Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 136
Filtrer
1.
Cell Host Microbe ; 32(6): 786-793, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38870896

RÉSUMÉ

Survival strategies of human-associated microbes to drug exposure have been mainly studied in the context of bona fide pathogens exposed to antibiotics. Less well understood are the survival strategies of non-pathogenic microbes and host-associated commensal communities to the variety of drugs and xenobiotics to which humans are exposed. The lifestyle of microbial commensals within complex communities offers a variety of ways to adapt to different drug-induced stresses. Here, we review the responses and survival strategies employed by gut commensals when exposed to drugs-antibiotics and non-antibiotics-at the individual and community level. We also discuss the factors influencing the recovery and establishment of a new community structure following drug exposure. These survival strategies are key to the stability and resilience of the gut microbiome, ultimately influencing the overall health and well-being of the host.


Sujet(s)
Antibactériens , Bactéries , Microbiome gastro-intestinal , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie , Bactéries/effets des médicaments et des substances chimiques , Xénobiotique/pharmacologie , Symbiose , Tube digestif/microbiologie , Tube digestif/effets des médicaments et des substances chimiques , Résistance bactérienne aux médicaments , Interactions hôte-microbes/effets des médicaments et des substances chimiques
2.
Microb Ecol ; 86(1): 86-96, 2023 Jul.
Article de Anglais | MEDLINE | ID: mdl-35809121

RÉSUMÉ

Skin harbors an important microbial ecosystem - the skin microbiota that is in homeostasis with its host and is beneficial for human health. Cosmetic products have the potential to interfere with this microbial community; therefore their impact should be assessed. The aim of this review is to highlight the importance of skin microbiota in the cosmetic industry. Several studies determined that cosmetic ingredients have the potential to disrupt the skin microbiota equilibrium leading to the development of skin diseases and dysregulation of immune response. These studies led their investigation by using different methodologies and models, concluding that methods must be chosen according to the aim of the study, the skin site to be evaluated, and the target population of the cosmetics. Overall, it is crucial to test the impact of cosmetics in the skin microbiota and to stablish standard procedures, as well as specific criteria that allow to classify a cosmetic product as skin microbiota friendly.


Sujet(s)
Cosmétiques , Interactions hôte-microbes , Microbiote , Peau , Humains , Cosmétiques/pharmacologie , Homéostasie , Microbiote/effets des médicaments et des substances chimiques , Peau/microbiologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Industrie/normes , Industrie/tendances
3.
Med Sci Monit ; 28: e936292, 2022 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-35256581

RÉSUMÉ

In the past 2 years, the coronavirus disease 2019 (COVID-19) pandemic has driven investigational studies and controlled clinical trials on antiviral treatments and vaccines that have undergone regulatory approval. Now that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants may become endemic over time, there remains a need to identify drugs that treat the symptoms of COVID-19 and prevent progression toward severe cases, hospitalization, and death. Understanding the molecular mechanisms of SARS-CoV-2 infection is extremely important for the development of effective therapies against COVID-19. This review outlines the key pathways involved in the host response to SARS-CoV-2 infection and discusses the potential role of antioxidant and anti-inflammatory pharmacological approaches for the management of early mild-to-moderate COVID-19, using the examples of combined indomethacin, low-dose aspirin, omeprazole, hesperidin, quercetin, and vitamin C. The pharmacological targets of these substances are described here for their possible synergism in counteracting SARS-CoV-2 replication and progression of the infection from the upper respiratory airways to the blood, avoiding vascular complications and cytokine and bradykinin storms.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Interactions hôte-microbes/effets des médicaments et des substances chimiques , SARS-CoV-2/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Antioxydants/pharmacologie , Antiviraux/usage thérapeutique , Maladies endémiques , Interactions hôte-microbes/physiologie , Humains , Phénomènes pharmacologiques/physiologie , SARS-CoV-2/pathogénicité
4.
Comput Math Methods Med ; 2022: 3179200, 2022.
Article de Anglais | MEDLINE | ID: mdl-35309841

RÉSUMÉ

Human immunodeficiency virus (HIV) infection is characterized not only by severe immunodeficiency but also by persistent inflammation and immune activation. These characteristics persist in people living with HIV (PLHIV) receiving effective antiretroviral therapy (ART) and are associated with morbidity and mortality in nonacquired immunodeficiency syndrome (AIDS) events. ART can inhibit HIV replication and promote immune reconstitution, which is currently the most effective way to control AIDS. However, despite effective long-term ART and overall suppression of plasma HIV RNA level, PLHIV still shows chronic low-level inflammation. The exact mechanisms that trigger chronic inflammation are unknown. Activation of the inflammasome is essential for the host response to pathogens, and some recent studies have confirmed the role of the inflammasome in the pathogenesis of inflammatory diseases. The NLRP3 inflammasome has been widely studied, which is a pyrin domain-containing protein 3 belonging to the family of nucleotide-binding and oligomerization domain-like receptors (NLRs). Recent studies suggest that inflammasome-mediated pyroptosis is associated with CD4+ T cell loss in the absence of persistent infectious HIV replication. This article reviews the mechanism of the NLRP3 inflammasome and its correlation with immune reconstitution in PLHIV treated with ART.


Sujet(s)
Agents antiVIH/usage thérapeutique , Infections à VIH/traitement médicamenteux , Infections à VIH/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/anatomopathologie , Biologie informatique , Infections à VIH/anatomopathologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/immunologie , Humains , Reconstitution immunitaire , Inflammasomes/effets des médicaments et des substances chimiques , Inflammasomes/immunologie , Inflammation/traitement médicamenteux , Inflammation/immunologie , Inflammation/anatomopathologie , Pyroptose/effets des médicaments et des substances chimiques , Pyroptose/immunologie
5.
Cells ; 11(1)2022 01 03.
Article de Anglais | MEDLINE | ID: mdl-35011708

RÉSUMÉ

Extracellular vesicles (EVs) and viruses share common features: size, structure, biogenesis and uptake. In order to generate EVs expressing the SARS-CoV-2 spike protein on their surface (S-EVs), we collected EVs from SARS-CoV-2 spike expressing human embryonic kidney (HEK-293T) cells by stable transfection with a vector coding for the S1 and S2 subunits. S-EVs were characterized using nanoparticle tracking analysis, ExoView and super-resolution microscopy. We obtained a population of EVs of 50 to 200 nm in size. Spike expressing EVs represented around 40% of the total EV population and co-expressed spike protein with tetraspanins on the surfaces of EVs. We subsequently used ACE2-positive endothelial and bronchial epithelial cells for assessing the internalization of labeled S-EVs using a cytofluorimetric analysis. Internalization of S-EVs was higher than that of control EVs from non-transfected cells. Moreover, S-EV uptake was significantly decreased by anti-ACE2 antibody pre-treatment. Furthermore, colchicine, a drug currently used in clinical trials, significantly reduced S-EV entry into the cells. S-EVs represent a simple, safe, and scalable model to study host-virus interactions and the mechanisms of novel therapeutic drugs.


Sujet(s)
COVID-19/métabolisme , Vésicules extracellulaires/métabolisme , SARS-CoV-2/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Angiotensin-converting enzyme 2/immunologie , Anticorps bloquants/pharmacologie , COVID-19/virologie , Lignée cellulaire , Cellules cultivées , Colchicine/pharmacologie , Cytométrie en flux/méthodes , Cellules HEK293 , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/virologie , Humains , Microscopie de fluorescence/méthodes , Liaison aux protéines/effets des médicaments et des substances chimiques , SARS-CoV-2/physiologie
6.
Sci Rep ; 12(1): 857, 2022 01 17.
Article de Anglais | MEDLINE | ID: mdl-35039591

RÉSUMÉ

Sorghum damping-off, caused by Fusarium solani (Mart.) Sacc., is a serious disease which causes economic loss in sorghum production. In this study, antagonistic activity of lavender essential oil (EO) at 0.5, 0.75, 1.0, 1.25, 1.5, and 1.6% against F. solani was studied in vitro. Their effects on regulation of three SbWRKY transcription factors, the response factor JERF3 and eight defense-related genes, which mediate different signaling pathways, in sorghum were investigated. Effects of application under greenhouse conditions were also evaluated. The results showed that lavender EO possesses potent antifungal activity against F. solani. A complete inhibition in the fungal growth was recorded for lavender EO at 1.6%. Gas chromatography-mass spectrometric analysis revealed that EO antifungal activity is most likely attributed to linalyl anthranilate, α-terpineol, eucalyptol, α-Pinene, and limonene. Observations using transmission electron microscopy revealed many abnormalities in the ultrastructures of the fungal mycelium as a response to treating with lavender EO, indicating that multi-mechanisms contributed to their antagonistic behavior. Results obtained from Real-time PCR investigations demonstrated that the genes studied were overexpressed, to varying extents in response to lavender EO. However, SbWRKY1 was the highest differentially expressed gene followed by JERF3, which suggest they play primary role(s) in synchronously organizing the transcription-regulatory-networks enhancing the plant resistance. Under greenhouse conditions, treating of sorghum grains with lavender EO at 1.5% prior to infection significantly reduced disease severity. Moreover, the growth parameters evaluated, the activities of antioxidant enzymes, and total phenolic and flavonoid contents were all enhanced. In contrast, lipid peroxidation was highly reduced. Results obtained from this study support the possibility of using lavender EO for control of sorghum damping-off. However, field evaluation is highly needed prior to any usage recommendation.


Sujet(s)
Antifongiques , Fusarium/effets des médicaments et des substances chimiques , Fusarium/pathogénicité , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/génétique , Expression des gènes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/génétique , Lavandula/composition chimique , Huile essentielle/pharmacologie , Maladies des plantes/génétique , Maladies des plantes/microbiologie , Huiles végétales/pharmacologie , Sorghum/génétique , Sorghum/microbiologie , Facteurs de transcription/génétique , Résistance des champignons aux médicaments , Expression des gènes/génétique , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Réseaux de régulation génique/génétique , Huile essentielle/isolement et purification , Huiles végétales/isolement et purification , Facteurs de transcription/métabolisme
7.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34902435

RÉSUMÉ

BACKGROUND: Treatments for coronavirus disease 2019, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are urgently needed but remain limited. SARS-CoV-2 infects cells through interactions of its spike (S) protein with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) on host cells. Multiple cells and organs are targeted, particularly airway epithelial cells. OM-85, a standardized lysate of human airway bacteria with strong immunomodulating properties and an impeccable safety profile, is widely used to prevent recurrent respiratory infections. We found that airway OM-85 administration inhibits Ace2 and Tmprss2 transcription in the mouse lung, suggesting that OM-85 might hinder SARS-CoV-2/host cell interactions. OBJECTIVES: We sought to investigate whether and how OM-85 treatment protects nonhuman primate and human epithelial cells against SARS-CoV-2. METHODS: ACE2 and TMPRSS2 mRNA and protein expression, cell binding of SARS-CoV-2 S1 protein, cell entry of SARS-CoV-2 S protein-pseudotyped lentiviral particles, and SARS-CoV-2 cell infection were measured in kidney, lung, and intestinal epithelial cell lines, primary human bronchial epithelial cells, and ACE2-transfected HEK293T cells treated with OM-85 in vitro. RESULTS: OM-85 significantly downregulated ACE2 and TMPRSS2 transcription and surface ACE2 protein expression in epithelial cell lines and primary bronchial epithelial cells. OM-85 also strongly inhibited SARS-CoV-2 S1 protein binding to, SARS-CoV-2 S protein-pseudotyped lentivirus entry into, and SARS-CoV-2 infection of epithelial cells. These effects of OM-85 appeared to depend on SARS-CoV-2 receptor downregulation. CONCLUSIONS: OM-85 inhibits SARS-CoV-2 epithelial cell infection in vitro by downregulating SARS-CoV-2 receptor expression. Further studies are warranted to assess whether OM-85 may prevent and/or reduce the severity of coronavirus disease 2019.


Sujet(s)
Adjuvants immunologiques/administration et posologie , COVID-19/prévention et contrôle , Extrait cellulaire/administration et posologie , Récepteurs viraux/antagonistes et inhibiteurs , Récepteurs viraux/immunologie , SARS-CoV-2/immunologie , Angiotensin-converting enzyme 2/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/immunologie , Animaux , COVID-19/immunologie , COVID-19/virologie , Cellules Caco-2 , Extrait cellulaire/immunologie , Cellules cultivées , Chlorocebus aethiops , Régulation négative/effets des médicaments et des substances chimiques , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Cellules HEK293 , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/immunologie , Humains , Techniques in vitro , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Poumon/virologie , Souris , Souris de lignée BALB C , Serine endopeptidases/effets des médicaments et des substances chimiques , Serine endopeptidases/génétique , Serine endopeptidases/immunologie , Transcription génétique/effets des médicaments et des substances chimiques , Transcription génétique/immunologie , Cellules Vero
8.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-34948270

RÉSUMÉ

In recent years, commensal bacteria colonizing the human body have been recognized as important determinants of health and multiple pathologic conditions. Among the most extensively studied commensal bacteria are the gut microbiota, which perform a plethora of functions, including the synthesis of bioactive products, metabolism of dietary compounds, and immunomodulation, both through attenuation and immunostimulation. An imbalance in the microbiota population, i.e., dysbiosis, has been linked to many human pathologies, including various cancer types and neurodegenerative diseases. Targeting gut microbiota and microbiome-host interactions resulting from probiotics, prebiotics, and postbiotics is a growing opportunity for the effective treatment of various diseases. As more research is being conducted, the microbiome field is shifting from simple descriptive analysis of commensal compositions to more molecular, cellular, and functional studies. Insight into these mechanisms is of paramount importance for understanding and modulating the effects that microbiota, probiotics, and their derivatives exert on host health.


Sujet(s)
Microbiome gastro-intestinal/physiologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Bactéries , Régime alimentaire , Dysbiose/microbiologie , Humains , Microbiote/physiologie , Prébiotiques/microbiologie , Probiotiques/métabolisme , Probiotiques/pharmacologie , Symbiose/physiologie
9.
Commun Biol ; 4(1): 1324, 2021 11 24.
Article de Anglais | MEDLINE | ID: mdl-34819611

RÉSUMÉ

The gut microbiome produces vitamins, nutrients, and neurotransmitters, and helps to modulate the host immune system-and also plays a major role in the metabolism of many exogenous compounds, including drugs and chemical toxicants. However, the extent to which specific microbial species or communities modulate hazard upon exposure to chemicals remains largely opaque. Focusing on the effects of collateral dietary exposure to the widely used herbicide atrazine, we applied integrated omics and phenotypic screening to assess the role of the gut microbiome in modulating host resilience in Drosophila melanogaster. Transcriptional and metabolic responses to these compounds are sex-specific and depend strongly on the presence of the commensal microbiome. Sequencing the genomes of all abundant microbes in the fly gut revealed an enzymatic pathway responsible for atrazine detoxification unique to Acetobacter tropicalis. We find that Acetobacter tropicalis alone, in gnotobiotic animals, is sufficient to rescue increased atrazine toxicity to wild-type, conventionally reared levels. This work points toward the derivation of biotic strategies to improve host resilience to environmental chemical exposures, and illustrates the power of integrative omics to identify pathways responsible for adverse health outcomes.


Sujet(s)
Atrazine/toxicité , Drosophila melanogaster/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Insecticides/toxicité , Acetobacter/génétique , Acetobacter/métabolisme , Animaux , Drosophila melanogaster/microbiologie , Femelle , Inactivation métabolique , Mâle
10.
Molecules ; 26(20)2021 Oct 14.
Article de Anglais | MEDLINE | ID: mdl-34684786

RÉSUMÉ

Two targeted sets of novel 1,5-diaryl-1H-imidazole-4-carboxylic acids 10 and carbohydrazides 11 were designed and synthesized from their corresponding ester intermediates 17, which were prepared via cycloaddition of ethyl isocyanoacetate 16 and diarylimidoyl chlorides 15. Evaluation of these new target scaffolds in the AlphaScreenTM HIV-1 IN-LEDGF/p75 inhibition assay identified seventeen compounds exceeding the pre-defined 50% inhibitory threshold at 100 µM concentration. Further evaluation of these compounds in the HIV-1 IN strand transfer assay at 100 µM showed that none of the compounds (with the exception of 10a, 10l, and 11k, with marginal inhibitory percentages) were actively bound to the active site, indicating that they are selectively binding to the LEDGF/p75-binding pocket. In a cell-based HIV-1 antiviral assay, compounds 11a, 11b, 11g, and 11h exhibited moderate antiviral percentage inhibition of 33-45% with cytotoxicity (CC50) values of >200 µM, 158.4 µM, >200 µM, and 50.4 µM, respectively. The antiviral inhibitory activity displayed by 11h was attributed to its toxicity. Upon further validation of their ability to induce multimerization in a Western blot gel assay, compounds 11a, 11b, and 11h appeared to increase higher-order forms of IN.


Sujet(s)
Protéines adaptatrices de la transduction du signal/antagonistes et inhibiteurs , Inhibiteurs de l'intégrase du VIH/composition chimique , Inhibiteurs de l'intégrase du VIH/synthèse chimique , Intégrase du VIH/effets des médicaments et des substances chimiques , Facteurs de transcription/antagonistes et inhibiteurs , Domaine catalytique , Lignée cellulaire , Simulation numérique , Conception de médicament , Évaluation préclinique de médicament , Intégrase du VIH/composition chimique , Intégrase du VIH/métabolisme , Inhibiteurs de l'intégrase du VIH/pharmacologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Humains , Imidazoles/synthèse chimique , Imidazoles/composition chimique , Imidazoles/pharmacologie , Simulation de docking moléculaire , Structure moléculaire , Multimérisation de protéines/effets des médicaments et des substances chimiques
11.
Comput Math Methods Med ; 2021: 4348910, 2021.
Article de Anglais | MEDLINE | ID: mdl-34707682

RÉSUMÉ

In this study, two types of epidemiological models called "within host" and "between hosts" have been studied. The within-host model represents the innate immune response, and the between-hosts model signifies the SEIR (susceptible, exposed, infected, and recovered) epidemic model. The major contribution of this paper is to break the chain of infectious disease transmission by reducing the number of susceptible and infected people via transferring them to the recovered people group with vaccination and antiviral treatment, respectively. Both transfers are considered with time delay. In the first step, optimal control theory is applied to calculate the optimal final time to control the disease within a host's body with a cost function. To this end, the vaccination that represents the effort that converts healthy cells into resistant-to-infection cells in the susceptible individual's body is used as the first control input to vaccinate the susceptible individual against the disease. Moreover, the next control input (antiviral treatment) is applied to eradicate the concentrations of the virus and convert healthy cells into resistant-to-infection cells simultaneously in the infected person's body to treat the infected individual. The calculated optimal time in the first step is considered as the delay of vaccination and antiviral treatment in the SEIR dynamic model. Using Pontryagin's maximum principle in the second step, an optimal control strategy is also applied to an SEIR mathematical model with a nonlinear transmission rate and time delay, which is computed as optimal time in the first step. Numerical results are consistent with the analytical ones and corroborate our theoretical results.


Sujet(s)
Modèles épidémiologiques , Grippe humaine/traitement médicamenteux , Grippe humaine/prévention et contrôle , Antiviraux/administration et posologie , Biologie informatique , Simulation numérique , Prédisposition aux maladies , Transmission de maladie infectieuse/prévention et contrôle , Transmission de maladie infectieuse/statistiques et données numériques , Épidémies/prévention et contrôle , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/immunologie , Humains , Immunité innée , Vaccins antigrippaux/administration et posologie , Grippe humaine/transmission , Dynamique non linéaire
12.
Viruses ; 13(9)2021 09 02.
Article de Anglais | MEDLINE | ID: mdl-34578337

RÉSUMÉ

The Michael addition reaction is a spontaneous and quick chemical reaction that is widely applied in various fields. This reaction is performed by conjugating an addition of nucleophiles with α, ß-unsaturated carbonyl compounds, resulting in the bond formation of C-N, C-S, C-O, and so on. In the development of molecular materials, the Michael addition is not only used to synthesize chemical compounds but is also involved in the mechanism of drug action. Several covalent drugs that bond via Michael addition are regarded as anticarcinogens and anti-inflammatory drugs. Although drug development is mainly focused on pharmaceutical drug discovery, target-based discovery can provide a different perspective for drug usage. However, considerable time and labor are required to define a molecular target through molecular biological experiments. In this review, we systematically examine the chemical structures of current FDA-approved antiviral drugs for potential Michael addition moieties with α, ß-unsaturated carbonyl groups, which may exert an unidentified broad-spectrum inhibitory mechanism to target viral or host factors. We thus propose that profiling the targets of antiviral agents, such as Michael addition products, can be achieved by employing a high-throughput LC-MS approach to comprehensively analyze the interaction between drugs and targets, and the subsequent drug responses in the cellular environment to facilitate drug repurposing and/or identify potential adverse effects, with a particular emphasis on the pros and cons of this shotgun proteomic approach.


Sujet(s)
Antiviraux/composition chimique , Découverte de médicament/méthodes , Composés chimiques organiques/composition chimique , Préparations pharmaceutiques/composition chimique , Protéomique/méthodes , Antiviraux/isolement et purification , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Humains
13.
BMC Med Genomics ; 14(1): 226, 2021 09 17.
Article de Anglais | MEDLINE | ID: mdl-34535131

RÉSUMÉ

BACKGROUND: Higher mortality of COVID-19 patients with lung disease is a formidable challenge for the health care system. Genetic association between COVID-19 and various lung disorders must be understood to comprehend the molecular basis of comorbidity and accelerate drug development. METHODS: Lungs tissue-specific neighborhood network of human targets of SARS-CoV-2 was constructed. This network was integrated with lung diseases to build a disease-gene and disease-disease association network. Network-based toolset was used to identify the overlapping disease modules and drug targets. The functional protein modules were identified using community detection algorithms and biological processes, and pathway enrichment analysis. RESULTS: In total, 141 lung diseases were linked to a neighborhood network of SARS-CoV-2 targets, and 59 lung diseases were found to be topologically overlapped with the COVID-19 module. Topological overlap with various lung disorders allows repurposing of drugs used for these disorders to hit the closely associated COVID-19 module. Further analysis showed that functional protein-protein interaction modules in the lungs, substantially hijacked by SARS-CoV-2, are connected to several lung disorders. FDA-approved targets in the hijacked protein modules were identified and that can be hit by exiting drugs to rescue these modules from virus possession. CONCLUSION: Lung diseases are clustered with COVID-19 in the same network vicinity, indicating the potential threat for patients with respiratory diseases after SARS-CoV-2 infection. Pathobiological similarities between lung diseases and COVID-19 and clinical evidence suggest that shared molecular features are the probable reason for comorbidity. Network-based drug repurposing approaches can be applied to improve the clinical conditions of COVID-19 patients.


Sujet(s)
Traitements médicamenteux de la COVID-19 , COVID-19/épidémiologie , Repositionnement des médicaments , Maladies pulmonaires/épidémiologie , Pandémies , SARS-CoV-2 , Algorithmes , Antiviraux/usage thérapeutique , COVID-19/génétique , Comorbidité , Découverte de médicament , Repositionnement des médicaments/méthodes , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/génétique , Humains , Maladies pulmonaires/traitement médicamenteux , Maladies pulmonaires/génétique , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/génétique , Biologie des systèmes
14.
PLoS Comput Biol ; 17(9): e1009418, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34555024

RÉSUMÉ

Increasing body of experimental evidence suggests that anticancer and antimicrobial therapies may themselves promote the acquisition of drug resistance by increasing mutability. The successful control of evolving populations requires that such biological costs of control are identified, quantified and included to the evolutionarily informed treatment protocol. Here we identify, characterise and exploit a trade-off between decreasing the target population size and generating a surplus of treatment-induced rescue mutations. We show that the probability of cure is maximized at an intermediate dosage, below the drug concentration yielding maximal population decay, suggesting that treatment outcomes may in some cases be substantially improved by less aggressive treatment strategies. We also provide a general analytical relationship that implicitly links growth rate, pharmacodynamics and dose-dependent mutation rate to an optimal control law. Our results highlight the important, but often neglected, role of fundamental eco-evolutionary costs of control. These costs can often lead to situations, where decreasing the cumulative drug dosage may be preferable even when the objective of the treatment is elimination, and not containment. Taken together, our results thus add to the ongoing criticism of the standard practice of administering aggressive, high-dose therapies and motivate further experimental and clinical investigation of the mutagenicity and other hidden collateral costs of therapies.


Sujet(s)
Résistance microbienne aux médicaments/génétique , Résistance aux médicaments antinéoplasiques/génétique , Anti-infectieux/administration et posologie , Antinéoplasiques/administration et posologie , Biologie informatique , Simulation numérique , Relation dose-effet des médicaments , Évolution moléculaire , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/génétique , Humains , Modèles biologiques , Mutation/effets des médicaments et des substances chimiques , Taux de mutation , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Phénotype , Processus stochastiques
15.
J Hepatol ; 75(6): 1452-1464, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34453966

RÉSUMÉ

Hepatic encephalopathy (HE) is a complication of cirrhosis characterised by neuropsychiatric and motor dysfunction. Microbiota-host interactions play an important role in HE pathogenesis. Therapies targeting microbial community composition and function have been explored for the treatment of HE. Prebiotics, probiotics and faecal microbiota transplant (FMT) have been used with the aim of increasing the abundance of potentially beneficial taxa, while antibiotics have been used to decrease the abundance of potentially harmful taxa. Other microbiome therapeutics, including postbiotics and absorbents, have been used to target microbial products. Microbiome-targeted therapies for HE have had some success, notably lactulose and rifaximin, with probiotics and FMT also showing promise. However, there remain several challenges to the effective application of microbiome therapeutics in HE, including the resilience of the microbiome to sustainable change and unpredictable clinical outcomes from microbiota alterations. Future work in this space should focus on rigorous trial design, microbiome therapy selection, and a personalised approach to HE.


Sujet(s)
Encéphalopathie hépatique/traitement médicamenteux , Microbiote/effets des médicaments et des substances chimiques , Transplantation de microbiote fécal/méthodes , Transplantation de microbiote fécal/statistiques et données numériques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Humains , Prébiotiques/administration et posologie , Probiotiques/usage thérapeutique
16.
Front Immunol ; 12: 700184, 2021.
Article de Anglais | MEDLINE | ID: mdl-34408749

RÉSUMÉ

Coronavirus disease 2019 (COVID-19), which has high incidence rates with rapid rate of transmission, is a pandemic that spread across the world, resulting in more than 3,000,000 deaths globally. Currently, several drugs have been used for the clinical treatment of COVID-19, such as antivirals (radecivir, baritinib), monoclonal antibodies (tocilizumab), and glucocorticoids (dexamethasone). Accumulating evidence indicates that long noncoding RNAs (lncRNAs) are essential regulators of virus infections and antiviral immune responses including biological processes that are involved in the regulation of COVID-19 and subsequent disease states. Upon viral infections, cellular lncRNAs directly regulate viral genes and influence viral replication and pathology through virus-mediated changes in the host transcriptome. Additionally, several host lncRNAs could help the occurrence of viral immune escape by inhibiting type I interferons (IFN-1), while others could up-regulate IFN-1 production to play an antiviral role. Consequently, understanding the expression and function of lncRNAs during severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection will provide insights into the development of lncRNA-based methods. In this review, we summarized the current findings of lncRNAs in the regulation of the strong inflammatory response, immune dysfunction and thrombosis induced by SARS-CoV-2 infection, discussed the underlying mechanisms, and highlighted the therapeutic challenges of COVID-19 treatment and its future research directions.


Sujet(s)
COVID-19/immunologie , Interactions hôte-microbes/génétique , Immunité innée/génétique , ARN long non codant/métabolisme , Thrombose/immunologie , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Marqueurs biologiques/analyse , COVID-19/complications , COVID-19/génétique , Dépistage de la COVID-19/méthodes , Cytokines/génétique , Cytokines/métabolisme , Régulation de l'expression des gènes viraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes viraux/immunologie , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Interactions hôte-microbes/immunologie , Humains , Échappement immunitaire/génétique , Pandémies/prévention et contrôle , ARN long non codant/analyse , ARN long non codant/antagonistes et inhibiteurs , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité , Transduction du signal/génétique , Transduction du signal/immunologie , Thrombose/génétique , Thrombose/virologie , Réplication virale/effets des médicaments et des substances chimiques , Réplication virale/génétique , Réplication virale/immunologie , Traitements médicamenteux de la COVID-19
17.
Viruses ; 13(8)2021 08 03.
Article de Anglais | MEDLINE | ID: mdl-34452398

RÉSUMÉ

Venezuelan equine encephalitis virus (VEEV) is a new world alphavirus and a category B select agent. Currently, no FDA-approved vaccines or therapeutics are available to treat VEEV exposure and resultant disease manifestations. The C-terminus of the VEEV non-structural protein 3 (nsP3) facilitates cell-specific and virus-specific host factor binding preferences among alphaviruses, thereby providing targets of interest when designing novel antiviral therapeutics. In this study, we utilized an overexpression construct encoding HA-tagged nsP3 to identify host proteins that interact with VEEV nsP3 by mass spectrometry. Bioinformatic analyses of the putative interactors identified 42 small molecules with the potential to inhibit the host interaction targets, and thus potentially inhibit VEEV. Three inhibitors, tomatidine, citalopram HBr, and Z-VEID-FMK, reduced replication of both the TC-83 strain and the Trinidad donkey (TrD) strain of VEEV by at least 10-fold in astrocytoma, astroglial, and microglial cells. Further, these inhibitors reduced replication of the related New World (NW) alphavirus Eastern equine encephalitis virus (EEEV) in multiple cell types, thus demonstrating broad-spectrum antiviral activity. Time-course assays revealed all three inhibitors reduced both infectious particle production and positive-sense RNA levels post-infection. Further evaluation of the putative host targets for the three inhibitors revealed an interaction of VEEV nsP3 with TFAP2A, but not eIF2S2. Mechanistic studies utilizing siRNA knockdowns demonstrated that eIF2S2, but not TFAP2A, supports both efficient TC-83 replication and genomic RNA synthesis, but not subgenomic RNA translation. Overall, this work reveals the composition of the VEEV nsP3 proteome and the potential to identify host-based, broad spectrum therapeutic approaches to treat new world alphavirus infections.


Sujet(s)
Antiviraux/pharmacologie , Virus de l'encéphalite équine du Venezuela/effets des médicaments et des substances chimiques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Protéines virales non structurales/antagonistes et inhibiteurs , Réplication virale/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire , Chlorocebus aethiops , Virus de l'encéphalite équine du Venezuela/génétique , Equus caballus , Humains , Protéome , Cellules Vero , Protéines virales non structurales/classification , Protéines virales non structurales/génétique
18.
Viruses ; 13(8)2021 08 03.
Article de Anglais | MEDLINE | ID: mdl-34452402

RÉSUMÉ

Background: Every year, millions of people are hospitalized and thousands die from influenza A virus (FLUAV) infection. Most cases of hospitalizations and death occur among the elderly. Many of these elderly patients are reliant on medical treatment of underlying chronic diseases, such as arthritis, diabetes, and hypertension. We hypothesized that the commonly prescribed medicines for treatment of underlying chronic diseases can affect host responses to FLUAV infection and thus contribute to the morbidity and mortality associated with influenza. Therefore, the aim of this study was to examine whether commonly prescribed medicines could affect host responses to virus infection in vitro. Methods: We first identified 45 active compounds from a list of commonly prescribed medicines. Then, we constructed a drug-target interaction network and identified the potential implication of these interactions for FLUAV-host cell interplay. Finally, we tested the effect of 45 drugs on the viability, transcription, and metabolism of mock- and FLUAV-infected human retinal pigment epithelial (RPE) cells. Results: In silico drug-target interaction analysis revealed that drugs such as atorvastatin, candesartan, and hydroxocobalamin could target and modulate FLUAV-host cell interaction. In vitro experiments showed that at non-cytotoxic concentrations, these compounds affected the transcription and metabolism of FLUAV- and mock-infected cells. Conclusion: Many commonly prescribed drugs were found to modulate FLUAV-host cell interactions in silico and in vitro and could therefore affect their interplay in vivo, thus contributing to the morbidity and mortality of patients with influenza virus infections.


Sujet(s)
Interactions hôte-microbes/effets des médicaments et des substances chimiques , Sous-type H1N1 du virus de la grippe A/effets des médicaments et des substances chimiques , Grippe humaine/virologie , Médicaments sur ordonnance/pharmacologie , Lignée cellulaire , Simulation numérique , Analyse de profil d'expression de gènes , Humains , Métabolomique , Projets pilotes , Médicaments sur ordonnance/effets indésirables , Étude de validation de principe
19.
Viruses ; 13(8)2021 08 14.
Article de Anglais | MEDLINE | ID: mdl-34452475

RÉSUMÉ

Human cytomegalovirus causes diseases in individuals with insufficient immunity. Cytomegaloviruses exploit the ubiquitin proteasome pathway to manipulate the proteome of infected cells. The proteasome degrades ubiquitinated proteins. The family of cullin RING ubiquitin ligases (CRL) regulates the stability of numerous important proteins. If the cullin within the CRL is modified with Nedd8 ("neddylated"), the CRL is enzymatically active, while CRLs lacking Nedd8 modifications are inactive. The Nedd8-activating enzyme (NAE) is indispensable for neddylation. By binding to NAE and inhibiting neddylation, the drug MLN4924 (pevonedistat) causes CRL inactivation and stabilization of CRL target proteins. We showed that MLN4924 elicits potent antiviral activity against cytomegaloviruses, suggesting that NAE might be a druggable host dependency factor (HDF). However, MLN4924 is a nucleoside analog related to AMP, and the antiviral activity of MLN4924 may have been influenced by off-target effects in addition to NAE inhibition. To test if NAE is indeed an HDF, we assessed the novel NAE inhibitor TAS4464 and observed potent antiviral activity against mouse and human cytomegalovirus. Additionally, we raised an MLN4924-resistant cell clone and showed that MLN4924 as well as TAS4464 lose their antiviral activity in these cells. Our results indicate that NAE, the neddylation process, and CRLs are druggable HDFs of cytomegaloviruses.


Sujet(s)
Antiviraux/pharmacologie , Cytomegalovirus/effets des médicaments et des substances chimiques , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Muromegalovirus/effets des médicaments et des substances chimiques , Ubiquitin-activating enzymes/antagonistes et inhibiteurs , Ubiquitin-activating enzymes/métabolisme , Animaux , Lignée cellulaire , Lignée cellulaire tumorale , Cullines/métabolisme , Cyclopentanes/métabolisme , Cytomegalovirus/pathogénicité , Humains , Souris , Muromegalovirus/pathogénicité , Protéine NEDD8/métabolisme , Maturation post-traductionnelle des protéines , Protéome , Pyrimidines/métabolisme , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Ubiquitin-activating enzymes/génétique , Ubiquitin-protein ligases/métabolisme
20.
Viruses ; 13(6)2021 06 17.
Article de Anglais | MEDLINE | ID: mdl-34204188

RÉSUMÉ

Mayaro virus (MAYV) hijacks the host's cell machinery to effectively replicate. The mitogen-activated protein kinases (MAPKs) p38, JNK, and ERK1/2 have emerged as crucial cellular factors implicated in different stages of the viral cycle. However, whether MAYV uses these MAPKs to competently replicate has not yet been determined. The aim of this study was to evaluate the impact of MAPK inhibition on MAYV replication using primary human dermal fibroblasts (HDFs) and HeLa cells. Viral yields in supernatants from MAYV-infected cells treated or untreated with inhibitors SB203580, SP600125, U0126, or Losmapimod were quantified using plaque assay. Additionally, viral protein expression was analyzed using immunoblot and immunofluorescence. Knockdown of p38⍺/p38ß isoforms was performed in HDFs using the PROTACs molecule NR-7h. Our data demonstrated that HDFs are highly susceptible to MAYV infection. SB203580, a p38 inhibitor, reduced MAYV replication in a dose-dependent manner in both HDFs and HeLa cells. Additionally, SB203580 significantly decreased viral E1 protein expression. Similarly, knockdown or inhibition of p38⍺/p38ß isoforms with NR-7h or Losmapimod, respectively, affected MAYV replication in a dose-dependent manner. Collectively, these findings suggest that p38 could play an important role in MAYV replication and could serve as a therapeutic target to control MAYV infection.


Sujet(s)
Alphavirus/physiologie , Fibroblastes/virologie , Réplication virale/génétique , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , p38 Mitogen-Activated Protein Kinases/génétique , Apoptose , Cellules cultivées , Cyclopropanes/pharmacologie , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Cellules HeLa , Interactions hôte-microbes/effets des médicaments et des substances chimiques , Humains , Imidazoles/pharmacologie , Système de signalisation des MAP kinases , Phosphorylation , Pyridines/pharmacologie , Peau/cytologie , Peau/virologie , Réplication virale/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...