Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
PLoS Pathog ; 20(7): e1012017, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39038029

ABSTRACT

Some respiratory viruses can cause a viral interference through the activation of the interferon (IFN) pathway that reduces the replication of another virus. Epidemiological studies of coinfections between SARS-CoV-2 and other respiratory viruses have been hampered by non-pharmacological measures applied to mitigate the spread of SARS-CoV-2 during the COVID-19 pandemic. With the ease of these interventions, SARS-CoV-2 and influenza A viruses can now co-circulate. It is thus of prime importance to characterize their interactions. In this work, we investigated viral interference effects between an Omicron variant and a contemporary influenza A/H3N2 strain, in comparison with an ancestral SARS-CoV-2 strain and the 2009 pandemic influenza A/H1N1 virus. We infected nasal human airway epitheliums with SARS-CoV-2 and influenza, either simultaneously or 24 h apart. Viral load was measured by RT-qPCR and IFN-α/ß/λ1/λ2 proteins were quantified by immunoassay. Expression of four interferon-stimulated genes (ISGs; OAS1/IFITM3/ISG15/MxA) was also measured by RT-droplet digital PCR. Additionally, susceptibility of each virus to IFN-α/ß/λ2 recombinant proteins was determined. Our results showed that influenza A, and especially A/H3N2, interfered with both SARS-CoV-2 viruses, but that SARS-CoV-2 did not significantly interfere with A/H3N2 or A/H1N1. Consistently with these results, influenza, and particularly the A/H3N2 strain, caused a higher production of IFN proteins and expression of ISGs than SARS-CoV-2. SARS-CoV-2 induced a marginal IFN production and reduced the IFN response during coinfections with influenza. All viruses were susceptible to exogenous IFNs, with the ancestral SARS-CoV-2 and Omicron being less susceptible to type I and type III IFNs, respectively. Thus, influenza A causes a viral interference towards SARS-CoV-2 most likely through an IFN response. The opposite is not necessarily true, and a concurrent infection with both viruses leads to a lower IFN response. Taken together, these results help us to understand how SARS-CoV-2 interacts with another major respiratory pathogen.

2.
Infect Dis Rep ; 16(1): 65-82, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38247977

ABSTRACT

Cytomegalovirus (CMV) infections may increase morbidity and mortality in immunocompromised patients. Until recently, standard antiviral drugs against CMV were limited to viral DNA polymerase inhibitors (val)ganciclovir, foscarnet and cidofovir with a risk for cross-resistance. These drugs may also cause serious side effects. This narrative review provides an update on new antiviral agents that were approved for the prevention and treatment of CMV infections in transplant recipients. Letermovir was approved in 2017 for CMV prophylaxis in CMV-seropositive adults who received an allogeneic hematopoietic stem cell transplant. Maribavir followed four years later, with an indication in the treatment of adult and pediatric transplant patients with refractory/resistant CMV disease. The target of letermovir is the CMV terminase complex (constituted of pUL56, pUL89 and pUL51 subunits). Letermovir prevents the cleavage of viral DNA and its packaging into capsids. Maribavir is a pUL97 kinase inhibitor, which interferes with the assembly of capsids and the egress of virions from the nucleus. Both drugs have activity against most CMV strains resistant to standard drugs and exhibit favorable safety profiles. However, high-level resistance mutations may arise more rapidly in the UL56 gene under letermovir than low-grade resistance mutations. Some mutations emerging in the UL97 gene under maribavir can be cross-resistant with ganciclovir. Thus, letermovir and maribavir now extend the drug arsenal available for the management of CMV infections and their respective niches are currently defined.

3.
Rev Med Virol ; 34(1): e2510, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38282407

ABSTRACT

Epidemic peaks of respiratory viruses that co-circulate during the winter-spring seasons can be synchronous or asynchronous. The occurrence of temporal patterns in epidemics caused by some respiratory viruses suggests that they could negatively interact with each other. These negative interactions may result from a programme of innate immune memory, known as trained immunity, which may confer broad protective effects against respiratory viruses. It is suggested that stimulation of innate immune cells by a vaccine or a pathogen could induce their long-term functional reprogramming through an interplay between metabolic and epigenetic changes, which influence the transcriptional response to a secondary challenge. During the coronavirus disease 2019 pandemic, the circulation of most respiratory viruses was prevented by non-pharmacological interventions and then resumed at unusual periods once sanitary measures were lifted. With time, respiratory viruses should find again their own ecological niches. This transition period provides an opportunity to study the interactions between respiratory viruses at the population level.


Subject(s)
COVID-19 , Vaccines , Viruses , Humans , Trained Immunity , Immunity, Innate
4.
J Alzheimers Dis ; 97(1): 171-178, 2024.
Article in English | MEDLINE | ID: mdl-38143354

ABSTRACT

Using APP/PS1 mice that overproduce amyloid-ß (Aß) peptides, we investigated whether intranasal infection with a neurovirulent clinical strain of herpes simplex virus 1 (HSV-1) before Aß deposition could accelerate or increase Alzheimer's disease-like pathology. After HSV-1 infection, APP/PS1 mice presented a similar disease as wild type animals based on body weight changes, clinical symptoms, and survival rates. The number and volume of Aß plaques, the number of microglia, and the percentages of circulating monocyte subsets were similar in APP/PS1 mice infected or not with HSV-1. Thus, intranasal infection with HSV-1 does not alter Aß pathology in this mouse model.


Subject(s)
Alzheimer Disease , Herpes Simplex , Herpesvirus 1, Human , Mice , Animals , Amyloid beta-Protein Precursor/genetics , Mice, Transgenic , Amyloid beta-Peptides , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Herpes Simplex/complications , Plaque, Amyloid/pathology , Disease Models, Animal , Presenilin-1/genetics
5.
Front Microbiol ; 13: 988058, 2022.
Article in English | MEDLINE | ID: mdl-36238591

ABSTRACT

[This corrects the article DOI: 10.3389/fmicb.2020.631736.].

6.
Antiviral Res ; 202: 105328, 2022 06.
Article in English | MEDLINE | ID: mdl-35490740

ABSTRACT

Letermovir (LTV) is approved for the prophylaxis of human cytomegalovirus (HCMV) infection in adult seropositive recipients of an allogeneic hematopoietic stem cell transplant. Here, we report on the in vitro activity of LTV against a large panel of clinical HCMV isolates and recombinant viruses with different drug susceptibility phenotypes to currently-approved DNA polymerase inhibitors or maribavir. No pre-existing mutations conferring resistance to LTV were detected by Sanger sequencing in clinical HCMV isolates susceptible or resistant to DNA polymerases inhibitors. The susceptibility of LTV against the different recombinant HCMV mutants with amino acid substitutions in the UL97 kinase or in the UL54 DNA polymerase was similar to that of the wild type virus. LTV was also effective against recombinant HCMV harboring UL97 mutations conferring resistance to maribavir.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Acetates , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cytomegalovirus/genetics , Drug Resistance, Viral/genetics , Ganciclovir/pharmacology , Humans , Mutation , Phenotype , Quinazolines
7.
Viruses ; 14(2)2022 02 15.
Article in English | MEDLINE | ID: mdl-35215988

ABSTRACT

The types of interactions between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory viruses are not well-characterized due to the low number of co-infection cases described since the onset of the pandemic. We have evaluated the interactions between SARS-CoV-2 (D614G mutant) and influenza A(H1N1)pdm09 or respiratory syncytial virus (RSV) in the nasal human airway epithelium (HAE) infected simultaneously or sequentially (24 h apart) with virus combinations. The replication kinetics of each virus were determined by RT-qPCR at different post-infection times. Our results showed that during simultaneous infection, SARS-CoV-2 interferes with RSV-A2 but not with A(H1N1)pdm09 replication. The prior infection of nasal HAE with SARS-CoV-2 reduces the replication kinetics of both respiratory viruses. SARS-CoV-2 replication is decreased by a prior infection with A(H1N1)pdm09 but not with RSV-A2. The pretreatment of nasal HAE with BX795, a TANK-binding kinase 1 inhibitor, partially alleviates the reduced replication of SARS-CoV-2 or influenza A(H1N1)pdm09 during sequential infection with both virus combinations. Thus, a prior infection of nasal HAE with SARS-CoV-2 interferes with the replication kinetics of A(H1N1)pdm09 and RSV-A2, whereas only A(H1N1)pdm09 reduces the subsequent infection with SARS-CoV-2. The mechanism involved in the viral interference between SARS-CoV-2 and A(H1N1)pdm09 is mediated by the production of interferon.


Subject(s)
Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/physiology , Nasopharynx/cytology , Respiratory Syncytial Virus, Human/physiology , SARS-CoV-2/physiology , Viral Interference , Virus Replication , Coinfection , Humans , Microbial Interactions , Nasopharynx/virology
8.
Emerg Infect Dis ; 28(2): 273-281, 2022 02.
Article in English | MEDLINE | ID: mdl-35075991

ABSTRACT

Multiple respiratory viruses can concurrently or sequentially infect the respiratory tract and lead to virus‒virus interactions. Infection by a first virus could enhance or reduce infection and replication of a second virus, resulting in positive (additive or synergistic) or negative (antagonistic) interaction. The concept of viral interference has been demonstrated at the cellular, host, and population levels. The mechanisms involved in viral interference have been evaluated in differentiated airway epithelial cells and in animal models susceptible to the respiratory viruses of interest. A likely mechanism is the interferon response that could confer a temporary nonspecific immunity to the host. During the coronavirus disease pandemic, nonpharmacologic interventions have prevented the circulation of most respiratory viruses. Once the sanitary restrictions are lifted, circulation of seasonal respiratory viruses is expected to resume and will offer the opportunity to study their interactions, notably with severe acute respiratory syndrome coronavirus 2.


Subject(s)
COVID-19 , Respiratory Tract Infections , Viruses , Animals , Humans , Pandemics , Respiratory Tract Infections/epidemiology , SARS-CoV-2 , Viral Interference
9.
Enzymes ; 50: 79-132, 2021.
Article in English | MEDLINE | ID: mdl-34861944

ABSTRACT

Human herpesviruses are large double-stranded DNA viruses belonging to the Herpesviridae family. The main characteristics of these viruses are their ability to establish a lifelong latency into the host with a potential to reactivate periodically. Primary infections and reactivations with herpesviruses are responsible for a large spectrum of diseases and may result in severe complications in immunocompromised patients. The viral DNA polymerase is a key enzyme in the replicative cycle of herpesviruses, and the target of most antiviral agents (i.e., nucleoside, nucleotide and pyrophosphate analogs). However, long-term prophylaxis and treatment with these antivirals may lead to the emergence of drug-resistant isolates harboring mutations in genes encoding viral enzymes that phosphorylate drugs (nucleoside analogs) and/or DNA polymerases, with potential cross-resistance between the different analogs. Drug resistance mutations mainly arise in conserved regions of the polymerase and exonuclease functional domains of these enzymes. In the polymerase domain, mutations associated with resistance to nucleoside/nucleotide analogs may directly or indirectly affect drug binding or incorporation into the primer strand, or increase the rate of extension of DNA to overcome chain termination. In the exonuclease domain, mutations conferring resistance to nucleoside/nucleotide analogs may reduce the rate of excision of incorporated drug, or continue DNA elongation after drug incorporation without excision. Mutations associated with resistance to pyrophosphate analogs may alter drug binding or the conformational changes of the polymerase domain required for an efficient activity of the enzyme. Novel herpesvirus inhibitors with a potent antiviral activity against drug-resistant isolates are thus needed urgently.


Subject(s)
Herpesviridae , Antiviral Agents/pharmacology , DNA-Directed DNA Polymerase/genetics , Herpesviridae/genetics , Humans , Nucleosides , Simplexvirus
10.
Sci Rep ; 11(1): 18688, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548521

ABSTRACT

Herpes simplex virus 1 (HSV-1) is responsible for herpes simplex virus encephalitis (HSE), associated with a 70% mortality rate in the absence of treatment. Despite intravenous treatment with acyclovir, mortality remains significant, highlighting the need for new anti-herpetic agents. Herein, we describe a novel neurovirulent recombinant HSV-1 (rHSV-1), expressing the fluorescent tdTomato and Gaussia luciferase (Gluc) enzyme, generated by the Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) (CRISPR-Cas9) system. The Gluc activity measured in the cell culture supernatant was correlated (P = 0.0001) with infectious particles, allowing in vitro monitoring of viral replication kinetics. A significant correlation was also found between brain viral titers and Gluc activity in plasma (R2 = 0.8510, P < 0.0001) collected from BALB/c mice infected intranasally with rHSV-1. Furthermore, evaluation of valacyclovir (VACV) treatment of HSE could also be performed by analyzing Gluc activity in mouse plasma samples. Finally, it was also possible to study rHSV-1 dissemination and additionally to estimate brain viral titers by in vivo imaging system (IVIS). The new rHSV-1 with reporter proteins is not only as a powerful tool for in vitro and in vivo antiviral screening, but can also be used for studying different aspects of HSE pathogenesis.


Subject(s)
Encephalitis, Herpes Simplex/physiopathology , Herpesvirus 1, Human/isolation & purification , Animals , Antiviral Agents/therapeutic use , Base Sequence , Blood-Brain Barrier , Brain/virology , Chlorocebus aethiops , Encephalitis, Herpes Simplex/drug therapy , Encephalitis, Herpes Simplex/virology , Genes, Reporter , Genes, Viral , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Luminescence , Mice , Mice, Inbred BALB C , Multiplex Polymerase Chain Reaction/methods , Valacyclovir/therapeutic use , Vero Cells , Viral Load , Virus Replication/genetics
11.
J Neuroinflammation ; 18(1): 178, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399779

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has been associated with several neurological complications in adult patients. METHODS: We used a mouse model deficient in TRIF and IPS-1 adaptor proteins, which are involved in type I interferon production, to study the role of microglia during brain infection by ZIKV. Young adult mice were infected intravenously with the contemporary ZIKV strain PRVABC59 (1 × 105 PFUs/100 µL). RESULTS: Infected mice did not present overt clinical signs of the disease nor body weight loss compared with noninfected animals. However, mice exhibited a viremia and a brain viral load that were maximal (1.3 × 105 genome copies/mL and 9.8 × 107 genome copies/g of brain) on days 3 and 7 post-infection (p.i.), respectively. Immunohistochemistry analysis showed that ZIKV antigens were distributed in several regions of the brain, especially the dorsal hippocampus. The number of Iba1+/TMEM119+ microglia remained similar in infected versus noninfected mice, but their cell body and arborization areas significantly increased in the stratum radiatum and stratum lacunosum-moleculare layers of the dorsal hippocampus cornu ammoni (CA)1, indicating a reactive state. Ultrastructural analyses also revealed that microglia displayed increased phagocytic activities and extracellular digestion of degraded elements during infection. Mice pharmacologically depleted in microglia with PLX5622 presented a higher brain viral load compared to untreated group (2.8 × 1010 versus 8.5 × 108 genome copies/g of brain on day 10 p.i.) as well as an increased number of ZIKV antigens labeled with immunogold in the cytoplasm and endoplasmic reticulum of neurons and astrocytes indicating an enhanced viral replication. Furthermore, endosomes of astrocytes contained nanogold particles together with digested materials, suggesting a compensatory phagocytic activity upon microglial depletion. CONCLUSIONS: These results indicate that microglia are involved in the control of ZIKV replication and/or its elimination in the brain. After depletion of microglia, the removal of ZIKV-infected cells by phagocytosis could be partly compensated by astrocytes.


Subject(s)
Brain/virology , Microglia/metabolism , Neurons/metabolism , Phagocytosis/physiology , Zika Virus Infection/metabolism , Animals , Brain/metabolism , Mice , Microglia/virology , Neurons/virology
12.
Adv Exp Med Biol ; 1322: 1-30, 2021.
Article in English | MEDLINE | ID: mdl-34258735

ABSTRACT

The discovery of the nucleoside analogue, acyclovir, represented a milestone in the management of infections caused by herpes simplex virus and varicella-zoster virus. Ganciclovir, another nucleoside analogue, was then used for the management of systemic and organ-specific human cytomegalovirus diseases. The pyrophosphate analogue, foscarnet, and the nucleotide analogue, cidofovir, have been approved subsequently and constitute the second-line antiviral drugs. However, the viral DNA polymerase is the ultimate target of all these antiviral agents with a possible emergence of cross-resistance between these drugs. Recently, letermovir that targets the viral terminase complex was approved for the prophylaxis of human cytomegalovirus infections in hematopoietic stem cell transplant recipients. Other viral targets such as the protein kinase and the helicase-primase complex are also evaluated for the development of novel potent inhibitors against herpesviruses.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cidofovir/pharmacology , Cytomegalovirus , Drug Resistance, Viral/genetics , Humans , Simplexvirus
13.
Antimicrob Agents Chemother ; 65(7): e0039021, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33875432

ABSTRACT

Amino acid substitutions conferring resistance of herpes simplex virus 1 (HSV-1) and human cytomegalovirus (HCMV) to foscarnet (PFA) are located in the genes UL30 and UL54, respectively, encoding the DNA polymerase (pol). In this study, we analyzed the impact of substitutions located in helix K and region II that are involved in the conformational changes of the DNA pol. Theoretical substitutions were identified by sequences alignment of the helix K and region II of human herpesviruses (susceptible to PFA) and bacteriophages (resistant to PFA) and introduced in viral genomes by recombinant phenotyping. We characterized the susceptibility of HSV-1 and HCMV mutants to PFA. In UL30, the substitutions I619K (helix K), V715S, and A719T (both in region II) increased mean PFA 50% effective concentrations (EC50s) by 2.5-, 5.6-, and 2.0-fold, respectively, compared to the wild type (WT). In UL54, the substitution Q579I (helix K) conferred hypersusceptibility to PFA (0.17-fold change), whereas the substitutions Q697P, V715S, and A719T (all in region II) increased mean PFA EC50s by 3.8-, 2.8- and 2.5-fold, respectively, compared to the WT. These results were confirmed by enzymatic assays using recombinant DNA pol harboring these substitutions. Three-dimensional modeling suggests that substitutions conferring resistance/hypersusceptibility to PFA located in helix K and region II of UL30 and UL54 DNA pol favor an open/closed conformation of these enzymes, resulting in a lower/higher drug affinity for the proteins. Thus, this study shows that both regions of UL30 and UL54 DNA pol are involved in the conformational changes of these proteins and can influence the susceptibility of both viruses to PFA.


Subject(s)
Herpesvirus 1, Human , Amino Acid Substitution , Antiviral Agents/pharmacology , Cytomegalovirus/genetics , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Viral/genetics , Foscarnet/pharmacology , Herpesvirus 1, Human/genetics , Humans , Mutation
14.
Antiviral Res ; 188: 105046, 2021 04.
Article in English | MEDLINE | ID: mdl-33588012

ABSTRACT

This study aimed at understanding the impact of different substitutions at codon 715 localized in the region II of the palm domain of herpes simplex virus 1 (HSV-1) and human cytomegalovirus (HCMV) DNA polymerases (pol). Here, we report a new theoretical mutation V715S that confers resistance of HSV-1 to foscarnet/acyclovir (5.6- and 9.2-fold increases EC50 values compared to wild type, respectively) and of HCMV to foscarnet/ganciclovir (2.8- and 2.9-fold increases in EC50 values compared to wild type, respectively). To further analyze the importance of this amino acid, we investigated the impact of the already known mutations V715M and V715G on the replicative capacities and drug susceptibilities of both viruses as well as on the activity and drug inhibition of the DNA pol. The V715G recombinant HSV-1 mutant was resistant to foscarnet and acyclovir (3.4- and 4.6-fold EC50 increase, respectively) whereas the V715M mutant was susceptible to foscarnet and resistant to acyclovir (3.4-fold EC50 increase). The V715G recombinant HCMV mutant did not grow and the V715M mutant was resistant to foscarnet (3.7-fold EC50 increase) and susceptible to ganciclovir. Finally, we showed by three-dimensional modeling that the differential impact of these mutations on the viral replicative capacity and drug resistance profile was related to different hydrophobic local environments for V715 in the DNA pol of the two viruses. Furthermore, we hypothesize that the DNA pol of HSV-1 is more tolerant to changes at this residue compared to that of HCMV because of a more hydrophobic environment stabilizing the region.


Subject(s)
Antiviral Agents/pharmacology , Codon , Cytomegalovirus/genetics , DNA-Directed DNA Polymerase/genetics , Exodeoxyribonucleases/genetics , Herpesvirus 1, Human/genetics , Mutation, Missense , Viral Proteins/genetics , Acyclovir/pharmacology , Animals , Chlorocebus aethiops , Cytomegalovirus/drug effects , Cytomegalovirus/enzymology , Cytomegalovirus/physiology , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Foscarnet/pharmacology , Ganciclovir/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/physiology , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Models, Molecular , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication
15.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32938766

ABSTRACT

The role of a signaling pathway through macrophage colony-stimulating factor (MCSF) and its receptor, macrophage colony-stimulating factor 1 receptor (CSF1R), during experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE) was studied by two different approaches. First, we evaluated the effect of stimulation of the MCSF/CSF1R axis before infection. Exogenous MCSF (40 µg/kg of body weight intraperitoneally [i.p.]) was administered once daily to BALB/c mice on days 4 and 2 before intranasal infection with 2,500 PFU of HSV-1. MCSF treatment significantly increased mouse survival compared to saline (50% versus 10%; P = 0.0169). On day 6 postinfection (p.i.), brain viral titers were significantly decreased, whereas beta interferon (IFN-ß) was significantly increased in mice treated with MCSF compared to mice treated with saline. The number of CD68+ (a phagocytosis marker) microglial cells was significantly increased in MCSF-treated mice compared to the saline-treated group. Secondly, we conditionally depleted CSF1R on microglial cells of CSF1R-loxP-CX3CR1-cre/ERT2 mice (in a C57BL/6 background) through induction with tamoxifen. The mice were then infected intranasally with 600,000 PFU of HSV-1. The survival rate of mice depleted of CSF1R (knockout [KO] mice) was significantly lower than that of wild-type (WT) mice (0% versus 67%). Brain viral titers and cytokine/chemokine levels were significantly higher in KO than in WT animals on day 6 p.i. Furthermore, increased infiltration of monocytes into the brains of WT mice was seen on day 6 p.i., but not in KO mice. Our results suggest that microglial cells are essential to control HSE at early stages of the disease and that the MCSF/CSF1R axis could be a therapeutic target to regulate their response to infection.IMPORTANCE Microglia appear to be one of the principal regulators of neuroinflammation in the central nervous system (CNS). An increasing number of studies have demonstrated that the activation of microglia could result in either beneficial or detrimental effects in different CNS disorders. Hence, the role of microglia during herpes simplex virus encephalitis (HSE) has not been fully characterized. Using experimental mouse models, we showed that an early activation of the MCSF/CSF1R axis improved the outcome of the disease, possibly by inducing a proliferation of microglia. In contrast, depletion of microglia before HSV-1 infection worsened the prognosis of HSE. Thus, an early microglial response followed by sustained infiltration of monocytes and T cells into the brain seem to be key components for a better clinical outcome. These data suggest that microglia could be a potential target for immunomodulatory strategies combined with antiviral therapy to better control the outcome of this devastating disease.


Subject(s)
Encephalitis, Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Microglia/metabolism , Microglia/virology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Brain/virology , Central Nervous System/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Phagocytosis , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Viral Load
16.
Methods Mol Biol ; 2142: 41-57, 2020.
Article in English | MEDLINE | ID: mdl-32367357

ABSTRACT

Detection of Zika virus (ZIKV) in the central nervous system (CNS) is a critical step when studying the pathogenesis of the infection in animal models. Both viral load determination and immunohistochemistry (IHC) staining are useful methods to quantitatively and qualitatively characterize viral infections in target tissues. Here, we describe viral RNA load determination by droplet digital PCR as well as protein detection by polymer-based IHC as effective techniques to quantify and localize ZIKV in the CNS of mice.


Subject(s)
Central Nervous System/virology , Immunohistochemistry/methods , Polymerase Chain Reaction/methods , Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System Viral Diseases/diagnosis , Central Nervous System Viral Diseases/metabolism , Central Nervous System Viral Diseases/pathology , Central Nervous System Viral Diseases/virology , Disease Models, Animal , Mice , Mice, Knockout , RNA, Viral/analysis , RNA, Viral/metabolism , Viral Load/methods , Zika Virus/genetics , Zika Virus/metabolism , Zika Virus Infection/metabolism , Zika Virus Infection/pathology , Zika Virus Infection/virology
17.
Sci Adv ; 6(5): eaax9318, 2020 01.
Article in English | MEDLINE | ID: mdl-32064341

ABSTRACT

Viral infections kill millions of people and new antivirals are needed. Nontoxic drugs that irreversibly inhibit viruses (virucidal) are postulated to be ideal. Unfortunately, all virucidal molecules described to date are cytotoxic. We recently developed nontoxic, broad-spectrum virucidal gold nanoparticles. Here, we develop further the concept and describe cyclodextrins, modified with mercaptoundecane sulfonic acids, to mimic heparan sulfates and to provide the key nontoxic virucidal action. We show that the resulting macromolecules are broad-spectrum, biocompatible, and virucidal at micromolar concentrations in vitro against many viruses [including herpes simplex virus (HSV), respiratory syncytial virus (RSV), dengue virus, and Zika virus]. They are effective ex vivo against both laboratory and clinical strains of RSV and HSV-2 in respiratory and vaginal tissue culture models, respectively. Additionally, they are effective when administrated in mice before intravaginal HSV-2 inoculation. Lastly, they pass a mutation resistance test that the currently available anti-HSV drug (acyclovir) fails.


Subject(s)
Cyclodextrins/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Virus Diseases/drug therapy , Acyclovir/chemistry , Acyclovir/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cyclodextrins/chemical synthesis , Cyclodextrins/chemistry , Female , Gold/chemistry , Heparitin Sulfate/chemistry , Heparitin Sulfate/pharmacology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/pathogenicity , Humans , Metal Nanoparticles/chemistry , Mice , Simplexvirus/drug effects , Simplexvirus/pathogenicity , Virus Diseases/virology , Zika Virus/drug effects , Zika Virus/pathogenicity
18.
Clin Microbiol Rev ; 33(2)2020 03 18.
Article in English | MEDLINE | ID: mdl-32051176

ABSTRACT

Herpes simplex virus 1 (HSV-1) can be responsible for life-threatening HSV encephalitis (HSE). The mortality rate of patients with HSE who do not receive antiviral treatment is 70%, with most survivors suffering from permanent neurological sequelae. The use of intravenous acyclovir together with improved diagnostic technologies such as PCR and magnetic resonance imaging has resulted in a reduction in the mortality rate to close to 20%. However, 70% of surviving patients still do not recover complete neurological functions. Thus, there is an urgent need to develop more effective treatments for a better clinical outcome. It is well recognized that cerebral damage resulting from HSE is caused by viral replication together with an overzealous inflammatory response. Both of these processes constitute potential targets for the development of innovative therapies against HSE. In this review, we discuss recent progress in therapy that may be used to ameliorate the outcome of patients with HSE, with a particular emphasis on immunomodulatory agents. Ideally, the administration of adjunctive immunomodulatory drugs should be initiated during the rise of the inflammatory response, and its duration should be limited in time to reduce undesired effects. This critical time frame should be optimized by the identification of reliable biomarkers of inflammation.


Subject(s)
Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Herpes Simplex/immunology , Encephalitis, Herpes Simplex/therapy , Immunomodulation , Acyclovir/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Animals , Antiviral Agents/therapeutic use , Drug Therapy , Genetic Predisposition to Disease , Humans , Immunity , Risk Factors , Simplexvirus/drug effects , Treatment Outcome
19.
Article in English | MEDLINE | ID: mdl-32015044

ABSTRACT

Herein, we phenotypically and enzymatically characterize the theoretical mutation Q579I in helix K and the already described clinical mutation K805Q in helix P of cytomegalovirus DNA polymerase for susceptibility to foscarnet. Q579I and K805Q recombinant viruses were hypersusceptible to foscarnet (respective mean 50% effective concentrations [EC50] of 0.12- and 0.19-fold that of the wild type). Three-dimensional modeling analysis suggested that both mutations favor the closed conformation of the enzyme to which foscarnet binds with a higher affinity.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , DNA-Directed DNA Polymerase/genetics , Foscarnet/pharmacology , Cytomegalovirus Infections/drug therapy , DNA, Viral/genetics , DNA-Directed DNA Polymerase/drug effects , Drug Resistance, Viral/genetics , Humans , Models, Molecular , Mutation
20.
Front Microbiol ; 11: 631736, 2020.
Article in English | MEDLINE | ID: mdl-33584597

ABSTRACT

The emergence and spread of infectious diseases with pandemic potential occurred regularly throughout history. Major pandemics and epidemics such as plague, cholera, flu, severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) have already afflicted humanity. The world is now facing the new coronavirus disease 2019 (COVID-19) pandemic. Many infectious diseases leading to pandemics are caused by zoonotic pathogens that were transmitted to humans due to increased contacts with animals through breeding, hunting and global trade activities. The understanding of the mechanisms of transmission of pathogens to humans allowed the establishment of methods to prevent and control infections. During centuries, implementation of public health measures such as isolation, quarantine and border control helped to contain the spread of infectious diseases and maintain the structure of the society. In the absence of pharmaceutical interventions, these containment methods have still been used nowadays to control COVID-19 pandemic. Global surveillance programs of water-borne pathogens, vector-borne diseases and zoonotic spillovers at the animal-human interface are of prime importance to rapidly detect the emergence of infectious threats. Novel technologies for rapid diagnostic testing, contact tracing, drug repurposing, biomarkers of disease severity as well as new platforms for the development and production of vaccines are needed for an effective response in case of pandemics.

SELECTION OF CITATIONS
SEARCH DETAIL
...