Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
1.
Clin Pharmacol Drug Dev ; 13(9): 1011-1023, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38993035

RÉSUMÉ

Avacopan 30 mg twice daily (BID) is approved for the treatment of severe active antineutrophil cytoplasmic autoantibody-associated vasculitis (granulomatosis with polyangiitis and microscopic polyangiitis). Food effect on avacopan pharmacokinetics (PKs) and PK bridging in Japanese participants were examined through 2 phase 1 studies involving healthy adult participants. In Study 1, an open-label, crossover trial, participants received oral administration of a single 30-mg dose of avacopan under fasted and fed conditions. Study 2 was a randomized, single-blind, placebo-controlled trial in Caucasian and Japanese participants: Part A investigated single doses of 10 and 30 mg of avacopan under fasted and fed conditions and Part B investigated 30 and 50 mg BID avacopan. The PKs of single-dose administrations of 10 and 30 mg in Japanese participants was compared with that in Caucasian participants under fasted conditions. Food substantially increased plasma avacopan area under the plasma concentration-time curve from time 0 to time infinity (AUC0-inf) by 1.72-fold, supporting the recommendation of taking avacopan with food. Maximum plasma concentration (Cmax) remained relatively unchanged. The median time to reach Cmax (tmax) was delayed by 3 hours. No significant food effect was observed on the active metabolite CCX168-M1 (M1) AUC. Avacopan and M1 exposures were <1.5-fold higher in Japanese participants than in Caucasian participants following multiple-dose administration of avacopan.


Sujet(s)
Aire sous la courbe , Asiatiques , Études croisées , Jeûne , Interactions aliments-médicaments , Volontaires sains , , Humains , Adulte , Mâle , Femelle , Méthode en simple aveugle , Jeune adulte , Administration par voie orale , Adulte d'âge moyen , Relation dose-effet des médicaments , Peuples d'Asie de l'Est
2.
Ann Allergy Asthma Immunol ; 133(2): 186-193.e2, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38719149

RÉSUMÉ

BACKGROUND: Nasal congestion could affect the absorption of an epinephrine nasal spray (ENS). OBJECTIVE: To compare the pharmacokinetics of 13.2 mg ENS with nasal congestion vs without congestion and vs intramuscular (IM) treatments. METHODS: This phase I, open-label, 4-period randomized crossover study enrolled 51 healthy adults with seasonal allergies into cohorts that received a single dose of 13.2 mg ENS (NDS1C; Bryn Pharma, Lebanon, New Jersey) administered as 2 consecutive sprays in either opposite nostrils (cohort 1) or the same nostril (cohort 2). Both cohorts received 13.2 mg ENS with and without nasal allergen challenge (NAC), 0.3 mg IM epinephrine by autoinjector, and 0.5 mg IM epinephrine by manual syringe (MS). RESULTS: The ENS after NAC resulted in higher extent and peak exposures and more rapid time to maximum plasma concentration vs ENS without NAC and IM treatments. In cohort 1, the maximum observed baseline-adjusted epinephrine plasma concentration (pg/mL) of ENS with NAC, IM autoinjector, IM MS, or ENS without NAC was 458.0, 279.0, 364.2, and 270.1, respectively, and in cohort 2 was 436.3, 228.2, 322.3, and 250.8, respectively. The maximum observed baseline-adjusted epinephrine plasma concentration geometric mean ratio (90% CI) for ENS with NAC vs without NAC in cohort 1 was 170% (123%-234%), and in cohort 2 was 174% (115%-263%). In cohort 1, the time to maximum plasma concentration was 15, 21, 45, and 25 minutes, respectively, and in cohort 2 was 18, 20, 45, and 20 minutes, respectively (P < .01 for ENS with NAC vs IM MS). The postdose mean heart rate and blood pressure remained stable and relatively similar to predose values regardless of plasma epinephrine concentration. Mild nausea and headache were the most common adverse events with ENS. CONCLUSION: The 13.2 mg ENS with congestion exhibited enhanced absorption vs IM treatments and ENS without congestion and seemed to be well tolerated. There was no clinically impactful relationship between pharmacodynamic effects and plasma epinephrine concentration.


Sujet(s)
Administration par voie nasale , Études croisées , Épinéphrine , Humains , Épinéphrine/pharmacocinétique , Épinéphrine/administration et posologie , Mâle , Adulte , Femelle , Adulte d'âge moyen , Jeune adulte , Pulvérisations nasales , Rhinite allergique saisonnière/traitement médicamenteux , Obstruction nasale/traitement médicamenteux , Adolescent , Allergènes/immunologie , Allergènes/administration et posologie , Allergènes/pharmacocinétique , Injections musculaires
3.
J Allergy Clin Immunol Glob ; 3(2): 100200, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38328805

RÉSUMÉ

Background: Recent acute anaphylaxis guideline updates have identified remaining unmet needs based on currently available therapeutic options as a critical focus. Objective: We compared the pharmacokinetic, pharmacodynamic, safety, and tolerability profiles of intranasal epinephrine with intramuscular epinephrine administered by autoinjector and manual syringe. Methods: An open-label, 3-period crossover study was conducted in 116 healthy adult volunteers to assess the bioavailability of a single 13.2 mg intranasal dose of epinephrine compared to a 0.3 mg intramuscular autoinjector and a 0.5 mg manual syringe. Patients with epinephrine concentrations of 50, 100, and 200 pg/mL at 10, 20, 30, and 60 minutes after dosing were also evaluated. Results: Pharmacokinetic parameters for the 13.2 mg intranasal dose exceeded those of the 0.3 mg autoinjector with a rapid and higher maximum observed concentration (intranasal, 429.4 pg/mL; autoinjector, 328.6 pg/mL) and greater systemic exposure (AUC0-360; intranasal, 39,060 pg∙min/mL; autoinjector, 17,440 pg∙min/mL). Similar results were observed compared to the 0.5 mg manual syringe. Pharmacokinetic parameters for opposite-nostril and same-nostril dosing were higher than both intramuscular doses, except time to reach maximum observed concentration, which was bracketed between the 2 intramuscular doses (intranasal opposite and same nostril, 20 minutes; autoinjector, 14.9 minutes; manual syringe, 45 minutes). Similar effects on blood pressure and heart rate were observed for intranasal and autoinjector administration. Intranasal epinephrine was safe and well tolerated. No serious or unexpected adverse events were reported, confirming results from earlier clinical studies. Conclusions: Bidose epinephrine spray addresses the unmet medical and patient needs for a needle-free, convenient, and effective dose-delivery system for self-administration of epinephrine that is as good as or better than the 0.3 mg autoinjector.

4.
Clin Pharmacol Drug Dev ; 13(5): 517-533, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38423992

RÉSUMÉ

Avacopan, a complement 5a receptor (C5aR) antagonist approved for treating severe active antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis, was evaluated in 2 clinical drug-drug interaction studies. The studies assessed the impact of avacopan on the pharmacokinetics (PK) of CYP3A4 substrates midazolam and simvastatin and CYP2C9 substrate celecoxib, and the influence of CYP3A4 inhibitor itraconazole and inducer rifampin on the PKs of avacopan. The results indicated that twice-daily oral administration of 30 mg of avacopan increased the area under the curve (AUC) of midazolam by 1.81-fold and celecoxib by 1.15-fold when administered without food, and twice-daily oral administration of 30 or 60 mg of avacopan increased the AUC of simvastatin by approximately 2.6-3.5-fold and the AUC of the active metabolite ß-hydroxy-simvastatin acid by approximately 1.4-1.7-fold when co-administered with food. Furthermore, the AUC of avacopan increased by approximately 2.19-fold when co-administered with itraconazole and decreased by approximately 13.5-fold when co-administered with rifampin. These findings provide critical insights into the potential drug-drug interactions involving avacopan, which could have significant implications for patient care and treatment planning. (NCT06207682).


Sujet(s)
Cytochrome P-450 CYP2C9 , Inhibiteurs du cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A , Interactions médicamenteuses , Volontaires sains , Itraconazole , Midazolam , Rifampicine , Simvastatine , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Administration par voie orale , Aire sous la courbe , Cytochrome P-450 CYP2C9/métabolisme , Cytochrome P-450 CYP3A/métabolisme , Inhibiteurs du cytochrome P-450 CYP3A/pharmacologie , Inhibiteurs du cytochrome P-450 CYP3A/administration et posologie , Inhibiteurs du cytochrome P-450 CYP3A/pharmacocinétique , Interactions aliments-médicaments , Itraconazole/pharmacologie , Itraconazole/administration et posologie , Itraconazole/pharmacocinétique , Midazolam/pharmacocinétique , Midazolam/administration et posologie , Rifampicine/pharmacologie , Rifampicine/administration et posologie , Rifampicine/pharmacocinétique , Simvastatine/pharmacocinétique , Simvastatine/administration et posologie , Simvastatine/effets indésirables
5.
Front Microbiol ; 14: 1320856, 2023.
Article de Anglais | MEDLINE | ID: mdl-38075874

RÉSUMÉ

The reduced pathogenicity of the omicron BA.1 sub-lineage compared to earlier variants is well described, although whether such attenuation is retained for later variants like BA.5 and XBB remains controversial. We show that BA.5 and XBB isolates were significantly more pathogenic in K18-hACE2 mice than a BA.1 isolate, showing increased neurotropic potential, resulting in fulminant brain infection and mortality, similar to that seen for original ancestral isolates. BA.5 also infected human cortical brain organoids to a greater extent than the BA.1 and original ancestral isolates. In the brains of mice, neurons were the main target of infection, and in human organoids neuronal progenitor cells and immature neurons were infected. The results herein suggest that evolving omicron variants may have increasing neurotropic potential.

6.
Cell Rep ; 42(5): 112322, 2023 05 30.
Article de Anglais | MEDLINE | ID: mdl-37105170

RÉSUMÉ

Crosstalk between cardiac cells is critical for heart performance. Here we show that vascular cells within human cardiac organoids (hCOs) enhance their maturation, force of contraction, and utility in disease modeling. Herein we optimize our protocol to generate vascular populations in addition to epicardial, fibroblast, and cardiomyocyte cells that self-organize into in-vivo-like structures in hCOs. We identify mechanisms of communication between endothelial cells, pericytes, fibroblasts, and cardiomyocytes that ultimately contribute to cardiac organoid maturation. In particular, (1) endothelial-derived LAMA5 regulates expression of mature sarcomeric proteins and contractility, and (2) paracrine platelet-derived growth factor receptor ß (PDGFRß) signaling from vascular cells upregulates matrix deposition to augment hCO contractile force. Finally, we demonstrate that vascular cells determine the magnitude of diastolic dysfunction caused by inflammatory factors and identify a paracrine role of endothelin driving dysfunction. Together this study highlights the importance and role of vascular cells in organoid models.


Sujet(s)
Cellules endothéliales , Myocytes cardiaques , Humains , Myocytes cardiaques/métabolisme , Péricytes/métabolisme , Transduction du signal , Organoïdes/métabolisme
7.
Theranostics ; 12(6): 2811-2832, 2022.
Article de Anglais | MEDLINE | ID: mdl-35401827

RÉSUMÉ

Rational: The mutating SARS-CoV-2 potentially impairs the efficacy of current vaccines or antibody-based treatments. Broad-spectrum and rapid anti-virus methods feasible for regular epidemic prevention against COVID-19 or alike are urgently called for. Methods: Using SARS-CoV-2 virus and bioengineered pseudoviruses carrying ACE2-binding spike protein domains, we examined the efficacy of cold atmospheric plasma (CAP) on virus entry prevention. Results: We found that CAP could effectively inhibit the entry of virus into cells. Direct CAP or CAP-activated medium (PAM) triggered rapid internalization and nuclear translocation of the virus receptor, ACE2, which began to return after 5 hours and was fully recovered by 12 hours. This was seen in vitro with both VERO-E6 cells and human mammary epithelial MCF10A cells, and in vivo. Hydroxyl radical (·OH) and species derived from its interactions with other species were found to be the most effective CAP components for triggering ACE2 nucleus translocation. The ERα/STAT3(Tyr705) and EGFR(Tyr1068/1086)/STAT3(Tyr705) axes were found to interact and collectively mediate the effects on ACE2 localization and expression. Conclusions: Our data support the use of PAM in helping control SARS-CoV-2 if developed into products for nose/mouth spray; an approach extendable to other viruses utilizing ACE2 for host entry.


Sujet(s)
COVID-19 , Gaz plasmas , Angiotensin-converting enzyme 2 , COVID-19/prévention et contrôle , Humains , Gaz plasmas/pharmacologie , Liaison aux protéines , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus/métabolisme
8.
Viruses ; 15(1)2022 12 31.
Article de Anglais | MEDLINE | ID: mdl-36680179

RÉSUMÉ

The global coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spawned an ongoing demand for new research reagents and interventions. Herein we describe a panel of monoclonal antibodies raised against SARS-CoV-2. One antibody showed excellent utility for immunohistochemistry, clearly staining infected cells in formalin-fixed and paraffin embedded lungs and brains of mice infected with the original and the omicron variants of SARS-CoV-2. We demonstrate the reactivity to multiple variants of concern using ELISAs and describe the use of the antibodies in indirect immunofluorescence assays, Western blots, and rapid antigen tests. Finally, we illustrate the ability of two antibodies to reduce significantly viral tissue titers in K18-hACE2 transgenic mice infected with the original and an omicron isolate of SARS-CoV-2.


Sujet(s)
Anticorps monoclonaux , COVID-19 , Animaux , Humains , Souris , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/génétique , Souris transgéniques , Anticorps antiviraux , Anticorps neutralisants
9.
J Biol Chem ; 298(2): 101547, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34971704

RÉSUMÉ

Complex diseases such as cancer and diabetes are underpinned by changes in metabolism, specifically by which and how nutrients are catabolized. Substrate utilization can be directly examined by measuring a metabolic endpoint rather than an intermediate (such as a metabolite in the tricarboxylic acid cycle). For instance, oxidation of specific substrates can be measured in vitro by incubation of live cultures with substrates containing radiolabeled carbon and measuring radiolabeled carbon dioxide. To increase throughput, we previously developed a miniaturized platform to measure substrate oxidation of both adherent and suspension cells using multiwell plates rather than flasks. This enabled multiple conditions to be examined simultaneously, ideal for drug screens and mechanistic studies. However, like many metabolic assays, this was not compatible with bicarbonate-buffered media, which is susceptible to alkalinization upon exposure to gas containing little carbon dioxide such as air. While other buffers such as HEPES can overcome this problem, bicarbonate has additional biological roles as a metabolic substrate and in modulating hormone signaling. Here, we create a bicarbonate-buffered well-plate platform to measure substrate oxidation. This was achieved by introducing a sealed environment within each well that was equilibrated with carbon dioxide, enabling bicarbonate buffering. As proof of principle, we assessed metabolic flux in cultured adipocytes, demonstrating that bicarbonate-buffered medium increased lipogenesis, glucose oxidation, and sensitivity to insulin in comparison to HEPES-buffered medium. This convenient and high-throughput method facilitates the study and screening of metabolic activity under more physiological conditions to aid biomedical research.


Sujet(s)
Hydrogénocarbonates , Dioxyde de carbone , Techniques de culture cellulaire , Milieux de culture , Substances tampon , HEPES , Oxydoréduction
10.
Commun Biol ; 4(1): 557, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33976375

RÉSUMÉ

Dengue virus (DENV) is spread from human to human through the bite of the female Aedes aegypti mosquito and leads to about 100 million clinical infections yearly. Treatment options and vaccine availability for DENV are limited. Defective interfering particles (DIPs) are considered a promising antiviral approach but infectious virus contamination has limited their development. Here, a DENV-derived DIP production cell line was developed that continuously produced DENV-free DIPs. The DIPs contained and could deliver to cells a DENV serotype 2 subgenomic defective-interfering RNA, which was originally discovered in DENV infected patients. The DIPs released into cell culture supernatant were purified and could potently inhibit replication of all DENV serotypes in cells. Antiviral therapeutics are limited for many viral infection. The DIP system described could be re-purposed to make antiviral DIPs for many other RNA viruses such as SARS-CoV-2, yellow fever, West Nile and Zika viruses.


Sujet(s)
Virus défectifs , Vaccins contre la dengue/usage thérapeutique , Virus de la dengue/croissance et développement , Dengue/prévention et contrôle , Réplication virale , Animaux , Lignée cellulaire tumorale , Chlorocebus aethiops , Virus défectifs/génétique , Virus défectifs/métabolisme , Dengue/virologie , Virus de la dengue/génétique , Virus de la dengue/métabolisme , Gènes rapporteurs , Cellules HEK293 , Interactions hôte-pathogène , Humains , Protéines luminescentes/biosynthèse , Protéines luminescentes/génétique , ARN viral/biosynthèse , ARN viral/génétique , Cellules Vero , Charge virale
11.
Cell ; 184(8): 2167-2182.e22, 2021 04 15.
Article de Anglais | MEDLINE | ID: mdl-33811809

RÉSUMÉ

Cardiac injury and dysfunction occur in COVID-19 patients and increase the risk of mortality. Causes are ill defined but could be through direct cardiac infection and/or inflammation-induced dysfunction. To identify mechanisms and cardio-protective drugs, we use a state-of-the-art pipeline combining human cardiac organoids with phosphoproteomics and single nuclei RNA sequencing. We identify an inflammatory "cytokine-storm", a cocktail of interferon gamma, interleukin 1ß, and poly(I:C), induced diastolic dysfunction. Bromodomain-containing protein 4 is activated along with a viral response that is consistent in both human cardiac organoids (hCOs) and hearts of SARS-CoV-2-infected K18-hACE2 mice. Bromodomain and extraterminal family inhibitors (BETi) recover dysfunction in hCOs and completely prevent cardiac dysfunction and death in a mouse cytokine-storm model. Additionally, BETi decreases transcription of genes in the viral response, decreases ACE2 expression, and reduces SARS-CoV-2 infection of cardiomyocytes. Together, BETi, including the Food and Drug Administration (FDA) breakthrough designated drug, apabetalone, are promising candidates to prevent COVID-19 mediated cardiac damage.


Sujet(s)
COVID-19/complications , Cardiotoniques/usage thérapeutique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Cardiopathies/traitement médicamenteux , Quinazolinones/usage thérapeutique , Facteurs de transcription/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/métabolisme , Animaux , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire , Cytokines/métabolisme , Femelle , Cardiopathies/étiologie , Cellules souches embryonnaires humaines , Humains , Inflammation/complications , Inflammation/traitement médicamenteux , Souris , Souris de lignée C57BL , Facteurs de transcription/métabolisme , Traitements médicamenteux de la COVID-19
12.
mBio ; 10(4)2019 08 27.
Article de Anglais | MEDLINE | ID: mdl-31455650

RÉSUMÉ

Nullbasic is a mutant form of the HIV-1 transcriptional activator protein (Tat) that strongly inhibits HIV-1 transcription and replication in lymphocytes in vitro To investigate Nullbasic inhibition in vivo, we employed an NSG mouse model where animals were engrafted with primary human CD4+ cells expressing a Nullbasic-ZsGreen1 (NB-ZSG) fusion protein or ZSG. NB-ZSG and ZSG were delivered by using a retroviral vector where CD4+ cells were transduced either prior to (preinfection) or following (postinfection) HIV-1 infection. The transduced cells were analyzed in vitro up to 10 days postinfection (dpi) and in vivo up to 39 dpi. Compared to ZSG, NB-ZSG strongly inhibited HIV-1 replication both in vitro and in vivo using preinfection treatment. In vitro, HIV-1 mRNA levels in cells were reduced by up to 60-fold. In vivo, HIV-1 RNA was undetectable in plasma samples during the course of the experiment, and HIV-1 mRNA levels in resident CD4+ cells in organ tissue were reduced up to 2,800-fold. Postinfection treatment of HIV-1-infected cells with NB-ZSG attenuated HIV-1 infection for up to 14 days. In vitro, a 25-fold reduction of viral mRNA in cells was observed but diminished to a <2-fold reduction by 10 dpi. In vivo, HIV-1 RNA was undetectable in plasma of NB-ZSG mice at 14 dpi but afterwards was not significantly different between NB-ZSG mice and control mice. However, we observed higher levels of CD4+ cells in NB-ZSG mice than in control mice, suggesting that NB-ZSG imparted a survival advantage to HIV-1-infected animals.IMPORTANCE HIV-1 infection is effectively controlled by antiviral therapy that inhibits virus replication and reduces viral loads below detectable levels in patients. However, therapy interruption leads to viral rebound due to latently infected cells, which serve as a source of continued viral infection. Interest in strategies leading to a functional cure for HIV-1 infection by long-term or permanent viral suppression is growing. Here, we show that a mutant form of the HIV-1 Tat protein, referred to as Nullbasic, inhibits HIV-1 transcription in infected CD4+ cells in vivo Analysis shows that stable expression of Nullbasic in CD4+ cells could lead to durable anti-HIV-1 activity. Nullbasic, as a gene therapy candidate, could be a part of a functional-cure strategy to suppress HIV-1 transcription and replication.


Sujet(s)
Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Protéines mutantes/pharmacologie , Réplication virale/effets des médicaments et des substances chimiques , Produits du gène tat du virus de l'immunodéficience humaine/pharmacologie , Animaux , Lymphocytes T CD4+/virologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Souris , Protéines mutantes/génétique , Protéines mutantes/métabolisme , Charge virale , Produits du gène tat du virus de l'immunodéficience humaine/génétique , Produits du gène tat du virus de l'immunodéficience humaine/métabolisme
13.
J Virol ; 93(12)2019 06 15.
Article de Anglais | MEDLINE | ID: mdl-30918071

RÉSUMÉ

HIV-1 replication requires direct interaction between HIV-1 reverse transcriptase (RT) and cellular eukaryotic translation elongation factor 1A (eEF1A). Our previous work showed that disrupting this interaction inhibited HIV-1 uncoating, reverse transcription, and replication, indicating its potential as an anti-HIV-1 target. In this study, we developed a sensitive, live-cell split-luciferase complementation assay (NanoBiT) to quantitatively measure inhibition of HIV-1 RT interaction with eEF1A. We used this to screen a small molecule library and discovered small-molecule oxazole-benzenesulfonamides (C7, C8, and C9), which dose dependently and specifically inhibited the HIV-1 RT interaction with eEF1A. These compounds directly bound to HIV-1 RT in a dose-dependent manner, as assessed by a biolayer interferometry (BLI) assay, but did not bind to eEF1A. These oxazole-benzenesulfonamides did not inhibit enzymatic activity of recombinant HIV-1 RT in a homopolymer assay but did inhibit reverse transcription and infection of both wild-type (WT) and nonnucleoside reverse transcriptase inhibitor (NNRTI)-resistant HIV-1 in a dose-dependent manner in HEK293T cells. Infection of HeLa cells was significantly inhibited by the oxazole-benzenesulfonamides, and the antiviral activity was most potent against replication stages before 8 h postinfection. In human primary activated CD4+ T cells, C7 inhibited HIV-1 infectivity and replication up to 6 days postinfection. The data suggest a novel mechanism of HIV-1 inhibition and further elucidate how the RT-eEF1A interaction is important for HIV-1 replication. These compounds provide potential to develop a new class of anti-HIV-1 drugs to treat WT and NNRTI-resistant strains in people infected with HIV.IMPORTANCE Antiretroviral drugs protect many HIV-positive people, but their success can be compromised by drug-resistant strains. To combat these strains, the development of new classes of HIV-1 inhibitors is essential and a priority in the field. In this study, we identified small molecules that bind directly to HIV-1 reverse transcriptase (RT) and inhibit its interaction with cellular eEF1A, an interaction which we have previously identified as crucial for HIV-1 replication. These compounds inhibit intracellular HIV-1 reverse transcription and replication of WT HIV-1, as well as HIV-1 mutants that are resistant to current RT inhibitors. A novel mechanism of action involving inhibition of the HIV-1 RT-eEF1A interaction is an important finding and a potential new way to combat drug-resistant HIV-1 strains in infected people.


Sujet(s)
Transcriptase inverse du VIH/effets des médicaments et des substances chimiques , Facteur-1 d'élongation de la chaîne peptidique/métabolisme , Agents antiVIH/pharmacologie , Cellules HEK293 , Infections à VIH/traitement médicamenteux , Transcriptase inverse du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Cellules HeLa , Humains , Oxazoles/métabolisme , Oxazoles/pharmacologie , Inhibiteurs de la transcriptase inverse/pharmacologie , Transcription inverse/effets des médicaments et des substances chimiques , Sulfonamides/métabolisme , Sulfonamides/pharmacologie , Réplication virale/effets des médicaments et des substances chimiques ,
14.
Virology ; 530: 65-74, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30782564

RÉSUMÉ

The eukaryotic translation elongation factor 1A (eEF1A) has two cell-type specific paralogs, eEF1A1 and eEF1A2. Both paralogs undertake a canonical function in delivering aminoacyl-tRNA to the ribosome for translation, but differences in other functions are emerging. eEF1A1 has been reported to be important for the replication of many viruses, but no study has specifically linked the eEF1A2 paralog. We have previously demonstrated that eEF1A1 directly interacts with HIV-1 RT and supports efficient reverse transcription. Here, we showed that RT interacted more strongly with eEF1A1 than with eEF1A2 in immunoprecipitation assay. Biolayer interferometry using eEF1A paralogs showed different association and dissociation rates with RT. Over expressed eEF1A1, but not eEF1A2, was able to restore HIV-1 reverse transcription efficiency in HEK293T cells with endogenous eEF1A knocked-down and HIV-1 reverse transcription efficiency correlated with the level of eEF1A1 mRNA, but not to eEF1A2 mRNA in both HEK293T and primary human skeletal muscle cells.


Sujet(s)
Transcriptase inverse du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , Interactions hôte-pathogène , Facteur-1 d'élongation de la chaîne peptidique/métabolisme , Transcription inverse , Cellules HEK293 , Humains , Immunoprécipitation , Cellules musculaires , Liaison aux protéines
15.
Virol J ; 15(1): 182, 2018 11 26.
Article de Anglais | MEDLINE | ID: mdl-30477508

RÉSUMÉ

Cellular protein eukaryotic translation elongation factor 1A (eEF1A) is an actin binding protein that plays a role in the formation of filamentous actin (F-actin) bundles. F-Actin regulates multiple stages of respiratory syncytial virus (RSV) replication including assembly and budding. Our previous study demonstrated that eEF1A knock-down significantly reduced RSV replication. Here we investigated if the eEF1A function in actin bundle formation was important for RSV replication and release. To investigate this, eEF1A function was impaired in HEp-2 cells by either knock-down of eEF1A with siRNA, or treatment with an eEF1A inhibitor, didemnin B (Did B). Cell staining and confocal microscopy analysis showed that both eEF1A knock-down and treatment with Did B resulted in disruption of cellular stress fiber formation and elevated accumulation of F-actin near the plasma membrane. When treated cells were then infected with RSV, there was also reduced formation of virus-induced cellular filopodia. Did B treatment, similarly to eEF1A knock-down, reduced the release of infectious RSV, but unlike eEF1A knock-down, did not significantly affect RSV genome replication. The lower infectious virus production in Did B treated cells also reduced RSV-induced cell death. In conclusion, the cellular factor eEF1A plays an important role in the regulation of F-actin stress fiber formation required for RSV assembly and release.


Sujet(s)
Actines/métabolisme , Facteur-1 d'élongation de la chaîne peptidique/génétique , Virus respiratoire syncytial humain/physiologie , Fibres de stress/physiologie , Réplication virale , Actines/génétique , Lignée cellulaire tumorale , Depsipeptides/pharmacologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/virologie , Techniques de knock-down de gènes , Humains , Pseudopodes/physiologie , Pseudopodes/virologie , Virus respiratoire syncytial humain/génétique
16.
Virol Sin ; 33(2): 142-152, 2018 Apr.
Article de Anglais | MEDLINE | ID: mdl-29541943

RÉSUMÉ

Nullbasic is a mutant form of HIV-1 Tat that has strong ability to protect cells from HIV-1 replication by inhibiting three different steps of viral replication: reverse transcription, Rev export of viral mRNA from the nucleus to the cytoplasm and transcription of viral mRNA by RNA polymerase II. We previously showed that Nullbasic inhibits transduction of human cells including T cells by HIV-1-based lentiviral vectors. Here we investigated whether the Nullbasic antagonists huTat2 (a Tat targeting intrabody), HIV-1 Tat or Rev proteins or cellular DDX1 protein could improve transduction by a HIV-1 lentiviral vector conveying Nullbasic-ZsGreen1 to human T cells. We show that overexpression of huTat2, Tat-FLAG and DDX1-HA in virus-like particle (VLP) producer cells significantly improved transduction efficiency of VLPs that convey Nullbasic in Jurkat cells. Specifically, co-expression of Tat-FLAG and DDX1-HA in the VLP producer cell improved transduction efficiency better than if used individually. Transduction efficiencies could be further improved by including a spinoculation step. However, the same optimised protocol and using the same VLPs failed to transduce primary human CD4+ T cells. The results imply that the effects of Nullbasic on VLPs on early HIV-1 replication are robust in human CD4+ T cells. Given this significant block to lentiviral vector transduction by Nullbasic in primary CD4+ T cells, our data indicate that gammaretroviral, but not lentiviral, vectors are suitable for delivering Nullbasic to primary human T cells.


Sujet(s)
Agents antiVIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Protéines mutantes/métabolisme , Lymphocytes T/virologie , Transduction génétique , Produits du gène tat du virus de l'immunodéficience humaine/métabolisme , Cellules cultivées , DEAD-box RNA helicases/génétique , DEAD-box RNA helicases/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , Humains , Protéines mutantes/génétique , Produits du gène rev du virus de l'immunodéficience humaine/génétique , Produits du gène rev du virus de l'immunodéficience humaine/métabolisme , Produits du gène tat du virus de l'immunodéficience humaine/génétique
17.
Virol J ; 14(1): 52, 2017 03 14.
Article de Anglais | MEDLINE | ID: mdl-28288662

RÉSUMÉ

BACKGROUND: Nullbasic is a mutant HIV-1 Tat protein that inhibits HIV-1 replication via three independent mechanisms that disrupts 1) reverse transcription of the viral RNA genome into a DNA copy, 2) HIV-1 Rev protein function required for viral mRNA transport from the nucleus to the cytoplasm and 3) HIV-1 mRNA transcription by RNA Polymerase II. The Nullbasic protein is derived from the subtype B strain HIV-1BH10 and has only been tested against other HIV-1 subtype B strains. However, subtype B strains only account for ~10% of HIV-1 infections globally and HIV-1 Tat sequences vary between subtypes especially for subtype C, which is responsible for ~50% HIV-1 infection worldwide. These differences could influence the ability of Tat to interact with RNA and cellular proteins and thus could affect the antiviral activity of Nullbasic. Therefore, Nullbasic was tested against representative HIV-1 strains from subtypes C, D and A/D recombinant to determine if it can inhibit their replication. METHODS: Nullbasic was delivered to human cells using a self-inactivating (SIN) γ-retroviral system. We evaluated Nullbasic-mCherry (NB-mCh) fusion protein activity against the HIV-1 strains in TZM-bl cell lines for inhibition of transactivation and virus replication. We also examined antiviral activity of Nullbasic-ZsGreen1 (NB-ZSG1) fusion protein against the same strains in primary CD4+ T cells. The Nullbasic expression was monitored by western blot and flow cytometry. The effects of Nullbasic on primary CD4+ T cells cytotoxicity, proliferation and apoptosis were also examined. RESULTS: The results show that Nullbasic inhibits Tat-mediated transactivation and virus replication of all the HIV-1 strains tested in TZM-bl cells. Importantly, Nullbasic inhibits replication of the HIV-1 strains in primary CD4+ T cells without affecting cell proliferation, cytotoxicity or level of apoptotic cells. CONCLUSION: A SIN-based γ-retroviral vector used to express Nullbasic fusion proteins improved protein expression particularly in primary CD4+ T cells. Nullbasic has antiviral activity against all strains from the subtypes tested although small differences in viral inhibition were observed. Further improvement of in γ-retroviral vector stable expression of Nullbasic expression may have utility in a future gene therapy approach applicable to genetically diverse HIV-1 strains.


Sujet(s)
Antiviraux/métabolisme , Génotype , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Protéines mutantes/métabolisme , Réplication virale , Produits du gène tat du virus de l'immunodéficience humaine/métabolisme , Lymphocytes T CD4+/physiologie , Lymphocytes T CD4+/virologie , Cellules cultivées , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/classification , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Protéines mutantes/génétique , Produits du gène tat du virus de l'immunodéficience humaine/génétique
18.
mBio ; 7(4)2016 07 05.
Article de Anglais | MEDLINE | ID: mdl-27381288

RÉSUMÉ

UNLABELLED: Nullbasic is a derivative of the HIV-1 transactivator of transcription (Tat) protein that strongly inhibits HIV-1 replication in lymphocytes. Here we show that lentiviral vectors that constitutively express a Nullbasic-ZsGreen1 (NB-ZSG1) fusion protein by the eEF1α promoter led to robust long-term inhibition of HIV-1 replication in Jurkat cells. Although Jurkat-NB-ZSG1 cells were infected by HIV-1, no virus production could be detected and addition of phorbol ester 12-myristate 13-acetate (PMA) and JQ1 had no effect, while suberanilohydroxamic acid (SAHA) modestly stimulated virus production but at levels 300-fold lower than those seen in HIV-1-infected Jurkat-ZSG1 cells. Virus replication was not recovered by coculture of HIV-1-infected Jurkat-NB-ZSG1 cells with uninfected Jurkat cells. Latently infected Jurkat latent 6.3 and ACH2 cells treated with latency-reversing agents produced measurable viral capsid (CA), but little or none was made when they expressed NB-ZSG1. When Jurkat cells chronically infected with HIV-1 were transduced with lentiviral virus-like particles conveying NB-ZSG1, a >3-log reduction in CA production was observed. Addition of PMA increased virus CA production but at levels 500-fold lower than those seen in nontransduced Jurkat cells. Transcriptome sequencing analysis confirmed that HIV-1 mRNA was strongly inhibited by NB-ZSG1 but indicated that full-length viral mRNA was made. Analysis of HIV-1-infected Jurkat cells expressing NB-ZSG1 by chromatin immunoprecipitation assays indicated that recruitment of RNA polymerase II (RNAPII) and histone 3 lysine 9 acetylation were inhibited. The reduction of HIV-1 promoter-associated RNAPII and epigenetic changes in viral nucleosomes indicate that Nullbasic can inhibit HIV-1 replication by enforcing viral silencing in cells. IMPORTANCE: HIV-1 infection is effectively controlled by antiviral therapy that inhibits virus replication and reduces measurable viral loads in patients below detectable levels. However, therapy interruption leads to viral rebound due to latently infected cells that serve as a source of continued viral infection. Interest in strategies leading to a functional cure of HIV infection by permanent viral suppression, which may be achievable, is growing. Here we show that a mutant form of the HIV-1 Tat protein, referred to as Nullbasic, can inhibit HIV-1 transcription in infected Jurkat T cell to undetectable levels. Analysis shows that Nullbasic alters the epigenetic state of the HIV-1 long terminal repeat promoter, inhibiting its association with RNA polymerase II. This study indicates that key cellular proteins and pathways targeted here can silence HIV-1 transcription. Further elucidation could lead to functional-cure strategies by suppression of HIV transcription, which may be achievable by a pharmacological method.


Sujet(s)
Antiviraux/métabolisme , Infections à VIH/thérapie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Protéines mutantes/pharmacologie , Transcription génétique/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Produits du gène tat du virus de l'immunodéficience humaine/métabolisme , Analyse de profil d'expression de gènes , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Cellules Jurkat , Protéines mutantes/génétique , Latence virale , Produits du gène tat du virus de l'immunodéficience humaine/génétique
19.
Blood ; 126(13): 1609-20, 2015 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-26206951

RÉSUMÉ

IL-17-producing cells are important mediators of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Here we demonstrate that a distinct CD8(+) Tc17 population develops rapidly after SCT but fails to maintain lineage fidelity such that they are unrecognizable in the absence of a fate reporter. Tc17 differentiation is dependent on alloantigen presentation by host dendritic cells (DCs) together with IL-6. Tc17 cells express high levels of multiple prototypic lineage-defining transcription factors (eg, RORγt, T-bet) and cytokines (eg, IL-17A, IL-22, interferon-γ, granulocyte macrophage colony-stimulating factor, IL-13). Targeted depletion of Tc17 early after transplant protects from lethal acute GVHD; however, Tc17 cells are noncytolytic and fail to mediate graft-versus-leukemia (GVL) effects. Thus, the Tc17 differentiation program during GVHD culminates in a highly plastic, hyperinflammatory, poorly cytolytic effector population, which we term "inflammatory iTc17" (iTc17). Because iTc17 cells mediate GVHD without contributing to GVL, therapeutic inhibition of iTc17 development in a clinical setting represents an attractive approach for separating GVHD and GVL.


Sujet(s)
Lymphocytes T CD8+/anatomopathologie , Maladie du greffon contre l'hôte/anatomopathologie , Réaction du greffon contre la leucémie , Interleukine-17/immunologie , Transplantation de cellules souches/effets indésirables , Cellules Th17/anatomopathologie , Animaux , Transplantation de moelle osseuse/effets indésirables , Antigènes CD8/immunologie , Lymphocytes T CD8+/immunologie , Femelle , Maladie du greffon contre l'hôte/immunologie , Humains , Souris de lignée BALB C , Souris de lignée C57BL , Cellules Th17/immunologie
20.
Blood ; 125(19): 2933-6, 2015 May 07.
Article de Anglais | MEDLINE | ID: mdl-25788702

RÉSUMÉ

Granulocyte colony-stimulating factor (G-CSF) is widely used clinically to prevent neutropenia after cytotoxic chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Autophagy, a process of cytoplasmic component recycling, maintains cellular homeostasis and protects the cell during periods of metabolic stress or nutrient deprivation. We have observed that G-CSF activates autophagy in neutrophils and HSCs from both mouse and human donors. Furthermore, G-CSF-induced neutrophil and HSC mobilization is impaired in the absence of autophagy. In contrast, autophagy is dispensable for direct HSC mobilization in response to the CXCR4 antagonist AMD3100. Altogether, these data demonstrate an important role for G-CSF in invoking autophagy within hematopoietic and myeloid cells and suggest that this pathway is critical for ensuring cell survival in response to clinically relevant cytokine-induced stress. These findings have direct relevance to HSC transplantation and the increasing clinical use of agents that modulate autophagy.


Sujet(s)
Autophagie , Facteur de stimulation des colonies de granulocytes/pharmacologie , Mobilisation de cellules souches hématopoïétiques , Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Animaux , Agents antiVIH/pharmacologie , Antigènes CD34/génétique , Antigènes CD34/métabolisme , Protéine-5 associée à l'autophagie , Benzylamines , Technique de Western , Cellules cultivées , Cyclames , Cytométrie en flux , Cellules souches hématopoïétiques/anatomopathologie , Composés hétérocycliques/pharmacologie , Humains , Souris , Souris knockout , Protéines associées aux microtubules/physiologie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/anatomopathologie , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteurs CXCR4/antagonistes et inhibiteurs , RT-PCR , Transplantation autologue
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE