Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
2.
Cancers (Basel) ; 12(3)2020 Mar 18.
Article En | MEDLINE | ID: mdl-32197329

Sterile alpha motif and histidine-aspartic acid domain-containing protein 1 (SAMHD1) is a dNTP triphosphohydrolase involved in the regulation of the intracellular dNTP pool, linked to viral restriction, cancer development and autoimmune disorders. SAMHD1 function is regulated by phosphorylation through a mechanism controlled by cyclin-dependent kinases and tightly linked to cell cycle progression. Recently, SAMHD1 has been shown to decrease the efficacy of nucleotide analogs used as chemotherapeutic drugs. Here, we demonstrate that SAMHD1 can enhance or decrease the efficacy of various classes of anticancer drug, including nucleotide analogues, but also anti-folate drugs and CDK inhibitors. Importantly, we show that selective CDK4/6 inhibitors are pharmacological activators of SAMHD1 that act by inhibiting its inactivation by phosphorylation. Combinations of a CDK4/6 inhibitor with nucleoside or folate antimetabolites potently enhanced drug efficacy, resulting in highly synergic drug combinations (CI < 0.04). Mechanistic analyses reveal that cell cycle-controlled modulation of SAMHD1 function is the central process explaining changes in anticancer drug efficacy, therefore providing functional proof of the potential of CDK4/6 inhibitors as a new class of adjuvants to boost chemotherapeutic regimens. The evaluation of SAMHD1 expression in cancer tissues allowed for the identification of cancer types that would benefit from the pharmacological modulation of SAMHD1 function. In conclusion, these results indicate that the modulation of SAMHD1 function may represent a promising strategy for the improvement of current antimetabolite-based treatments.

3.
Antiviral Res ; 168: 18-27, 2019 08.
Article En | MEDLINE | ID: mdl-31077767

Midostaurin is a multi-kinase inhibitor with antineoplastic activity. We assessed the capacity of midostaurin to affect early and late steps of HIV-1 infection and to reactivate HIV-1 latently infected cells, alone or in combination with histone deacetylase inhibitors (HDACi) known to act as latency-reversing agents (LRA). Acute HIV-1 infection was assessed by flow cytometry in three cell types treated with midostaurin in the presence or absence of SAMHD1. Non-infected cells were treated with midostaurin and harvested for Western blot analysis. Macrophage infections were also measured by quantitative RT-PCR. HIV-1 latency reactivation was assessed in several latency models. Midostaurin induced G2/M arrest and inhibited CDK2, preventing the phosphorylation of SAMHD1 associated to inhibition of its dNTPase activity. In the presence of SAMHD1, midostaurin blocked HIV-1 DNA formation and viral replication. However, following Vpx-mediated SAMHD1 degradation, midostaurin increased viral transcripts and virus replication. In three out of four HIV-1 latency models, including primary CD4+ T cells, midostaurin effectively reversed HIV-1 latency and was synergistic in combination with LRA vorinostat and panobinostat. Our study describes a dual effect for midostaurin in HIV-1 infection, antiviral or proviral depending on SAMHD1 activation, and highlights a role for active SAMHD1 in regulating the activity of potential HIV-1 latency reversal agents.


HIV Infections/virology , HIV-1/drug effects , Protein Kinase Inhibitors/pharmacology , Staurosporine/analogs & derivatives , Virus Latency/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Cell Cycle Checkpoints/drug effects , Cells, Cultured , HIV-1/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Macrophages/drug effects , Macrophages/virology , Phosphorylation/drug effects , SAM Domain and HD Domain-Containing Protein 1/antagonists & inhibitors , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/metabolism , Staurosporine/pharmacology , Virus Activation/drug effects , Virus Replication/drug effects
4.
Nat Commun ; 9(1): 2739, 2018 07 16.
Article En | MEDLINE | ID: mdl-30013105

CD32 has been shown to be preferentially expressed in latently HIV-1-infected cells in an in vitro model of quiescent CD4 T cells. Here we show that stimulation of CD4+ T cells with IL-2, IL-7, PHA, and anti-CD3/CD28 antibodies induces T-cell proliferation, co-expression of CD32 and the activation of the markers HLA-DR and CD69. HIV-1 infection increases CD32 expression. 79.2% of the CD32+/CD4+ T cells from HIV+ individuals under antiretroviral treatment were HLA-DR+. Resting CD4+ T cells infected in vitro generally results in higher integration of provirus. We observe no difference in provirus integration or replication-competent inducible latent HIV-1 in CD32+ or CD32- CD4+ T cells from HIV+ individuals. Our results demonstrate that CD32 expression is a marker of CD4+ T cell activation in HIV+ individuals and raises questions regarding the immune resting status of CD32+ cells harboring HIV-1 proviruses.


CD4-Positive T-Lymphocytes/immunology , HIV Infections/genetics , HIV-1/immunology , Host-Pathogen Interactions/immunology , Lymphocyte Activation/genetics , Receptors, IgG/genetics , Adult , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Gene Expression , HIV Infections/immunology , HIV Infections/virology , HIV-1/genetics , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Interleukin-2/pharmacology , Interleukin-7/pharmacology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation/drug effects , Male , Middle Aged , Phytohemagglutinins/pharmacology , Primary Cell Culture , Proviruses/genetics , Proviruses/immunology , Receptors, IgG/immunology , Virus Integration
5.
Antiviral Res ; 156: 116-127, 2018 08.
Article En | MEDLINE | ID: mdl-29906476

The hepatitis C virus (HCV) is a globally prevalent infectious pathogen. As many as 80% of people infected with HCV do not control the virus and develop a chronic infection. Response to interferon (IFN) therapy is widely variable in chronic HCV infected patients, suggesting that HCV has evolved mechanisms to suppress and evade innate immunity responsible for its control and elimination. Adenosine deaminase acting on RNA 1 (ADAR1) is a relevant factor in the regulation of the innate immune response. The loss of ADAR1 RNA-editing activity and the resulting loss of inosine bases in RNA are critical in producing aberrant RLR-mediated innate immune response, mediated by RNA sensors MDA5 and RIG-I. Here, we describe ADAR1 role as a regulator of innate and antiviral immune function in HCV infection, both in vitro and in patients. Polymorphisms within ADAR1 gene were found significantly associated to poor clinical outcome to HCV therapy and advanced liver fibrosis in a cohort of HCV and HIV-1 coinfected patients. Moreover, ADAR1 knockdown in primary macrophages and Huh7 hepatoma cells enhanced IFN and IFN stimulated gene expression and increased HCV replication in vitro. Overall, our results demonstrate that ADAR1 regulates innate immune signaling and is an important contributor to the outcome of the HCV virus-host interaction. ADAR1 is a potential target to boost antiviral immune response in HCV infection.


Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Genetic Predisposition to Disease , Hepacivirus/immunology , Hepatitis C/immunology , Host-Pathogen Interactions , Immunity, Innate , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Cells, Cultured , Humans , Polymorphism, Genetic
6.
Sci Rep ; 7(1): 13339, 2017 10 17.
Article En | MEDLINE | ID: mdl-29042669

ADAR1-dependent A-to-I editing has recently been recognized as a key process for marking dsRNA as self, therefore, preventing innate immune activation and affecting the development and resolution of immune-mediated diseases and infections. Here, we have determined the role of ADAR1 as a regulator of innate immune activation and modifier of viral susceptibility in primary myeloid and lymphoid cells. We show that ADAR1 knockdown significantly enhanced interferon, cytokine and chemokine production in primary macrophages that function as antiviral paracrine factors, rendering them resistant to HIV-1 infection. ADAR1 knockdown induced deregulation of the RLRs-MAVS signaling pathway, by increasing MDA5, RIG-I, IRF7 and phospho-STAT1 expression, an effect that was partially rescued by pharmacological blockade of the pathway. In summary, our results demonstrate a role of ADAR1 in regulating innate immune function in primary macrophages, suggesting that macrophages may play an essential role in disease associated to ADAR1 dysfunction. We also show that viral inhibition is exclusively dependent on innate immune activation consequence of ADAR1 knockdown, pointing towards ADAR1 as a potential target to boost antiviral immune response.


Adenosine Deaminase/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Macrophages/immunology , Macrophages/metabolism , RNA Editing , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Gene Knockdown Techniques , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Interferon Type I/metabolism , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/virology , Signal Transduction , Virus Diseases/etiology , Virus Replication
7.
Article En | MEDLINE | ID: mdl-28874382

The persistence of HIV despite suppressive antiretroviral therapy is a major roadblock to HIV eradication. Current strategies focused on inducing the expression of latent HIV fail to clear the persistent reservoir, prompting the development of new approaches for killing HIV-positive cells. Recently, acitretin was proposed as a pharmacological enhancer of the innate cellular defense network that led to virus reactivation and preferential death of infected cells. We evaluated the capacity of acitretin to reactivate and/or to facilitate immune-mediated clearance of HIV-positive cells. Acitretin did not induce HIV reactivation in latently infected cell lines (J-Lat and ACH-2). We could observe only modest induction of HIV reactivation by acitretin in latently green fluorescent protein-HIV-infected Jurkat cells, comparable to suboptimal concentrations of vorinostat, a known latency-reversing agent (LRA). Acitretin induction was insignificant, however, compared to optimal concentrations of LRAs. Acitretin failed to reactivate HIV in a model of latently infected primary CD4+ T cells but induced retinoic acid-inducible gene I (RIG-I) and mitochondrial antiviral signaling (MAVS) expression in infected and uninfected cells, confirming the role of acitretin as an innate immune modulator. However, this effect was not associated with selective killing of HIV-positive cells. In conclusion, acitretin-mediated stimulation of the RIG-I pathway for HIV reactivation is modest and thus may not meaningfully affect the HIV reservoir. Stimulation of the RIG-I-dependent interferon (IFN) cascade by acitretin may not significantly affect the selective destruction of latently infected HIV-positive cells.


Acitretin/pharmacology , HIV Infections/immunology , Immunity, Innate/drug effects , Immunologic Factors/pharmacology , Virus Latency/drug effects , DEAD Box Protein 58/metabolism , HIV Infections/drug therapy , HIV-1/pathogenicity , HIV-1/physiology , Humans , Receptors, Immunologic , Signal Transduction/drug effects
...