Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
2.
J Cell Biol ; 222(11)2023 11 06.
Article in English | MEDLINE | ID: mdl-37672657

ABSTRACT

Stress granules (SGs) are formed in the cytoplasm in response to various toxic agents and are believed to play a critical role in the regulation of mRNA metabolism during stress. In SGs, mRNAs are stored in an abortive translation initiation complex that can be routed to either translation initiation or degradation. Here, we show that G3BP, a phosphorylation-dependent endoribonuclease that interacts with RasGAP, is recruited to SGs in cells exposed to arsenite. G3BP may thus determine the fate of mRNAs during cellular stress. Remarkably, SG assembly can be either dominantly induced by G3BP overexpression, or on the contrary, inhibited by expressing a central domain of G3BP. This region binds RasGAP and contains serine 149 whose dephosphorylation is induced by arsenite treatment. Critically, a non-phosphorylatable G3BP mutant (S149A) oligomerizes and assembles SG. These results suggest that G3BP is an effector of SG assembly and that Ras signaling contributes to this process by regulating G3BP dephosphorylation.


Subject(s)
Endoribonucleases , Stress Granules , ras GTPase-Activating Proteins , Arsenites/pharmacology , Endoribonucleases/genetics , GTPase-Activating Proteins/genetics , RNA, Messenger/genetics
4.
Clin Transl Gastroenterol ; 14(4): e00560, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36573890

ABSTRACT

Advanced therapies have transformed the treatment of inflammatory bowel disease; however, many patients fail to respond, highlighting the need for therapies tailored to the underlying cell and molecular disease drivers. The first-in-class oral molecule ABX464 (obefazimod), which selectively upregulates miR-124, has demonstrated its ability to be a well-tolerated treatment with rapid and sustained efficacy in patients with ulcerative colitis (UC). Here, we provide evidence that ABX464 affects the immune system in vitro , in the murine model of inflammatory bowel disease, and in patients with UC. In vitro , ABX464 treatment upregulated miR-124 and led to decreases in proinflammatory cytokines including interleukin (IL) 17 and IL6, and in the chemokine CCL2. Consistently, miR-124 expression was upregulated in the rectal biopsies and blood samples of patients with UC, and a parallel reduction in Th17 cells and IL17a levels was observed in serum samples. In a mouse model of induced intestinal inflammation with dextran sulfate sodium, ABX464 reversed the increases in multiple proinflammatory cytokines in the colon and the upregulation of IL17a secretion in the mesenteric lymph nodes. By upregulating miR-124, ABX464 acts as "a physiological brake" of inflammation, which may explain the efficacy of ABX464 with a favorable tolerability and safety profile in patients with UC.


Subject(s)
Colitis, Ulcerative , Inflammatory Bowel Diseases , MicroRNAs , Humans , Animals , Mice , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , MicroRNAs/genetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Cytokines/metabolism , Inflammation
5.
Drug Discov Today ; 26(4): 1030-1039, 2021 04.
Article in English | MEDLINE | ID: mdl-33387693

ABSTRACT

Inflammatory diseases are believed to develop as a result of dysregulated inflammatory responses to environmental factors on susceptible genetic backgrounds. Operating at the level of post-transcriptional gene regulation, miRNAs are a class of endogenous, small noncoding RNAs that can promote downregulation of protein expression by translational repression and/or mRNA degradation of target mRNAs involved in inflammation. MiR-124 is a crucial modulator of inflammation and innate immunity that could provide therapeutic restitution of physiological pathways lost in inflammatory diseases. A recently discovered small quinoline, ABX464, was shown to upregulate miR-124 in human immune cells. In vivo, in a proof-of-concept clinical study, ABX464 showed robust and consistent efficacy in ulcerative colitis (UC). In this review, we examine the current therapeutic options proposed for UC and discuss the drug candidate ABX464 in this context.


Subject(s)
Colitis, Ulcerative , MicroRNAs/immunology , Quinolines/pharmacology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/therapy , Gene Expression Regulation/drug effects , Humans , Immunologic Factors/pharmacology , Inflammation/drug therapy , Inflammation/genetics
6.
EMBO J ; 40(3): e104895, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33320356

ABSTRACT

The Hippo signaling pathway is a major regulator of organ growth, which controls the activity of the transcription coactivator Yorkie (Yki) in Drosophila and its homolog YAP in mammals. Both Yki and YAP proteins exist as alternatively spliced isoforms containing either one or two WW domains. The biological importance of this conserved alternative splicing event is unknown. Here, we identify the splicing factor B52 as a regulator of yki alternative splicing in Drosophila and show that B52 modulates growth in part through modulation of yki alternative splicing. Yki isoforms differ by their transcriptional activity as well as their ability to bind and bridge PPxY motifs-containing partners, and can compete in vivo. Strikingly, flies in which yki alternative splicing has been abrogated, thus expressing only Yki2 isoform, exhibit fluctuating wing asymmetry, a signal of developmental instability. Our results identify yki alternative splicing as a new level of modulation of the Hippo pathway, that is required for growth equilibration during development. This study provides the first demonstration that the process of alternative splicing contributes to developmental robustness.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Splicing Factors/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Alternative Splicing , Animals , Cell Line , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Nuclear Proteins/chemistry , Protein Binding , Protein Domains , RNA Splicing Factors/genetics , Sequence Analysis, RNA , Trans-Activators/chemistry , Wings, Animal/growth & development , Wings, Animal/metabolism , YAP-Signaling Proteins
7.
J Clin Invest ; 131(4)2021 02 15.
Article in English | MEDLINE | ID: mdl-33332285

ABSTRACT

To clarify the function of cyclin A2 in colon homeostasis and colorectal cancer (CRC), we generated mice deficient for cyclin A2 in colonic epithelial cells (CECs). Colons of these mice displayed architectural changes in the mucosa and signs of inflammation, as well as increased proliferation of CECs associated with the appearance of low- and high-grade dysplasias. The main initial events triggering those alterations in cyclin A2-deficient CECs appeared to be abnormal mitoses and DNA damage. Cyclin A2 deletion in CECs promoted the development of dysplasia and adenocarcinomas in a murine colitis-associated cancer model. We next explored the status of cyclin A2 expression in clinical CRC samples at the mRNA and protein levels and found higher expression in tumors of patients with stage 1 or 2 CRC compared with those of patients with stage 3 or 4 CRC. A meta-analysis of 11 transcriptome data sets comprising 2239 primary CRC tumors revealed different expression levels of CCNA2 (the mRNA coding for cyclin A2) among the CRC tumor subtypes, with the highest expression detected in consensus molecular subtype 1 (CMS1) and the lowest in CMS4 tumors. Moreover, we found high expression of CCNA2 to be a new, independent prognosis factor for CRC tumors.


Subject(s)
Colon/metabolism , Colorectal Neoplasms/metabolism , Cyclin A2/metabolism , Homeostasis , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Animals , Colon/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclin A2/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Staging , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Prognosis
8.
RNA Biol ; 17(6): 857-871, 2020 06.
Article in English | MEDLINE | ID: mdl-32150510

ABSTRACT

Alternative splicing (AS) plays a central role during cell-fate determination. However, how the core spliceosomal factors (CSFs) are involved in this process is poorly understood. Here, we report the down-regulation of the U2AF1 CSF during stem cell differentiation. To investigate its function in stemness and differentiation, we downregulated U2AF1 in human induced pluripotent stem cells (hiPSCs), using an inducible-shRNA system, to the level found in differentiated ectodermal, mesodermal and endodermal cells. RNA sequencing and computational analysis reveal that U2AF1 down-regulation modulates the expression of development-regulating genes and regulates transcriptional networks involved in cell-fate determination. Furthermore, U2AF1 down-regulation induces a switch in the AS of transcription factors (TFs) required to establish specific cell lineages, and favours the splicing of a differentiated cell-specific isoform of DNMT3B. Our results showed that the differential expression of the core spliceosomal factor U2AF1, between stem cells and the precursors of the three germ layers regulates a cell-type-specific alternative splicing programme and a transcriptional network involved in cell-fate determination via the modulation of gene expression and alternative splicing of transcription regulators.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , RNA Splicing , Splicing Factor U2AF/metabolism , Alternative Splicing , Cell Differentiation/genetics , Cell Line , Cell Self Renewal/genetics , Computational Biology/methods , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Models, Biological , RNA, Messenger/genetics , Spliceosomes/metabolism
9.
Diabetes ; 69(4): 647-660, 2020 04.
Article in English | MEDLINE | ID: mdl-32005707

ABSTRACT

Aging-dependent changes in tissue function are associated with the development of metabolic diseases. However, the molecular connections linking aging, obesity, and diabetes remain unclear. Lamin A, lamin C, and progerin, products of the Lmna gene, have antagonistic functions on energy metabolism and life span. Lamin C, albeit promoting obesity, increases life span, suggesting that this isoform is crucial for maintaining healthy conditions under metabolic stresses. Because ß-cell loss during obesity or aging leads to diabetes, we investigated the contribution of lamin C to ß-cell function in physiopathological conditions. We demonstrate that aged lamin C only-expressing mice (Lmna LCS/LCS ) become obese but remain glucose tolerant due to adaptive mechanisms including increased ß-cell mass and insulin secretion. Triggering diabetes in young mice revealed that Lmna LCS/LCS animals normalize their fasting glycemia by both increasing insulin secretion and regenerating ß-cells. Genome-wide analyses combined to functional analyses revealed an increase of mitochondrial biogenesis and global translational rate in Lmna LCS/LCS islets, two major processes involved in insulin secretion. Altogether, our results demonstrate for the first time that the sole expression of lamin C protects from glucose intolerance through a ß-cell-adaptive transcriptional program during metabolic stresses, highlighting Lmna gene processing as a new therapeutic target for diabetes treatment.


Subject(s)
Aging/metabolism , Diabetes Mellitus/metabolism , Glucose Intolerance/metabolism , Insulin-Secreting Cells/metabolism , Lamin Type A/metabolism , Obesity/metabolism , Aging/genetics , Animals , Blood Glucose/metabolism , Diabetes Mellitus/genetics , Energy Metabolism/physiology , Glucagon/metabolism , Glucose Intolerance/genetics , Insulin/metabolism , Lamin Type A/genetics , Mice , Mice, Transgenic , Obesity/genetics , Pancreas/metabolism
10.
J Cell Biol ; 218(7): 2433-2434, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31171633

ABSTRACT

In this issue, Panas et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201801214) challenge the data published in the Tourrière et al. (2013. J. Cell Biol. https://doi.org/10.1083/jcb.200212128) paper on the role of G3BP phosphorylation in stress granule (SG) assembly. This reply addresses that letter and suggests that more work is needed to understand the role of this modification in SG formation.


Subject(s)
DNA Helicases , ras GTPase-Activating Proteins , Endoribonucleases , Phosphorylation , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins
11.
J Virus Erad ; 5(1): 10-22, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30800421

ABSTRACT

OBJECTIVES: To assess the safety and tolerability as well as antiretroviral impact of ABX464, an oral investigational drug with a novel mechanism of HIV-1 inhibition (ClinicalTrials.gov NCT02735863). METHODS: Randomised, double-blind, placebo-controlled, Phase IIa study in individuals living with HIV-1 on antiretroviral therapy at six clinical centres in Spain, France and Belgium. ABX464 was administered once a day to 22 fully controlled HIV-1-positive participants at two doses (50 mg, n=6 and 150 mg, n=16) versus placebo, which was given to eight participants for 28 days in combination with a boosted protease inhibitor (darunavir/ritonavir or darunavir/cobicistat). The primary objective of the study was to assess ABX464 safety and tolerability when used in combination with darunavir boosted therapy. The secondary objective was to study antiretroviral efficacy on viral reservoirs using time to viral rebound following treatment interruption. The impact of ABX464 on HIV-1 reservoirs was further assessed by measuring levels of total HIV-1 in peripheral blood mononuclear cells (PBMCs) in the intervention arm versus placebo. A positive response was defined as an absolute reduction in HIV-1 DNA of at least 50 copies/106 PBMCs and a relative decrease >25% of HIV-1 DNA level. RESULTS: Twenty-six of the 30 randomly allocated participants completed the study according to the study protocol. ABX464 was found to be safe and well tolerated with the majority of adverse events (AEs) being mild or moderate. Of the participants, 22 (73.3%) experienced treatment-associated AEs (93.8%, 66.7%, 37.5% in the ABX464 150-mg, 50-mg dose and placebo arms, respectively). Percentages for combined grade 3/4 AEs for the three arms were 6.3%, 0% and 12.5%, respectively. Median time (Kaplan-Meier estimates) to viral rebound for ABX464 150-mg, 50-mg and placebo arms were 12.0 (95% confidence interval [CI]: 10-15), 15.5 (95% CI 14-22) and 15.5 (95% CI 1-22) days, respectively with no significant difference between the 150-mg treatment arm and placebo. Median changes in total HIV-1 DNA copies/106 PBMCs for ABX464 150-mg, 50-mg and placebo arms after 28 days of treatment were -40 (range -434 to +194), -115 (range -116 to -114) and 25 (range -35 to +218), respectively, showing a decrease in the intervention arms. There were 6/14, 2/2, and 0/4 responders for ABX464 150 mg, 50 mg and placebo, respectively. No significant difference was seen between treatment arms and placebo with respect to these virological parameters. CONCLUSIONS: This small controlled study confirmed the good safety and tolerability of ABX464 and provides some evidence of a potential reduction of the HIV-1 reservoir in terms of HIV-1 DNA levels in PBMCs when it was added to an HIV-1 protease inhibitor-based regimen. These results will need to be confirmed in a larger study.

12.
Sci Rep ; 9(1): 792, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692590

ABSTRACT

ABX464 is a first-in-class, clinical-stage, small molecule for oral administration that has shown strong anti-inflammatory effects in the DSS-model for inflammatory bowel disease (IBD) and also prevents replication of the HIV virus. ABX464 which binds to cap binding complex (CBC) has demonstrated safety and efficacy in a phase 2a proof-of-concept clinical trial in patients with Ulcerative colitis. Previously, with limited technologies, it was not possible to quantify the effect of ABX464 on viral and cellular RNA biogenesis. Here, using RNA CaptureSeq and deep sequencing, we report that ABX464 enhances the splicing of HIV RNA in infected PBMCs from six healthy individuals and also the expression and splicing of a single long noncoding RNA to generate the anti-inflammatory miR-124 both ex vivo and in HIV patients. While ABX464 has no effect on pre-mRNA splicing of cellular genes, depletion of CBC complex by RNAi leads to accumulation of intron retention transcripts. These results imply that ABX464 did not inhibit the function of CBC in splicing but rather strengthens it under pathological condition like inflammation and HIV infection. The specific dual ability of ABX464 to generate both anti-inflammatory miR-124 and spliced viral RNA may have applicability for the treatment of both inflammatory diseases and HIV infection.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Leukocytes, Mononuclear/cytology , Quinolines/pharmacology , RNA Splicing/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , HIV/drug effects , HIV/genetics , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , MicroRNAs/drug effects , MicroRNAs/genetics , RNA, Viral/drug effects , RNA, Viral/genetics , Sequence Analysis, RNA
13.
Sci Data ; 4: 170150, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29039845

ABSTRACT

RNA-Seq enables the generation of extensive transcriptome information providing the capability to characterize transcripts (including alternative isoforms and polymorphism), to quantify expression and to identify differential regulation in a single experiment. To reveal the capacity of new anti-HIV ABX464 candidate in modulating the expression of genes, datasets were generated and validated using RNA-seq approach. This comprehensive dataset will be useful to deepen the comprehensive understanding of the progression of human immunodeficiency virus (HIV) associated with mucosal damage in the gastrointestinal (GI) tract and subsequent inflammation, providing an opportunity to generate new therapies, diagnoses, and preventive strategies.


Subject(s)
Anti-HIV Agents/adverse effects , Macrophage Activation/drug effects , Macrophage Activation/genetics , Quinolines/adverse effects , Gastroenteritis/complications , Gastroenteritis/drug therapy , HIV Infections/complications , HIV Infections/drug therapy , Humans , Sequence Analysis, RNA
14.
AIDS ; 31(18): 2443-2454, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28926402

ABSTRACT

OBJECTIVE: In this study, we looked for a new family of latency reversing agents. DESIGN: We searched for G-protein-coupled receptors (GPCR) coexpressed with the C-C chemokine receptor type 5 (CCR5) in primary CD4 T cells that activate infected cells and boost HIV production. METHODS: GPCR coexpression was unveiled by reverse transcriptase-PCR. We used fluorescence resonance energy transfer to analyze the dimerization with CCR5 of the expressed GPCR. Viral entry was measured by flow cytometry, reverse transcription by quantitative PCR, nuclear factor-kappa B translocation by immunofluorescence, long terminal repeat activation using a gene reporter assay and viral production by p24 quantification. RESULTS: Gαi-coupled sphingosine-1-phophate receptor 1 (S1P1) is highly coexpressed with CCR5 on primary CD4 T cells and dimerizes with it. The presence of S1P1 had major effects neither on viral entry nor on reverse transcription. Yet, S1P1 signaling induced NFκB activation, boosting the expression of the HIV LTR. Consequently, in culture medium containing sphingosine-1-phophate, the presence of S1P1 enhanced the replication of a CCR5-, but also of a CXCR4-using HIV-1 strain. The S1P1 ligand FTY720, a drug used in multiple sclerosis treatment, inhibited HIV-1 productive infection of monocyte-derived dendritic cells and of severe combined immunodeficiency mice engrafted with human peripheral blood mononuclear cells. Conversely, S1P1 agonists were able to force latently infected peripheral blood mononuclear cells and lymph node cells to produce virions in vitro. CONCLUSION: Altogether these data indicate that the presence of S1P1 facilitates HIV-1 replicative cycle by boosting viral genome transcription, S1P1 antagonists have anti-HIV effects and S1P1 agonists are HIV latency reversing agents.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Signal Transduction , Virus Latency/drug effects , Virus Replication/drug effects , Animals , Cells, Cultured , Gene Expression Profiling , HIV-1/growth & development , Humans , Mice, SCID , Receptors, CCR5/biosynthesis , Receptors, Lysosphingolipid/biosynthesis
15.
Sci Rep ; 7(1): 4860, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28687795

ABSTRACT

The progression of human immunodeficiency virus (HIV) is associated with mucosal damage in the gastrointestinal (GI) tract. This damage enables bacterial translocation from the gut and leads to subsequent inflammation. Dextran sulfate sodium (DSS-exposure) is an established animal model for experimental colitis that was recently shown to recapitulate the link between GI-tract damage and pathogenic features of SIV infection. The current study tested the protective properties of ABX464, a first-in-class anti-HIV drug candidate currently in phase II clinical trials. ABX464 treatment strongly attenuated DSS-induced colitis in mice and produced a long-term protection against prolonged DSS-exposure after drug cessation. Consistently, ABX464 reduced the colonic production of the inflammatory cytokines IL-6 and TNFα as well as that of the chemoattractant MCP-1. However, RNA profiling analysis revealed the capacity of ABX464 to induce the expression of IL-22, a cytokine involved in colitis tissue repair, both in DSS-treated mice and in LPS-stimulated bone marrow-derived macrophages. Importantly, anti-IL-22 antibodies significantly reduced the protective effect of ABX464 on colitis in DSS-treated mice. Because reduced IL-22 production in the gut mucosa is an established factor of HIV and DSS-induced immunopathogenesis, our data suggest that the anti-inflammatory properties of ABX464 warrant exploration in both HIV and inflammatory ulcerative colitis (UC) disease.


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Colitis/drug therapy , Interleukins/metabolism , Macrophages/immunology , Quinolines/administration & dosage , Animals , Anti-HIV Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/pathology , Cytokines/analysis , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Gene Expression Profiling , Interleukins/genetics , Intestinal Mucosa/pathology , Macrophages/drug effects , Mice , Quinolines/pharmacology , Treatment Outcome , Interleukin-22
16.
Article in English | MEDLINE | ID: mdl-28507108

ABSTRACT

We investigated the safety and antiviral effects of an anti-HIV compound (ABX464) with a unique mechanism of viral replication inhibition. This was a randomized, double-blind, placebo-controlled, dose-ranging study in treatment-naive HIV-infected patients. Participants were assigned to eight groups; each group included eight subjects receiving either the study compound, ABX464 (n = 6), or the corresponding placebo (n = 2), according to a randomization code. The first dose administered was 25 mg, given once or 3 times a day over a 2- to 3-week period. Ascending doses of up to 150 mg were delivered after review of the safety data. The primary objective of the study was to assess the safety and tolerability of ABX464 after repeated oral administrations in subjects infected by HIV. Sixty-six subjects were enrolled and were randomized. Sixty-three subjects completed the study according to the study protocol. Twenty-one adverse events (AEs) were reported by 7 subjects out of 16 (44%) who received placebo, and 158 AEs were reported by 39 subjects out of 50 (78%) who received the study drug. In the ABX464 treatment group, all of these adverse events were mild to moderate. No subjects discontinued treatment due to drug-related AEs. Administration of ABX464 at up to 150 mg once a day was safe and well tolerated in HIV-infected subjects. An efficacy signal with respect to a reduction of the viral load by ABX464 was detected, mainly in subjects treated at the highest dose. Further studies will be required to demonstrate antiviral effects in HIV-infected subjects in combination with other antiretroviral therapies. (This study is registered on the ClinicalTrials.gov website under registration no. NCT02452242.).


Subject(s)
Anti-HIV Agents/therapeutic use , HIV-1/drug effects , HIV-1/pathogenicity , Quinolines/therapeutic use , Adolescent , Adult , Aged , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Double-Blind Method , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Quinolines/adverse effects , Quinolines/pharmacokinetics , Treatment Outcome , Viral Load/drug effects , Virus Replication/drug effects , Young Adult
17.
J Antimicrob Chemother ; 72(3): 820-828, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27999038

ABSTRACT

Background: An anti-HIV compound (ABX464) has been developed with a novel mechanism of activity in that it blocks viral gene expression in cells that are already infected. Objectives: A first-in-man study was conducted to determine the pharmacokinetic and safety profiles of ABX464. This was carried out as an open label, parallel group, single ascending dose, exploratory study. Methods: Twenty-four male subjects in good health without HIV infection, aged from 18 to 55 years old, with BMIs of 18-27 kg/m 2 were included. A single oral dose of ABX464 (50, 100, 150 or 200 mg) was administered on the morning of day 0 after overnight fasting, with follow-up for 45 days. Safety assessments consisted of vital signs, electrocardiogram, physical examination, laboratory tests and urinalysis. Pharmacokinetic parameters were calculated for ABX464 and its main metabolite ABX-464- N -glucuronide (ABX464-NGlc). The study was registered at https://www.clinicaltrials (trial number NCT02792686). Results: ABX464 was well tolerated; the most frequent related treatment-emergent adverse events were headaches, nausea and vomiting; they were not considered as treatment-limiting effects. ABX464's C max was observed approximately 2 h after administration in all groups. ABX464 was rapidly and substantially metabolized into ABX464-NGlc. The C max of ABX464-NGlc was observed approximately 4 h post-dose and was about 160-fold higher than that of the parent with a much longer t 1/2 (90-110 h). The ratio of metabolite to parent drug was consistent across the complete dose range. Conclusions: These studies confirmed that ABX464 is well tolerated and rapidly and substantially metabolized into ABX464-NGlc in human subjects.


Subject(s)
Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Quinolines/adverse effects , Quinolines/pharmacokinetics , Adolescent , Adult , Dose-Response Relationship, Drug , Electrocardiography , Healthy Volunteers , Humans , Male , Middle Aged , Urinalysis
18.
Article in English | MEDLINE | ID: mdl-27799203

ABSTRACT

ABX464 is an antiviral that provides a novel approach to the reduction and control of HIV infection. Investigation of food influence is important in the optimization of treatment. An open-label, food effect, randomized study which included 2 groups of 24 subjects each was carried out to assess the bioavailability and safety of single (group 1) and repeated (group 2) oral doses of ABX464 (50 mg) under fed or fasted conditions. The maximum concentration (Cmax) and the area under the concentration-time curve from time zero to infinity (AUC0-∞) of ABX464 were demonstrated to increase with food after a single dose of ABX464 (219% and 188%, respectively). The apparent terminal elimination half-lives (t1/2s) under fed and fasted conditions were comparable, at about 0.80 h. The median time to maximum concentration (Tmax) was delayed from 1.5 to 2.8 h, and the ratio of the AUC0-∞ obtained under fed conditions to the AUC0-∞ obtained under fasted conditions (Frel) was 2.69. Comparable results were obtained on day 1 and day 10 in group 2. The increases in Cmax and AUC0-∞ of the metabolite ABX464-N-glucuronide (ABX464-NGlc) were, however, much more limited when ABX464 was given with food. The t1/2s were also comparable under the two conditions (around 100 h). Between day 1 and day 10, the Cmax increased by 5% under the fasted condition and by 25% under the fed condition. The most common related treatment-emergent adverse events were headaches, vomiting, and nausea. It was concluded that food has a significant impact on the levels of ABX464 in plasma with a delay in absorption and increased relative bioavailability, with a lesser impact on its biotransformation into ABX464-NGlc. ABX464 was well tolerated under both fasted and fed conditions. (This study has been registered at ClinicalTrials.gov under registration no. NCT02731885.).


Subject(s)
Antiviral Agents/therapeutic use , Glucuronides/therapeutic use , Administration, Oral , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Body Mass Index , Food-Drug Interactions , Glucuronides/administration & dosage , Glucuronides/chemistry , HIV Infections/drug therapy , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
19.
J Neurochem ; 139(3): 349-368, 2016 11.
Article in English | MEDLINE | ID: mdl-27513819

ABSTRACT

Neuronal granules play an important role in the localization and transport of translationally silenced messenger ribonucleoproteins in neurons. Among the factors associated with these granules, the RNA-binding protein G3BP1 (stress-granules assembly factor) is involved in neuronal plasticity and is induced in Alzheimer's disease. We immunopurified a stable complex containing G3BP1 from mouse brain and performed high-throughput sequencing and cross-linking immunoprecipitation to identify the associated RNAs. The G3BP-complex contained the deubiquitinating protease USP10, CtBP1 and the RNA-binding proteins Caprin-1, G3BP2a and splicing factor proline and glutamine rich, or PSF. The G3BP-complex binds preferentially to transcripts that retain introns, and to non-coding sequences like 3'-untranslated region and long non-coding RNAs. Specific transcripts with retained introns appear to be enriched in the cerebellum compared to the rest of the brain and G3BP1 depletion decreased this intron retention in the cerebellum of G3BP1 knockout mice. Among the enriched transcripts, we found an overrepresentation of genes involved in synaptic transmission, especially glutamate-related neuronal transmission. Notably, G3BP1 seems to repress the expression of the mature Grm5 (metabotropic glutamate receptor 5) transcript, by promoting the retention of an intron in the immature transcript in the cerebellum. Our results suggest that G3BP is involved in a new functional mechanism to regulate non-coding RNAs including intron-retaining transcripts, and thus have broad implications for neuronal gene regulation, where intron retention is widespread.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/metabolism , Cerebellum/metabolism , Introns/genetics , 3' Untranslated Regions/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cross-Linking Reagents , Cytoplasmic Granules/metabolism , DNA Helicases , DNA-Binding Proteins/metabolism , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Knockout , Poly-ADP-Ribose Binding Proteins , RNA/biosynthesis , RNA/genetics , RNA Helicases , RNA Recognition Motif Proteins , RNA, Long Noncoding/genetics , Transcription, Genetic , Ubiquitin Thiolesterase/metabolism
20.
Retrovirology ; 12: 30, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25889234

ABSTRACT

BACKGROUND: Current therapies have succeeded in controlling AIDS pandemic. However, there is a continuing need for new drugs, in particular those acting through new and as yet unexplored mechanisms of action to achieve HIV infection cure. We took advantage of the unique feature of proviral genome to require both activation and inhibition of splicing of viral transcripts to develop molecules capable of achieving long lasting effect on viral replication in humanized mouse models through inhibition of Rev-mediated viral RNA biogenesis. RESULTS: Current HIV therapies reduce viral load during treatment but titers rebound after treatment is discontinued. We devised a new drug that has a long lasting effect after viral load reduction. We demonstrate here that ABX464 compromises HIV replication of clinical isolates of different subtypes without selecting for drug resistance in PBMCs or macrophages. ABX464 alone, also efficiently compromised viral proliferation in two humanized mouse models infected with HIV that require a combination of 3TC, Raltegravir and Tenofovir (HAART) to achieve viral inhibition in current protocols. Crucially, while viral load increased dramatically just one week after stopping HAART treatment, only slight rebound was observed following treatment cessation with ABX464 and the magnitude of the rebound was maintained below to that of HAART for two months after stopping the treatment. Using a system to visualize single HIV RNA molecules in living cells, we show that ABX464 inhibits viral replication by preventing Rev-mediated export of unspliced HIV-1 transcripts to the cytoplasm and by interacting with the Cap Binding Complex (CBC). Deep sequencing of viral RNA from treated cells established that retained viral RNA is massively spliced but importantly, normal cellular splicing is unaffected by the drug. Consistently ABX464 is non-toxic in humans and therefore represents a promising complement to current HIV therapies. CONCLUSIONS: ABX464 represents a novel class of anti-HIV molecules with unique properties. ABX464 has a long lasting effect in humanized mice and neutralizes the expression of HIV-1 proviral genome of infected immune cells including reservoirs and it is therefore a promising drug toward a functional cure of HIV.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/isolation & purification , Viral Load , Adult , Animals , Anti-HIV Agents/pharmacology , Disease Models, Animal , HIV-1/drug effects , Humans , Mice, SCID , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...