Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 19.475
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000469

RÉSUMÉ

Coronavirus can cause various diseases, from mild symptoms to the recent severe COVID-19. The coronavirus RNA genome is frequently mutated due to its RNA nature, resulting in many pathogenic and drug-resistant variants. Therefore, many medicines should be prepared to respond to the various coronavirus variants. In this report, we demonstrated that Forsythia viridissima fruit ethanol extract (FVFE) effectively reduces coronavirus replication. We attempted to identify the active compounds and found that actigenin from FVFE effectively reduces human coronavirus replication. Arctigenin treatment can reduce coronavirus protein expression and coronavirus-induced cytotoxicity. These results collectively suggest that arctigenin is a potent natural compound that prevents coronavirus replication.


Sujet(s)
Forsythia , Fruit , Furanes , Lignanes , Extraits de plantes , Réplication virale , Forsythia/composition chimique , Lignanes/pharmacologie , Réplication virale/effets des médicaments et des substances chimiques , Furanes/pharmacologie , Humains , Fruit/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , SARS-CoV-2/effets des médicaments et des substances chimiques , SARS-CoV-2/physiologie , Antiviraux/pharmacologie , Antiviraux/composition chimique , Animaux , Chlorocebus aethiops , Cellules Vero
2.
Virol J ; 21(1): 154, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978059

RÉSUMÉ

BACKGROUND: Rabies is a fatal zoonotic disease whose pathogenesis has not been fully elucidated, and vaccination is the only effective method for protecting against rabies virus infection. Most inactivated vaccines are produced using Vero cells, which are African green monkey kidney cells, to achieve large-scale production. However, there is a potential carcinogenic risk due to nonhuman DNA contamination. Thus, replacing Vero cells with human diploid cells may be a safer strategy. In this study, we developed a novel 2BS cell-adapted rabies virus strain and analysed its sequence, virulence and immunogenicity to determine its application potential as a human diploid cell inactivated vaccine. METHODS AND RESULTS: The 2BS cell-adapted rabies virus strain 2aG4-B40 was established by passage for 40 generations and selection of plaques in 2BS cells. RNA sequence analysis revealed that mutations in 2BS cell-adapted strains were not located at key sites that regulate the production of neutralizing antibodies or virulence in the aG strain (GQ412744.1). The gradual increase in virulence (remaining above 7.0 logLD50/ml from the 40th to 55th generation) and antigen further indicated that these mutations may increase the affinity of the adapted strains for human diploid cells. Identification tests revealed that the 2BS cell-adapted virus strain was neutralized by anti-rabies serum, with a neutralization index of 19,952. PrEP and PEP vaccination and the NIH test further indicated that the vaccine prepared with the 2aG4-B40 strain had high neutralizing antibody levels (2.24 to 46.67 IU/ml), immunogenicity (protection index 270) and potency (average 11.6 IU/ml). CONCLUSIONS: In this study, a 2BS cell-adapted strain of the 2aG4 rabies virus was obtained by passage for 40 generations. The results of sequencing analysis and titre determination of the adapted strain showed that the mutations in the adaptive process are not located at key sequence regions of the virus, and these mutations may enhance the affinity of the adapted strain for human diploid cells. Moreover, vaccines made from the adapted strain 2aG4-B40 had high potency and immunogenicity and could be an ideal candidate rabies virus strain for inactivated vaccine preparation.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins antirabiques , Virus de la rage , Rage (maladie) , Virus de la rage/immunologie , Virus de la rage/génétique , Virus de la rage/pathogénicité , Animaux , Vaccins antirabiques/immunologie , Vaccins antirabiques/génétique , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Rage (maladie)/prévention et contrôle , Rage (maladie)/immunologie , Rage (maladie)/virologie , Humains , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Chlorocebus aethiops , Virulence , Vaccins inactivés/immunologie , Cellules Vero , Chine , Souris , Lignée cellulaire , Mutation , Femelle , Immunogénicité des vaccins
3.
Int J Nanomedicine ; 19: 6931-6943, 2024.
Article de Anglais | MEDLINE | ID: mdl-39005960

RÉSUMÉ

Purpose: Over the past three years, extensive research has been dedicated to understanding and combating COVID-19. Targeting the interaction between the SARS-CoV-2 Spike protein and the ACE2 receptor has emerged as a promising therapeutic strategy against SARS-CoV-2. This study aimed to develop ACE2-coated virus-like particles (ACE2-VLPs), which can be utilized to prevent viral entry into host cells and efficiently neutralize the virus. Methods: Virus-like particles were generated through the utilization of a packaging plasmid in conjunction with a plasmid containing the ACE2 envelope sequence. Subsequently, ACE2-VLPs and ACE2-EVs were purified via ultracentrifugation. The quantification of VLPs was validated through multiple methods, including Nanosight 3000, TEM imaging, and Western blot analysis. Various packaging systems were explored to optimize the ACE2-VLP configuration for enhanced neutralization capabilities. The evaluation of neutralization effectiveness was conducted using pseudoviruses bearing different spike protein variants. Furthermore, the study assessed the neutralization potential against the Omicron BA.1 variant in Vero E6 cells. Results: ACE2-VLPs showed a high neutralization capacity even at low doses and demonstrated superior efficacy in in vitro pseudoviral assays compared to extracellular vesicles carrying ACE2. ACE2-VLPs remained stable under various environmental temperatures and effectively blocked all tested variants of concern in vitro. Notably, they exhibited significant neutralization against Omicron BA.1 variant in Vero E6 cells. Given their superior efficacy compared to extracellular vesicles and proven success against live virus, ACE2-VLPs stand out as crucial candidates for treating SARS-CoV-2 infections. Conclusion: This novel therapeutic approach of coating VLPs with receptor particles provides a proof-of-concept for designing effective neutralization strategies for other viral diseases in the future.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Angiotensin-converting enzyme 2/métabolisme , Animaux , Cellules Vero , Chlorocebus aethiops , Humains , COVID-19/virologie , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Anticorps neutralisants/pharmacologie , Cellules HEK293 , Pénétration virale/effets des médicaments et des substances chimiques
4.
J Med Virol ; 96(7): e29782, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39011762

RÉSUMÉ

Extracellular vesicles (EVs) are shown to be a novel viral transmission model capable of increasing a virus's tropism. According to our earlier research, cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or transfected with envelope protein plasmids generate a novel type of EVs that are micrometer-sized and able to encase virus particles. Here, we showed the capacity of these EVs to invade various animals both in vitro and in vivo independent of the angiotensin-converting enzyme 2 receptor. First, via macropinocytosis, intact EVs produced from Vero E6 (monkey) cells were able to enter cells from a variety of animals, including cats, dogs, bats, hamsters, and minks, and vice versa. Second, when given to zebrafish with cutaneous wounds, the EVs showed favorable stability in aqueous environments and entered the fish. Moreover, infection of wild-type (WT) mice with heterogeneous EVs carrying SARS-CoV-2 particles led to a strong cytokine response and a notable amount of lung damage. Conversely, free viral particles did not infect WT mice. These results highlight the variety of processes behind viral transmission and cross-species evolution by indicating that EVs may be possible vehicles for SARS-CoV-2 spillover and raising risk concerns over EVs' potential for viral gene transfer.


Sujet(s)
COVID-19 , Vésicules extracellulaires , SARS-CoV-2 , Animaux , Vésicules extracellulaires/virologie , Vésicules extracellulaires/métabolisme , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , SARS-CoV-2/génétique , COVID-19/transmission , COVID-19/virologie , Souris , Chlorocebus aethiops , Cellules Vero , Humains , Cricetinae , Protéines d'enveloppe des coronavirus/métabolisme , Protéines d'enveloppe des coronavirus/génétique , Chiens , Danio zébré/virologie , Chats , Chiroptera/virologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique
5.
Bull Exp Biol Med ; 177(1): 137-139, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38960960

RÉSUMÉ

We studied toxicity of recombinant Streptococcus pneumoniae pneumolysin protein in experiments on mice and its cytopathogenic effect on cultures of Vero green monkey kidney cells and human lung carcinoma A549 cells in vitro. In vivo and in vitro experiments proved the absence of compromised toxicity and direct cytopathogenic action of the recombinant protein.


Sujet(s)
Protéines bactériennes , Protéines recombinantes , Streptococcus pneumoniae , Streptolysines , Streptolysines/toxicité , Streptolysines/génétique , Animaux , Protéines bactériennes/toxicité , Protéines bactériennes/génétique , Chlorocebus aethiops , Souris , Cellules Vero , Streptococcus pneumoniae/effets des médicaments et des substances chimiques , Humains , Protéines recombinantes/toxicité , Protéines recombinantes/génétique , Cellules A549
6.
Molecules ; 29(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38999077

RÉSUMÉ

Various cancer-associated morbidities remain a growing global health challenge, resulting in a significant burden on healthcare systems worldwide due to high mortality rates and a frequent lack of novel therapeutic options for advanced and localized disease. Reactive oxygen species (ROS) play an important role in cancer pathogenesis and response to chemotherapeutics; therefore, it is crucial to develop novel compounds with both antioxidant and anticancer activity. In this study, a series of previously reported 3-((4-hydroxyphenyl)amino)propanoic acid derivatives (compounds 1-36) were evaluated for their anticancer and antioxidant activities. Compounds 12, 20-22, and 29 were able to reduce A549 cell viability by 50% and suppress A549 cell migration in vitro. These compounds also showed favorable cytotoxicity properties towards noncancerous Vero cells. The most promising candidate, compound 20, exhibited potent antioxidant properties in the DPPH radical scavenging assay. These results demonstrate that 3-((4-hydroxyphenyl)amino)propanoic acid could be further explored as an attractive scaffold for the development of novel anticancer and antioxidant candidates.


Sujet(s)
Antinéoplasiques , Antioxydants , Survie cellulaire , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Antioxydants/pharmacologie , Antioxydants/composition chimique , Cellules Vero , Chlorocebus aethiops , Animaux , Cellules A549 , Survie cellulaire/effets des médicaments et des substances chimiques , Propionates/pharmacologie , Propionates/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Structure moléculaire
7.
Sci Rep ; 14(1): 15363, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965389

RÉSUMÉ

The therapeutic potential of insect-derived bioactive molecules as anti-SARS-CoV-2 agents has shown promising results. Hymenopteran venoms, notably from Apis mellifera (honeybee) and Vespa orientalis (oriental wasp), were examined for the first time in an in vitro setting for their potential anti-COVID-19 activity. This assessment utilized an immunodiagnostic system to detect the SARS-CoV-2 nucleocapsid antigen titer reduction. Further analyses, including cytotoxicity assays, plaque reduction assays, and in silico docking-based screening, were performed to evaluate the efficacy of the most potent venom. Results indicated that bee and wasp venoms contain bioactive molecules with potential therapeutic effects against SARS-CoV-2.Nevertheless, the wasp venom exhibited superior efficacy compared to bee venom, achieving a 90% maximal (EC90) concentration effect of antigen depletion at 0.184 mg/mL, in contrast to 2.23 mg/mL for bee venom. The cytotoxicity of the wasp venom was assessed on Vero E6 cells 48 h post-treatment using the MTT assay. The CC 50 of the cell growth was 0.16617 mg/mL for Vero E6 cells. The plaque reduction assay of wasp venom revealed 50% inhibition (IC50) at a 0.208 mg/mL concentration. The viral count at 50% inhibition was 2.5 × 104 PFU/mL compared to the initial viral count of 5 × 104 PFU/mL. In silico data for the wasp venom revealed a strong attraction to binding sites on the ACE2 protein, indicating ideal interactions. This substantiates the potential of wasp venom as a promising viral inhibitor against SARS-CoV-2, suggesting its consideration as a prospective natural preventive and curative antiviral drug. In conclusion, hymenopteran venoms, particularly wasp venom, hold promise as a source of potential therapeutic biomolecules against SARS-CoV-2. More research and clinical trials are needed to evaluate these results and investigate their potential for translation into innovative antiviral therapies.


Sujet(s)
Antiviraux , Traitements médicamenteux de la COVID-19 , COVID-19 , Simulation de docking moléculaire , SARS-CoV-2 , Venins de guêpe , Cellules Vero , SARS-CoV-2/effets des médicaments et des substances chimiques , Chlorocebus aethiops , Animaux , Humains , Antiviraux/pharmacologie , COVID-19/virologie , Venins de guêpe/pharmacologie , Venins de guêpe/composition chimique , Venins d'abeille/pharmacologie , Venins d'abeille/composition chimique , Égypte , Abeilles , Guêpes
8.
Sci Rep ; 14(1): 15289, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961137

RÉSUMÉ

The limited availability of antivirals for new highly pathogenic strains of virus has become a serious public health. Therefore, news products against these pathogens has become an urgent necessity. Among the multiple sources for news antibiotics and antivirals, insect exudates or their products has become an increasingly frequent option. Insects emerged 350 million years ago and have showed a high adaptability and resistance to the most varied biomes. Their survival for so long, in such different environments, is an indication that they have a very efficient protection against environmental infections, despite not having a developed immune system like mammals. Since the ancient civilizations, the products obtained from the bee have been of great pharmacological importance, being used as antimicrobial, anti-inflammatory, antitumor and several other functions. Investigations of biological activity of propolis have been carried out, mainly in the species Apis mellifera, and its product have showed activity against some important viruses. However, for the Meliponini species, known as stingless bees, there are few studies, either on their chemical composition or on their biological activities. The importance of studying these bees is because they come from regions with native forests, and therefore with many species of plants not yet studied, in addition to which they are regions still free of pesticides, which guarantees a greater fidelity of the obtained data. Previous studies by our group with crude hydroalcoholic extract of propolis demonstrated an intense antiviral activity against Herpes, influenza, and rubella viruses. In this work, we chose to use aqueous extracts, which eliminates the presence of other compounds besides those originally present in propolis, in addition to extracting substances different from those obtained in alcoholic extracts. Therefore, this study aimed to identify, isolate and characterize compounds with antiviral effects from aqueous propolis extracts from Scaptotrigona aff postica, in emerging viruses such as zicavirus, chikungunya, and mayaro virus. The evaluation of the antiviral activity of the crude and purified material was performed by reducing infectious foci in VERO cell cultures. The results obtained with crude propolis, indicate a high reduction of zica virus (64×) and mayaro (128×) when was used 10% v/v of propolis. The reduction of chikungunya virus was of 256 fold, even when was used 5% v/v of propolis. The chemical characterization of the compounds present in the extracts was performed by high-pressure liquid chromatography. Through the purification of propolis by HPLC and mass spectrometry, it was possible to identify and isolate a peak with antiviral activity. This substance showed activity against all viruses tested. When purified fraction was used, the reduction observed was of 16 fold for zicavirus, 32 fold for mayaro virus and 512 fold for chikungunya virus. Likewise, it was observed that the antiviral response was concentration dependent, being more intense when propolis was added 2 h after the viral infection. Now we are carrying out the chemical characterization of the purified compounds that showed antiviral action.


Sujet(s)
Antiviraux , Propolis , Propolis/pharmacologie , Propolis/composition chimique , Animaux , Antiviraux/pharmacologie , Antiviraux/composition chimique , Abeilles , Virus du chikungunya/effets des médicaments et des substances chimiques , Chlorocebus aethiops , Cellules Vero
9.
BMC Vet Res ; 20(1): 288, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961481

RÉSUMÉ

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) mainly causes acute and severe porcine epidemic diarrhea (PED), and is highly fatal in neonatal piglets. No reliable therapeutics against the infection exist, which poses a major global health issue for piglets. Luteolin is a flavonoid with anti-viral activity toward several viruses. RESULTS: We evaluated anti-viral effects of luteolin in PEDV-infected Vero and IPEC-J2 cells, and identified IC50 values of 23.87 µM and 68.5 µM, respectively. And found PEDV internalization, replication and release were significantly reduced upon luteolin treatment. As luteolin could bind to human ACE2 and SARS-CoV-2 main protease (Mpro) to contribute viral entry, we first identified that luteolin shares the same core binding site on pACE2 with PEDV-S by molecular docking and exhibited positive pACE2 binding with an affinity constant of 71.6 µM at dose-dependent increases by surface plasmon resonance (SPR) assay. However, pACE2 was incapable of binding to PEDV-S1. Therefore, luteolin inhibited PEDV internalization independent of PEDV-S binding to pACE2. Moreover, luteolin was firmly embedded in the groove of active pocket of Mpro in a three-dimensional docking model, and fluorescence resonance energy transfer (FRET) assays confirmed that luteolin inhibited PEDV Mpro activity. In addition, we also observed PEDV-induced pro-inflammatory cytokine inhibition and Nrf2-induced HO-1 expression. Finally, a drug resistant mutant was isolated after 10 cell culture passages concomitant with increasing luteolin concentrations, with reduced PEDV susceptibility to luteolin identified at passage 10. CONCLUSIONS: Our results push forward that anti-PEDV mechanisms and resistant-PEDV properties for luteolin, which may be used to combat PED.


Sujet(s)
Antiviraux , Lutéoline , Virus de la diarrhée porcine épidémique , Lutéoline/pharmacologie , Virus de la diarrhée porcine épidémique/effets des médicaments et des substances chimiques , Animaux , Antiviraux/pharmacologie , Chlorocebus aethiops , Cellules Vero , Suidae , Simulation de docking moléculaire , Pénétration virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Lignée cellulaire , Simulation numérique , Maladies des porcs/virologie , Maladies des porcs/traitement médicamenteux
10.
Commun Biol ; 7(1): 839, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987278

RÉSUMÉ

Clostridioides difficile causes a wide range of intestinal diseases through the action of two main cytotoxins, TcdA and TcdB. Ingested spores germinate in the intestine establishing a population of cells that produce toxins and spores. The pathogenicity locus, PaLoc, comprises several genes, including those coding for TcdA/B, for the holin-like TcdE protein, and for TcdR, an auto-regulatory RNA polymerase sigma factor essential for tcdA/B and tcdE expression. Here we show that tcdR, tcdA, tcdB and tcdE are expressed in a fraction of the sporulating cells, in either the whole sporangium or in the forespore. The whole sporangium pattern is due to protracted expression initiated in vegetative cells by σD, which primes the TcdR auto-regulatory loop. In contrast, the forespore-specific regulatory proteins σG and SpoVT control TcdR production and tcdA/tcdB and tcdE expression in this cell. We detected TcdA at the spore surface, and we show that wild type and ΔtcdA or ΔtcdB spores but not ΔtcdR or ΔtcdA/ΔtcdB spores are cytopathic against HT29 and Vero cells, indicating that spores may serve as toxin-delivery vehicles. Since the addition of TcdA and TcdB enhance binding of spores to epithelial cells, this effect may occur independently of toxin production by vegetative cells.


Sujet(s)
Toxines bactériennes , Clostridioides difficile , Spores bactériens , Spores bactériens/métabolisme , Spores bactériens/génétique , Clostridioides difficile/génétique , Clostridioides difficile/métabolisme , Toxines bactériennes/métabolisme , Toxines bactériennes/génétique , Humains , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Régulation de l'expression des gènes bactériens , Animaux , Chlorocebus aethiops , Cellules Vero , Entérotoxines/métabolisme , Entérotoxines/génétique
11.
Proc Natl Acad Sci U S A ; 121(29): e2310421121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976733

RÉSUMÉ

We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Tests de neutralisation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Humains , Anticorps neutralisants/immunologie , Souris , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Anticorps antiviraux/immunologie , Tests de neutralisation/méthodes , Souris transgéniques , Coronavirus humain OC43/immunologie , Coronavirus humain OC43/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/immunologie , Chlorocebus aethiops , Cellules Vero , Macaca mulatta
12.
Antiviral Res ; 228: 105923, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38844175

RÉSUMÉ

There are no approved vaccines or therapeutics for Lassa virus (LASV) infections. To identify compounds with anti-LASV activity, we conducted a cell-based screening campaign at biosafety level 4 and tested almost 60,000 compounds for activity against an infectious reporter LASV. Hits from this screen included several structurally related macrocycles. The most potent, Mac128, had a sub-micromolar EC50 against the reporter virus, inhibited wild-type clade IV LASV, and reduced viral titers by 4 orders of magnitude. Mechanistic studies suggested that Mac128 inhibited viral replication at the level of the polymerase.


Sujet(s)
Antiviraux , Virus de Lassa , Composés macrocycliques , Réplication virale , Virus de Lassa/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , Antiviraux/composition chimique , Réplication virale/effets des médicaments et des substances chimiques , Composés macrocycliques/pharmacologie , Composés macrocycliques/composition chimique , Humains , Animaux , Chlorocebus aethiops , Cellules Vero , Fièvre de Lassa/virologie , Fièvre de Lassa/traitement médicamenteux , Lignée cellulaire , Évaluation préclinique de médicament , Protéines virales/antagonistes et inhibiteurs , Protéines virales/métabolisme , Protéines virales/génétique
13.
J Ethnopharmacol ; 333: 118428, 2024 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-38852639

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Lizhong decoction (LZD) is a frequently utilized traditional Chinese remedy for diarrhea. It is unknown how effective it is as an antiviral against PEDV infection. AIM OF THE STUDY: In vitro and in vivo PEDV infection models were used to evaluate the anti-PEDV potential of LZD extract. MATERIALS AND METHODS: LC-MS was used for qualitative analysis of LZD. The antiviral effect of LZD against PEDV using flow cytometry (FC), Quantitative real-time polymerase chain reaction (QPCR), immunofluorescence assay (IFA) analysis in Vero and IPEC-J2 cells. Additionally, we measured the survival rate, clinical symptoms, body weights, fecal scores, temperature, histological analysis, and viral load in a model of newborn piglets infected with PEDV in order to assess the antiviral impact of LZD in vivo. RESULTS: In total, 648 compounds were identified, including 144 Alkaloids, 128 Terpenoids, etc. LZD effectively suppressed PEDV replication in vitro. According to time of addition experiments, LZD mostly inhibited PEDV during the viral life cycle's replication stages. During PEDV infection, LZD can Significantly decrease the apoptotic rate of IPEC-J2 cells and Vero cells. In comparison to the model group, LZD was able to decrease the viral titers in the infected piglets' intestinal and visceral tissues, ameliorate their intestinal pathology, cause a significant increase in body weight growth and increase the piglet survival rate. CONCLUSION: Our findings indicate that the aqueous solution derived from LZD suppressed PEDV replication both in vitro and in vivo, indicating its potential as a candidate for pharmaceutical development.


Sujet(s)
Infections à coronavirus , Médicaments issus de plantes chinoises , Virus de la diarrhée porcine épidémique , Maladies des porcs , Lignée cellulaire , Infections à coronavirus/traitement médicamenteux , Infections à coronavirus/médecine vétérinaire , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Virus de la diarrhée porcine épidémique/effets des médicaments et des substances chimiques , Suidae , Maladies des porcs/traitement médicamenteux , Cellules Vero , Réplication virale/effets des médicaments et des substances chimiques , Analyse de survie , Résultat thérapeutique , Poids/effets des médicaments et des substances chimiques , Charge virale/effets des médicaments et des substances chimiques , Eau/composition chimique , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique , Antiviraux/composition chimique , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Animaux
14.
Proc Natl Acad Sci U S A ; 121(25): e2316376121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38861603

RÉSUMÉ

Human parainfluenza virus type 3 (HPIV3) is a major pediatric respiratory pathogen lacking available vaccines or antiviral drugs. We generated live-attenuated HPIV3 vaccine candidates by codon-pair deoptimization (CPD). HPIV3 open reading frames (ORFs) encoding the nucleoprotein (N), phosphoprotein (P), matrix (M), fusion (F), hemagglutinin-neuraminidase (HN), and polymerase (L) were modified singly or in combination to generate 12 viruses designated Min-N, Min-P, Min-M, Min-FHN, Min-L, Min-NP, Min-NPM, Min-NPL, Min-PM, Min-PFHN, Min-MFHN, and Min-PMFHN. CPD of N or L severely reduced growth in vitro and was not further evaluated. CPD of P or M was associated with increased and decreased interferon (IFN) response in vitro, respectively, but had little effect on virus replication. In Vero cells, CPD of F and HN delayed virus replication, but final titers were comparable to wild-type (wt) HPIV3. In human lung epithelial A549 cells, CPD F and HN induced a stronger IFN response, viral titers were reduced 100-fold, and the expression of F and HN proteins was significantly reduced without affecting N or P or the relative packaging of proteins into virions. Following intranasal infection in hamsters, replication in the nasal turbinates and lungs tended to be the most reduced for viruses bearing CPD F and HN, with maximum reductions of approximately 10-fold. Despite decreased in vivo replication (and lower expression of CPD F and HN in vitro), all viruses induced titers of serum HPIV3-neutralizing antibodies similar to wt and provided complete protection against HPIV3 challenge. In summary, CPD of HPIV3 yielded promising vaccine candidates suitable for further development.


Sujet(s)
Codon , Virus parainfluenza humain de type 3 , Vaccins atténués , Réplication virale , Animaux , Virus parainfluenza humain de type 3/immunologie , Virus parainfluenza humain de type 3/génétique , Humains , Vaccins atténués/immunologie , Vaccins atténués/génétique , Codon/génétique , Cricetinae , Infections à respirovirus/immunologie , Infections à respirovirus/prévention et contrôle , Infections à respirovirus/virologie , Chlorocebus aethiops , Cellules Vero , Cadres ouverts de lecture/génétique , Mesocricetus , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Vaccins antiviraux/immunologie , Vaccins antiviraux/génétique , Protéines virales/immunologie , Protéines virales/génétique , Vaccins contre les virus para-influenza/immunologie , Vaccins contre les virus para-influenza/génétique
15.
J Biomed Sci ; 31(1): 60, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849802

RÉSUMÉ

BACKGROUND: Flavivirus is a challenge all over the world. The replication of flavivirus takes place within membranous replication compartments (RCs) derived from endoplasmic reticulum (ER). Flavivirus NS1 proteins have been proven essential for the formation of viral RCs by remodeling the ER. The glycosylation of flavivirus NS1 proteins is important for viral replication, yet the underlying mechanism remains unclear. METHODS: HeLa cells were used to visualize the ER remodeling effects induced by NS1 expression. ZIKV replicon luciferase assay was performed with BHK-21 cells. rZIKV was generated from BHK-21 cells and the plaque assay was done with Vero Cells. Liposome co-floating assay was performed with purified NS1 proteins from 293T cells. RESULTS: We found that the glycosylation of flavivirus NS1 contributes to its ER remodeling activity. Glycosylation deficiency of NS1, either through N-glycosylation sites mutations or tunicamycin treatment, compromises its ER remodeling activity and interferes with viral RCs formation. Disruption of NS1 glycosylation results in abnormal aggregation of NS1, rather than reducing its membrane-binding activity. Consequently, deficiency in NS1 glycosylation impairs virus replication. CONCLUSIONS: In summary, our results highlight the significance of NS1 glycosylation in flavivirus replication and elucidate the underlying mechanism. This provides a new strategy for combating flavivirus infections.


Sujet(s)
Protéines virales non structurales , Réplication virale , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Glycosylation , Humains , Animaux , Compartiments de réplication virale/métabolisme , Cellules HeLa , Chlorocebus aethiops , Flavivirus/physiologie , Réticulum endoplasmique/métabolisme , Réticulum endoplasmique/virologie , Cellules Vero
16.
Nat Commun ; 15(1): 5112, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879641

RÉSUMÉ

Virus infectivity is traditionally determined by endpoint titration in cell cultures, and requires complex processing steps and human annotation. Here we developed an artificial intelligence (AI)-powered automated framework for ready detection of virus-induced cytopathic effect (DVICE). DVICE uses the convolutional neural network EfficientNet-B0 and transmitted light microscopy images of infected cell cultures, including coronavirus, influenza virus, rhinovirus, herpes simplex virus, vaccinia virus, and adenovirus. DVICE robustly measures virus-induced cytopathic effects (CPE), as shown by class activation mapping. Leave-one-out cross-validation in different cell types demonstrates high accuracy for different viruses, including SARS-CoV-2 in human saliva. Strikingly, DVICE exhibits virus class specificity, as shown with adenovirus, herpesvirus, rhinovirus, vaccinia virus, and SARS-CoV-2. In sum, DVICE provides unbiased infectivity scores of infectious agents causing CPE, and can be adapted to laboratory diagnostics, drug screening, serum neutralization or clinical samples.


Sujet(s)
Intelligence artificielle , Effet cytopathogène viral , Microscopie , SARS-CoV-2 , Humains , SARS-CoV-2/pathogénicité , SARS-CoV-2/physiologie , Microscopie/méthodes , COVID-19/virologie , , Animaux , Virus de la vaccine/physiologie , Virus de la vaccine/pathogénicité , Salive/virologie , Chlorocebus aethiops , Cellules Vero , Rhinovirus/pathogénicité , Rhinovirus/physiologie , Lignée cellulaire
17.
Emerg Microbes Infect ; 13(1): 2368217, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38865205

RÉSUMÉ

Nipah virus (NiV), a highly pathogenic Henipavirus in humans, has been responsible for annual outbreaks in recent years. Experiments involving live NiV are highly restricted to biosafety level 4 (BSL-4) laboratories, which impedes NiV research. In this study, we developed transcription and replication-competent NiV-like particles (trVLP-NiV) lacking N, P, and L genes. This trVLP-NiV exhibited the ability to infect and continuously passage in cells ectopically expressing N, P, and L proteins while maintaining stable genetic characteristics. Moreover, the trVLP-NiV displayed a favourable safety profile in hamsters. Using the system, we found the NiV nucleoprotein residues interacting with viral RNA backbone affected viral replication in opposite patterns. This engineered system was sensitive to well-established antiviral drugs, innate host antiviral factors, and neutralizing antibodies. We then established a high-throughput screening platform utilizing the trVLP-NiV, leading to the identification of tunicamycin as a potential anti-NiV compound. Evidence showed that tunicamycin inhibited NiV replication by decreasing the infectivity of progeny virions. In conclusion, this trVLP-NiV system provided a convenient and versatile molecular tool for investigating NiV molecular biology and conducting antiviral drug screening under BSL-2 conditions. Its application will contribute to the development of medical countermeasures against NiV infections.


Sujet(s)
Infections à hénipavirus , Virus Nipah , Réplication virale , Virus Nipah/physiologie , Virus Nipah/génétique , Virus Nipah/effets des médicaments et des substances chimiques , Animaux , Cricetinae , Humains , Infections à hénipavirus/virologie , Transcription génétique , Virion/génétique , Protéines virales/génétique , Protéines virales/métabolisme , Antiviraux/pharmacologie , Cellules Vero , Chlorocebus aethiops , Lignée cellulaire , ARN viral/génétique
18.
Front Cell Infect Microbiol ; 14: 1393680, 2024.
Article de Anglais | MEDLINE | ID: mdl-38938877

RÉSUMÉ

Hand, foot, and mouth disease (HFMD) is a common infectious disease caused by enterovirus 71 (EV71) that frequently affects children, leading to severe infections in some cases. In general, when infection occurs, the body upregulates inflammatory responses to eliminate pathogenic microorganisms to protect the host from infection. However, EV71 may inhibit host's innate immunity to promote virus infection. At present, it is not fully understood how EV71 hijack the host cells for its own replication. Toll-like receptor 4 (TLR4), a natural immune receptor, historically associated with bacterial endotoxin-induced inflammatory responses. However, it is still unclear whether and how TLR4 is altered during EV71 infection. In this study, we observed a reduction in both TLR4 protein and gene transcript levels in RD, GES-1, and Vero cells following EV71 infection, as detected by RT-qPCR, immunofluorescence staining and western blot. Furthermore, we observed that the TLR4 downstream molecules of MYD88, p-NF-κB p65, p-TBK1 and related inflammatory cytokines were also reduced, suggesting that antiviral innate immune and inflammatory response were suppressed. To determine the impact of TLR4 changes on EV71 infection, we interfered EV71-infected RD cells with TLR4 agonist or inhibitor and the results showed that activation of TLR4 inhibited EV71 replication, while inhibition of TLR4 promote EV71 replication. Besides, EV71 replication was also promoted in TLR4 siRNA-transfected and EV71-infected RD cells. This suggests that down-regulation the expression of TLR4 by EV71 can inhibit host immune defense to promote EV71 self-replication. This novel mechanism may be a strategy for EV71 to evade host immunity.


Sujet(s)
Entérovirus humain A , Immunité innée , Transduction du signal , Récepteur de type Toll-4 , Réplication virale , Humains , Lignée cellulaire , Chlorocebus aethiops , Cytokines/métabolisme , Entérovirus humain A/immunologie , Syndrome mains-pieds-bouche/immunologie , Syndrome mains-pieds-bouche/virologie , Interactions hôte-pathogène/immunologie , Inflammation/métabolisme , Inflammation/immunologie , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Cellules Vero , Animaux
19.
Nat Commun ; 15(1): 4996, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862527

RÉSUMÉ

Assessing the impact of SARS-CoV-2 on organelle dynamics allows a better understanding of the mechanisms of viral replication. We combine label-free holotomographic microscopy with Artificial Intelligence to visualize and quantify the subcellular changes triggered by SARS-CoV-2 infection. We study the dynamics of shape, position and dry mass of nucleoli, nuclei, lipid droplets and mitochondria within hundreds of single cells from early infection to syncytia formation and death. SARS-CoV-2 infection enlarges nucleoli, perturbs lipid droplets, changes mitochondrial shape and dry mass, and separates lipid droplets from mitochondria. We then used Bayesian network modeling on organelle dry mass states to define organelle cross-regulation networks and report modifications of organelle cross-regulation that are triggered by infection and syncytia formation. Our work highlights the subcellular remodeling induced by SARS-CoV-2 infection and provides an Artificial Intelligence-enhanced, label-free methodology to study in real-time the dynamics of cell populations and their content.


Sujet(s)
Théorème de Bayes , COVID-19 , Gouttelettes lipidiques , Mitochondries , SARS-CoV-2 , SARS-CoV-2/physiologie , Humains , COVID-19/virologie , COVID-19/métabolisme , Mitochondries/métabolisme , Gouttelettes lipidiques/métabolisme , Gouttelettes lipidiques/virologie , Intelligence artificielle , Nucléole/métabolisme , Nucléole/virologie , Réplication virale , Noyau de la cellule/métabolisme , Noyau de la cellule/virologie , Animaux , Chlorocebus aethiops , Cellules Vero
20.
Commun Biol ; 7(1): 721, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862613

RÉSUMÉ

The genome folds into complex configurations and structures thought to profoundly impact its function. The intricacies of this dynamic structure-function relationship are not well understood particularly in the context of viral infection. To unravel this interplay, here we provide a comprehensive investigation of simultaneous host chromatin structural (via Hi-C and ATAC-seq) and functional changes (via RNA-seq) in response to vaccinia virus infection. Over time, infection significantly impacts global and local chromatin structure by increasing long-range intra-chromosomal interactions and B compartmentalization and by decreasing chromatin accessibility and inter-chromosomal interactions. Local accessibility changes are independent of broad-scale chromatin compartment exchange (~12% of the genome), underscoring potential independent mechanisms for global and local chromatin reorganization. While infection structurally condenses the host genome, there is nearly equal bidirectional differential gene expression. Despite global weakening of intra-TAD interactions, functional changes including downregulated immunity genes are associated with alterations in local accessibility and loop domain restructuring. Therefore, chromatin accessibility and local structure profiling provide impactful predictions for host responses and may improve development of efficacious anti-viral counter measures including the optimization of vaccine design.


Sujet(s)
Chromatine , Virus de la vaccine , Chromatine/métabolisme , Chromatine/génétique , Animaux , Virus de la vaccine/génétique , Virus de la vaccine/physiologie , Chlorocebus aethiops , Cellules Vero , Vaccine/virologie , Vaccine/immunologie , Interactions hôte-pathogène/génétique , Multi-omique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...