanding the pathogenesis, prevention, diagnosis, and treatment of BKVAN. These challenges include: 1. The mechanism of BKV reactivation under immunosuppressive conditions has not been well elucidated, leading to difficulties in breakthroughs in clinical research on prevention, diagnosis, and treatment. 2. Lack of proper identification of high-risk individuals, and effective personalized clinical management strategies. 3.Lack of early and sensitive diagnostic markers. 4. Lack of direct and effective treatment options due to the absence of specific antiviral drugs. The purpose of this review is to summarize the current status and cutting-edge advancements in BKV-related research, providing new methods and perspectives to address future research challenges.


Sujet(s)
Virus BK , Transplantation rénale , Infections à polyomavirus , Infections à virus oncogènes , Humains , Transplantation rénale/effets indésirables , Infections à polyomavirus/diagnostic , Infections à polyomavirus/immunologie , Infections à virus oncogènes/traitement médicamenteux , Infections à virus oncogènes/immunologie , Infections à virus oncogènes/virologie , Animaux , Activation virale , Immunosuppresseurs/usage thérapeutique , Immunosuppresseurs/effets indésirables , Antiviraux/usage thérapeutique
19.
JCI Insight ; 9(19)2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39163135

RÉSUMÉ

Drugs that inhibit HIV transcription and/or reactivation of latent HIV have been proposed as a strategy to reduce HIV-associated immune activation or to achieve a functional cure, yet comparative studies are lacking. We evaluated 26 drugs, including drugs previously reported to inhibit HIV transcription (inhibitors of Tat-dependent HIV transcription, Rev, HSF-1/PTEF-b, HSP90, Jak/Stat, or SIRT1/Tat deacetylation) and other agents that were not tested before (inhibitors of PKC, NF-κB, SP-1, or histone acetyltransferase; NR2F1 agonists), elongation (inhibitors of CDK9/ PTEF-b), completion (inhibitors of PolyA-polymerase), or splicing (inhibitors of human splice factors). To investigate if those drugs would vary in their ability to affect different blocks to HIV transcription, we measured levels of initiated, elongated, midtranscribed, completed, and multiply spliced HIV RNA in PBMCs from antiretroviral therapy-suppressed individuals following ex vivo treatment with each drug and subsequent T cell activation. We identified new drugs that prevent HIV reactivation, including CDK and splicing inhibitors. While some drugs inhibited 1 or 2 steps, other drugs (CDK inhibitors, splicing inhibitors, tanespimycin, and triptolide) inhibited multiple stages of HIV transcription and blocked the production of supernatant viral RNA. These drugs and targets deserve further study in strategies aimed at reducing HIV-associated immune activation or achieving a functional cure.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Activation virale , Latence virale , Humains , Latence virale/effets des médicaments et des substances chimiques , Activation virale/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Agents antiVIH/pharmacologie , Agents antiVIH/usage thérapeutique , Phénanthrènes/pharmacologie , Phénanthrènes/usage thérapeutique , Diterpènes/pharmacologie , Composés époxy/pharmacologie , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/virologie , Transcription génétique/effets des médicaments et des substances chimiques , Activation des lymphocytes/effets des médicaments et des substances chimiques , ARN viral , Mâle , Triterpènes pentacycliques
20.
Clin Res Hepatol Gastroenterol ; 48(8): 102447, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39181184

RÉSUMÉ

BACKGROUND: The hepatitis B surface antigen (HBsAg)-negative and antibody to hepatitis B core antigen (anti-HBc)-positive patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT) are at risk of HBV reactivation (HBVr). METHODS: To analyze the risk factors for HBVr, a total of 1,042 leukemia patients(≥18years of age), who underwent allo-HSCT from January 2016 to April 2022 in The First Affiliated Hospital of Soochow University, were enrolled in the study. Finally, 193 leukemia patients with resolved HBV infection were included into the study. RESULTS: HBVr occurred in 22 patients (11.39 %), and the median time to HBVr was 24 months (with a range of 11-51months). Hepatitis flares developed in 22.73 % of patients with HBVr, and hepatic failure occurred in 1 patient. During the follow-up period, only 1(1.3 %) patient experienced HBVr among 79 patients with antiviral prophylaxis. While 21(18.42 %) patients experienced HBVr among 114 patients without antiviral prophylaxis. The cumulative incidence of HBV reactivation at 3 years was 44.4. % for anti-HBs-negative donors/recipients with a low anti-HBs titer (<100IU/L) and 7.1 % for anti-HBs-positive donors/recipients with a high anti-HBs titer (≥100IU/L) respectively. In addition, univariate and multivariate Cox regression analyses confirmed the use of rituximab as a risk factor for HBV reactivation. CONCLUSION: The univariate and multivariate analyses confirmed that the anti-HBs titer in both recipients and donors are protective indicators to prevent incidence of HBVr. In addition, antiviral prophylaxis can significantly reduce the incidence of HBVr.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Hépatite B , Leucémies , Activation virale , Humains , Mâle , Facteurs de risque , Femelle , Transplantation de cellules souches hématopoïétiques/effets indésirables , Adulte , Adulte d'âge moyen , Hépatite B/prévention et contrôle , Leucémies/thérapie , Virus de l'hépatite B/physiologie , Jeune adulte , Transplantation homologue/effets indésirables , Anticorps de l'hépatite B/sang , Études rétrospectives , Adolescent , Antiviraux/usage thérapeutique , Antigènes de surface du virus de l'hépatite B/sang , Incidence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE