Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.380
Filter
1.
PLoS One ; 19(5): e0304534, 2024.
Article in English | MEDLINE | ID: mdl-38820303

ABSTRACT

Highly variable pandemic coronavirus SARS-CoV-2, which causes the hazardous COVID-19 infection, has been persistent in the human population since late 2019. A prompt assessment of individual and herd immunity against the infection can be accomplished by using rapid tests to determine antiviral antibody levels. The microneutralization assay (MN) is one of the most widely used diagnostic methods that has been proposed to assess the qualitative and quantitative characteristics of virus-specific humoral immunity in COVID-19 convalescents or vaccine recipients. However, some aspects of the assay, such as sensitivity and time cost, need improvement. Here, we developed an express test, which may be potentially used in clinical practice for the assessment of serum-caused SARS-CoV-2 inhibition in infected cell cultures. It implies the detection and counting of coronaviral fluorescent-forming units (FFU) and includes two sequentially used developing components: biotinylated mouse monoclonal antibodies against the recombinant N protein of SARS-CoV-2 (B.1) and the recombinant EGFP-streptavidin fusion protein. Due to the universal specificity of the antibodies, our analytical tool is suitable for the detection of various strains of SARS-CoV-2 when determining both the infectious titer of viruses and the titer of serum virus-neutralizing antibodies. The developed two-component test system is characterized by high sensitivity, a reduced number of analytic stages and low assay cost, as well as by flexibility, since it may be modified for detection of other pathogens using the appropriate antibodies.


Subject(s)
Antibodies, Viral , COVID-19 , SARS-CoV-2 , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Humans , COVID-19/diagnosis , COVID-19/virology , COVID-19/immunology , Animals , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vero Cells , Chlorocebus aethiops , Mice , Fluorescent Antibody Technique/methods , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood
2.
Sci Transl Med ; 16(748): eadj4504, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776389

ABSTRACT

Despite the wide availability of several safe and effective vaccines that prevent severe COVID-19, the persistent emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that can evade vaccine-elicited immunity remains a global health concern. In addition, the emergence of SARS-CoV-2 VOCs that can evade therapeutic monoclonal antibodies underscores the need for additional, variant-resistant treatment strategies. Here, we characterize the antiviral activity of GS-5245, obeldesivir (ODV), an oral prodrug of the parent nucleoside GS-441524, which targets the highly conserved viral RNA-dependent RNA polymerase (RdRp). We show that GS-5245 is broadly potent in vitro against alphacoronavirus HCoV-NL63, SARS-CoV, SARS-CoV-related bat-CoV RsSHC014, Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV-2 WA/1, and the highly transmissible SARS-CoV-2 BA.1 Omicron variant. Moreover, in mouse models of SARS-CoV, SARS-CoV-2 (WA/1 and Omicron B1.1.529), MERS-CoV, and bat-CoV RsSHC014 pathogenesis, we observed a dose-dependent reduction in viral replication, body weight loss, acute lung injury, and pulmonary function with GS-5245 therapy. Last, we demonstrate that a combination of GS-5245 and main protease (Mpro) inhibitor nirmatrelvir improved outcomes in vivo against SARS-CoV-2 compared with the single agents. Together, our data support the clinical evaluation of GS-5245 against coronaviruses that cause or have the potential to cause human disease.


Subject(s)
Antiviral Agents , Prodrugs , SARS-CoV-2 , Animals , SARS-CoV-2/drug effects , Prodrugs/pharmacology , Prodrugs/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , Mice , Administration, Oral , Chlorocebus aethiops , Vero Cells , COVID-19 Drug Treatment , COVID-19/virology , Virus Replication/drug effects , Nucleosides/pharmacology , Nucleosides/therapeutic use , Nucleosides/chemistry , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Female , Disease Models, Animal
3.
Carbohydr Polym ; 337: 122157, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710573

ABSTRACT

Seaweed polysaccharides, particularly sulfated ones, exhibited potent antiviral activity against a wide variety of enveloped viruses, such as herpes simplex virus and respiratory viruses. Different mechanisms of action were suggested, which may range from preventing infection to intracellular antiviral activity, at different stages of the viral cycle. Herein, we generated two chemically engineered sulfated fucans (C303 and C304) from Cystoseira indica by an amalgamated extraction-sulfation procedure using chlorosulfonic acid-pyridine/N,N-dimethylformamide and sulfur trioxide-pyridine/N,N-dimethylformamide reagents, respectively. These compounds exhibited activity against HSV-1 and RSV with 50 % inhibitory concentration values in the range of 0.75-2.5 µg/mL and low cytotoxicity at concentrations up to 500 µg/mL. The antiviral activities of chemically sulfated fucans (C303 and C304) were higher than the water (C301) and CaCl2 extracted (C302) polysaccharides. Compound C303 had a (1,3)-linked fucan backbone and was branched. Sulfates were present at positions C-2, C-4, and C-2,4 of Fucp, and C-6 of Galp residues of this polymer. Compound C304 had a comparable structure but with more sulfates at C-4 of Fucp residue. Both C303 and C304 were potent antiviral candidates, acting in a dose-dependent manner on the adsorption and other intracellular stages of HSV-1 and RSV replication, in vitro.


Subject(s)
Antiviral Agents , Herpesvirus 1, Human , Polysaccharides , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Chlorocebus aethiops , Herpesvirus 1, Human/drug effects , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Animals , Vero Cells , Humans , Sulfates/chemistry , Sulfates/pharmacology , Respiratory Syncytial Viruses/drug effects
4.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731543

ABSTRACT

Ribonuclease P (RNase P) complexed with an external guide sequence (EGS) represents a promising nucleic acid-based gene targeting approach for gene expression knock-down and modulation. The RNase P-EGS strategy is unique as an EGS can be designed to basepair any mRNA sequence and recruit intracellular RNase P for hydrolysis of the target mRNA. In this study, we provide the first direct evidence that the RNase P-based approach effectively blocks the gene expression and replication of herpes simplex virus 2 (HSV-2), the causative agent of genital herpes. We constructed EGSs to target the mRNA encoding HSV-2 single-stranded DNA binding protein ICP8, which is essential for viral DNA genome replication and growth. In HSV-2 infected cells expressing a functional EGS, ICP8 levels were reduced by 85%, and viral growth decreased by 3000 folds. On the contrary, ICP8 expression and viral growth exhibited no substantial differences between cells expressing no EGS and those expressing a disabled EGS with mutations precluding RNase P recognition. The anti-ICP8 EGS is specific in targeting ICP8 because it only affects ICP8 expression but does not affect the expression of the other viral immediate-early and early genes examined. This study shows the effective and specific anti-HSV-2 activity of the RNase P-EGS approach and demonstrates the potential of EGS RNAs for anti-HSV-2 applications.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 2, Human , Virus Replication , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/physiology , Humans , Ribonuclease P/metabolism , Ribonuclease P/genetics , Animals , Viral Proteins/genetics , Viral Proteins/metabolism , Chlorocebus aethiops , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vero Cells , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , DNA-Binding Proteins
5.
BMC Vet Res ; 20(1): 192, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734600

ABSTRACT

BACKGROUND: Natural antimicrobial agents such as nisin were used to control the growth of foodborne pathogens in dairy products. The current study aimed to examine the inhibitory effect of pure nisin and nisin nanoparticles (nisin NPs) against methicillin resistant Staphylococcus aureus (MRSA) and E.coli O157:H7 during the manufacturing and storage of yoghurt. Nisin NPs were prepared using new, natural, and safe nano-precipitation method by acetic acid. The prepared NPs were characterized using zeta-sizer and transmission electron microscopy (TEM). In addition, the cytotoxicity of nisin NPs on vero cells was assessed using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The minimum inhibitory concentrations (MICs) of nisin and its nanoparticles were determined using agar well-diffusion method. Further, fresh buffalo's milk was inoculated with MRSA or E.coli O157:H7 (1 × 106 CFU/ml) with the addition of either nisin or nisin NPs, and then the inoculated milk was used for yoghurt making. The organoleptic properties, pH and bacterial load of the obtained yoghurt were evaluated during storage in comparison to control group. RESULTS: The obtained results showed a strong antibacterial activity of nisin NPs (0.125 mg/mL) against MRSA and E.coli O157:H7 in comparison with control and pure nisin groups. Notably, complete eradication of MRSA and E.coli O157:H7 was observed in yoghurt formulated with nisin NPs after 24 h and 5th day of storage, respectively. The shelf life of yoghurt inoculated with nisin nanoparticles was extended than those manufactured without addition of such nanoparticles. CONCLUSIONS: Overall, the present study indicated that the addition of nisin NPs during processing of yoghurt could be a useful tool for food preservation against MRSA and E.coli O157:H7 in dairy industry.


Subject(s)
Anti-Bacterial Agents , Escherichia coli O157 , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Nanoparticles , Nisin , Yogurt , Nisin/pharmacology , Nisin/chemistry , Yogurt/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Escherichia coli O157/drug effects , Nanoparticles/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Food Preservatives/pharmacology , Vero Cells , Food Microbiology , Chlorocebus aethiops , Food Preservation/methods
6.
Signal Transduct Target Ther ; 9(1): 125, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734691

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a 'highly transmissible respiratory pathogen, leading to severe multi-organ damage. However, knowledge regarding SARS-CoV-2-induced cellular alterations is limited. In this study, we report that SARS-CoV-2 aberrantly elevates mitochondrial bioenergetics and activates the EGFR-mediated cell survival signal cascade during the early stage of viral infection. SARS-CoV-2 causes an increase in mitochondrial transmembrane potential via the SARS-CoV-2 RNA-nucleocapsid cluster, thereby abnormally promoting mitochondrial elongation and the OXPHOS process, followed by enhancing ATP production. Furthermore, SARS-CoV-2 activates the EGFR signal cascade and subsequently induces mitochondrial EGFR trafficking, contributing to abnormal OXPHOS process and viral propagation. Approved EGFR inhibitors remarkably reduce SARS-CoV-2 propagation, among which vandetanib exhibits the highest antiviral efficacy. Treatment of SARS-CoV-2-infected cells with vandetanib decreases SARS-CoV-2-induced EGFR trafficking to the mitochondria and restores SARS-CoV-2-induced aberrant elevation in OXPHOS process and ATP generation, thereby resulting in the reduction of SARS-CoV-2 propagation. Furthermore, oral administration of vandetanib to SARS-CoV-2-infected hACE2 transgenic mice reduces SARS-CoV-2 propagation in lung tissue and mitigates SARS-CoV-2-induced lung inflammation. Vandetanib also exhibits potent antiviral activity against various SARS-CoV-2 variants of concern, including alpha, beta, delta and omicron, in in vitro cell culture experiments. Taken together, our findings provide novel insight into SARS-CoV-2-induced alterations in mitochondrial dynamics and EGFR trafficking during the early stage of viral infection and their roles in robust SARS-CoV-2 propagation, suggesting that EGFR is an attractive host target for combating COVID-19.


Subject(s)
COVID-19 , ErbB Receptors , Mitochondria , SARS-CoV-2 , Virus Replication , SARS-CoV-2/drug effects , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/drug effects , Humans , Animals , Mice , COVID-19/virology , COVID-19/metabolism , COVID-19/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Virus Replication/drug effects , Energy Metabolism/drug effects , Energy Metabolism/genetics , Vero Cells , Chlorocebus aethiops , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Membrane Potential, Mitochondrial/drug effects , Oxidative Phosphorylation/drug effects , Signal Transduction/drug effects
7.
Proc Natl Acad Sci U S A ; 121(22): e2314166121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38768348

ABSTRACT

The nonstructural protein 1 (Nsp1) of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is a virulence factor that targets multiple cellular pathways to inhibit host gene expression and antiviral response. However, the underlying mechanisms of the various Nsp1-mediated functions and their contributions to SARS-CoV-2 virulence remain unclear. Among the targets of Nsp1 is the mRNA (messenger ribonucleic acid) export receptor NXF1-NXT1, which mediates nuclear export of mRNAs from the nucleus to the cytoplasm. Based on Nsp1 crystal structure, we generated mutants on Nsp1 surfaces and identified an acidic N-terminal patch that is critical for interaction with NXF1-NXT1. Photoactivatable Nsp1 probe reveals the RNA Recognition Motif (RRM) domain of NXF1 as an Nsp1 N-terminal binding site. By mutating the Nsp1 N-terminal acidic patch, we identified a separation-of-function mutant of Nsp1 that retains its translation inhibitory function but substantially loses its interaction with NXF1 and reverts Nsp1-mediated mRNA export inhibition. We then generated a recombinant (r)SARS-CoV-2 mutant on the Nsp1 N-terminal acidic patch and found that this surface is key to promote NXF1 binding and inhibition of host mRNA nuclear export, viral replication, and pathogenicity in vivo. Thus, these findings provide a mechanistic understanding of Nsp1-mediated mRNA export inhibition and establish the importance of this pathway in the virulence of SARS-CoV-2.


Subject(s)
Active Transport, Cell Nucleus , COVID-19 , Nucleocytoplasmic Transport Proteins , RNA, Messenger , RNA-Binding Proteins , SARS-CoV-2 , Viral Nonstructural Proteins , Humans , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/genetics , Animals , COVID-19/virology , COVID-19/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Virus Replication , Cell Nucleus/metabolism , Vero Cells , Virulence , Chlorocebus aethiops , HEK293 Cells
8.
BMC Mol Cell Biol ; 25(1): 15, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741034

ABSTRACT

BACKGROUND: Transfection is an important analytical method for studying gene expression in the cellular environment. There are some barriers to efficient DNA transfection in host cells, including circumventing the plasma membrane, escaping endosomal compartmentalization, autophagy, immune sensing pathways, and translocating the nuclear envelope. Therefore, it would be very useful to introduce an optimum transfection approach to achieve a high transfection efficiency in the Vero cell line. The aim of this study was to compare various transfection techniques and introduce a highly efficient method for gene delivery in Vero cells. METHODS: In the current study, three transfection methods were used, including chemical transfection, electroporation, and lentiviral vector transduction, to obtain the optimum transfection conditions in the Vero cell line. Vero cells were cultured and transfected with chemical transfection reagents, electroporation, or HIV-1-based lentivectors under different experimental conditions. Transfection efficiency was assessed using flow cytometry and fluorescence microscopy to detect GFP-positive cells. RESULTS: Among the tested methods, TurboFect™ chemical transfection exhibited the highest efficiency. Optimal transfection conditions were achieved using 1 µg DNA and 4 µL TurboFect™ in 6 × 104 Vero cells. CONCLUSION: TurboFect™, a cationic polymer transfection reagent, demonstrated superior transfection efficiency in Vero cells compared with electroporation and lentivirus particles, and is the optimal choice for chemical transfection in the Vero cell line.


Subject(s)
Electroporation , Genetic Vectors , Transfection , Animals , Chlorocebus aethiops , Vero Cells , Electroporation/methods , Transfection/methods , Genetic Vectors/genetics , Lentivirus/genetics , Transduction, Genetic/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans
9.
Methods Mol Biol ; 2808: 1-7, 2024.
Article in English | MEDLINE | ID: mdl-38743358

ABSTRACT

We have adopted a real-time assay based on a dual-split reporter to assess cell-cell fusion mediated by the measles virus (MeV) membrane fusion machinery. This reporter system is comprised of two expression vectors, each encoding a segment of Renilla luciferase fused to a segment of GFP. To regain function, the two segments need to associate, which is dependent on cell-cell fusion between effector cells expressing the MeV fusion machinery and target cells expressing the corresponding MeV receptor. By measuring reconstituted luciferase activity, we can follow the kinetics of cell-cell fusion and quantify the extent of fusion. This assay lends itself to the study of the MeV fusion machinery comprised of the attachment and fusion glycoproteins, the matrix protein, and the MeV receptors. Moreover, entry inhibitors targeting attachment or fusion can be readily screened using this assay. Finally, this assay can be easily adopted to study the entry of other members of the Paramyxoviridae, as we have demonstrated for the henipaviruses.


Subject(s)
Cell Fusion , Measles virus , Virus Internalization , Measles virus/genetics , Measles virus/physiology , Humans , Animals , Cell Fusion/methods , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Chlorocebus aethiops , Cell Line , Vero Cells , Luciferases, Renilla/genetics , Luciferases, Renilla/metabolism
10.
Methods Mol Biol ; 2808: 209-224, 2024.
Article in English | MEDLINE | ID: mdl-38743373

ABSTRACT

The plaque reduction neutralization test (PRNT) and the enzyme-linked immunosorbent assay (ELISA) are both widely used to assess immunity to infectious diseases such as measles, but they use two different measurement principles: ELISA measures the ability of antibodies to bind to virus components, while the PRNT detects the aptitude of antibodies to prevent the infection of a susceptible cell. As a result, detection of measles virus (MV) neutralizing antibodies is the gold standard for assessing immunity to measles. However, the assay is laborious and requires experience and excellent technical skills. In addition, the result is only available after several days. Therefore, the classical PRNT is not suitable for high-throughput testing. By using an immunocolorimetric assay (ICA) to detect MV-infected cells, the standard PRNT has been developed into a focus reduction neutralization test (FRNT). This assay is faster and has improved specificity. The FRNT described here is extremely useful when immunity to measles virus needs to be assessed in patients with a specific medical condition, such as immunocompromised individuals in whom presumed residual immunity needs to be assessed. The FRNT is not generally recommended for use with large numbers of specimens, such as in a seroprevalence study.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Measles virus , Measles , Neutralization Tests , Neutralization Tests/methods , Measles virus/immunology , Measles/immunology , Measles/diagnosis , Measles/virology , Humans , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Chlorocebus aethiops , Animals , Vero Cells , Viral Plaque Assay/methods , Enzyme-Linked Immunosorbent Assay/methods
11.
Org Biomol Chem ; 22(19): 3986-3994, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38695061

ABSTRACT

Algae-based marine carbohydrate drugs are typically decorated with negative ion groups such as carboxylate and sulfate groups. However, the precise synthesis of highly sulfated alginates is challenging, thus impeding their structure-activity relationship studies. Herein we achieve a microwave-assisted synthesis of a range of highly sulfated mannuronate glycans with up to 17 sulfation sites by overcoming the incomplete sulfation due to the electrostatic repulsion of crowded polyanionic groups. Although the partially sulfated tetrasaccharide had the highest affinity for the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant, the fully sulfated octasaccharide showed the most potent interference with the binding of the RBD to angiotensin-converting enzyme 2 (ACE2) and Vero E6 cells, indicating that the sulfated oligosaccharides might inhibit the RBD binding to ACE2 in a length-dependent manner.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antiviral Agents , Microwaves , Polysaccharides , SARS-CoV-2 , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Chlorocebus aethiops , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/chemistry , Vero Cells , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polysaccharides/chemical synthesis , Humans , Animals , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Hexuronic Acids/chemical synthesis , Sulfates/chemistry , Sulfates/pharmacology , Sulfates/chemical synthesis , COVID-19 Drug Treatment , Structure-Activity Relationship
12.
Biochem Biophys Res Commun ; 716: 149954, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38704887

ABSTRACT

Membrane lipids and proteins form dynamic domains crucial for physiological and pathophysiological processes, including viral infection. Many plasma membrane proteins, residing within membrane domains enriched with cholesterol (CHOL) and sphingomyelin (SM), serve as receptors for attachment and entry of viruses into the host cell. Among these, human coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use proteins associated with membrane domains for initial binding and internalization. We hypothesized that the interaction of lipid-binding proteins with CHOL in plasma membrane could sequestrate lipids and thus affect the efficiency of virus entry into host cells, preventing the initial steps of viral infection. We have prepared CHOL-binding proteins with high affinities for lipids in the plasma membrane of mammalian cells. Binding of the perfringolysin O domain four (D4) and its variant D4E458L to membrane CHOL impaired the internalization of the receptor-binding domain of the SARS-CoV-2 spike protein and the pseudovirus complemented with the SARS-CoV-2 spike protein. SARS-CoV-2 replication in Vero E6 cells was also decreased. Overall, our results demonstrate that the integrity of CHOL-rich membrane domains and the accessibility of CHOL in the membrane play an essential role in SARS-CoV-2 cell entry.


Subject(s)
Cell Membrane , Cholesterol , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Vero Cells , Chlorocebus aethiops , Cholesterol/metabolism , Animals , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Cell Membrane/metabolism , Cell Membrane/virology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Humans , Carrier Proteins/metabolism , COVID-19/virology , COVID-19/metabolism , Protein Binding
13.
Nat Commun ; 15(1): 4018, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740820

ABSTRACT

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Subject(s)
Dependovirus , Gene Editing , Herpes Simplex , Herpesvirus 1, Human , Viral Load , Virus Shedding , Animals , Gene Editing/methods , Female , Dependovirus/genetics , Mice , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Herpes Simplex/genetics , Herpes Simplex/virology , Herpes Simplex/therapy , Disease Models, Animal , Virus Latency/genetics , Humans , Genetic Vectors/genetics , Vero Cells , Genetic Therapy/methods , Herpes Genitalis/therapy , Herpes Genitalis/virology , DNA, Viral/genetics
14.
Sci Rep ; 14(1): 10942, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740839

ABSTRACT

Pradimicin U is a new dihydrobenzo[a]naphthacenequinone compound found to be active on a screen designed to investigate compounds with antimicrobial activity, produced by the actinomycete designated strain FMUSA5-5T. The strain was isolated from a bio-fertilizer of Musa spp. collected from Suphanburi province, Thailand. The chemotaxonomic characteristics and 16S rRNA gene analysis revealed that strain FMUSA5-5T is a member of the genus Nonomuraea. Low genome-based taxonomic criteria, average nucleotide identity (ANI) (82.8-88.3%), average amino-acid identity (AAI) (79.4-87.3%), and digital DNA-DNA hybridization (dDDH) (29.5-38.5%) values and several phenotypic differences between strain FMUSA5-5T and its closest type strains of the genus Nonomuraea indicated that strain FMUSA5-5T represents a novel species of the genus Nonomuraea and the name Nonomuraea composti sp. nov. is proposed for the strain. The crude extract from the culture broth of strain FMUSA5-5T displayed promising antimicrobial activity against several pathogens and led to the isolation of a novel secondary metabolite, pradimicin U. Interestingly, this compound displayed a broad spectrum of biological activities such as antimalarial activity against Plasmodium falciparum K1 (IC50 value = 3.65 µg/mL), anti-Mycobacterium tuberculosis H37Ra (MIC value = 25.0 µg/mL), anti-Alternaria brassicicola BCC 42724 (MIC value = 25.0 µg/mL), anti-Bacillus cereus ATCC 11778 and anti-Staphylococcus aureus ATCC 29213 (MIC values = 6.25 and 1.56 µg/mL, respectively). Moreover, the compound possessed strong anti-human small cell lung cancer (NCI-H187) activity with IC50 value of 5.69 µg/mL, while cytotoxicity against human breast cancer (MCF-7) and Vero cells was very weak (IC50 values of 52.49 and 21.84 µg/mL, respectively).


Subject(s)
Anti-Infective Agents , RNA, Ribosomal, 16S , Humans , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , RNA, Ribosomal, 16S/genetics , Microbial Sensitivity Tests , Phylogeny , Actinomycetales/genetics , Actinomycetales/isolation & purification , Animals , Thailand , Vero Cells , Musa/microbiology , Plasmodium falciparum/drug effects , Chlorocebus aethiops
15.
Methods Cell Biol ; 187: 99-116, 2024.
Article in English | MEDLINE | ID: mdl-38705632

ABSTRACT

Correlative Light Electron Microscopy (CLEM) is a powerful technique to investigate the ultrastructure of specific cells and organelles at sub-cellular resolution. Transmission Electron Microscopy (TEM) is particularly useful to the field of virology, given the small size of the virion, which is below the limit of detection by light microscopy. Furthermore, viral infection results in the rearrangement of host organelles to form spatially defined compartments that facilitate the replication of viruses. With the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there has been great interest to study the viral replication complex using CLEM. In this chapter we provide an exemplary workflow describing the safe preparation and processing of cells grown on coverslips and infected with SARS-CoV-2.


Subject(s)
COVID-19 , SARS-CoV-2 , SARS-CoV-2/ultrastructure , Humans , COVID-19/virology , Vero Cells , Chlorocebus aethiops , Animals , Microscopy, Electron, Transmission/methods , Virus Replication , Microscopy, Electron/methods
16.
Biol Pharm Bull ; 47(5): 930-940, 2024.
Article in English | MEDLINE | ID: mdl-38692871

ABSTRACT

The coronavirus disease 2019 (COVID-19) is caused by the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19, with the recurrent epidemics of new variants of SARS-CoV-2, remains a global public health problem, and new antivirals are still required. Some cholesterol derivatives, such as 25-hydroxycholesterol, are known to have antiviral activity against a wide range of enveloped and non-enveloped viruses, including SARS-CoV-2. At the entry step of SARS-CoV-2 infection, the viral envelope fuses with the host membrane dependent of viral spike (S) glycoproteins. From the screening of cholesterol derivatives, we found a new compound 26,27-dinorcholest-5-en-24-yne-3ß,20-diol (Nat-20(S)-yne) that inhibited the SARS-CoV-2 S protein-dependent membrane fusion in a syncytium formation assay. Nat-20(S)-yne exhibited the inhibitory activities of SARS-CoV-2 pseudovirus entry and intact SARS-CoV-2 infection in a dose-dependent manner. Among the variants of SARS-CoV-2, inhibition of infection by Nat-20(S)-yne was stronger in delta and Wuhan strains, which predominantly invade into cells via fusion at the plasma membrane, than in omicron strains. The interaction between receptor-binding domain of S proteins and host receptor ACE2 was not affected by Nat-20(S)-yne. Unlike 25-hydroxycholesterol, which regulates various steps of cholesterol metabolism, Nat-20(S)-yne inhibited only de novo cholesterol biosynthesis. As a result, plasma membrane cholesterol content was substantially decreased in Nat-20(S)-yne-treated cells, leading to inhibition of SARS-CoV-2 infection. Nat-20(S)-yne having a new mechanism of action may be a potential therapeutic candidate for COVID-19.


Subject(s)
Antiviral Agents , COVID-19 , Cholesterol , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Humans , COVID-19/virology , Cholesterol/metabolism , Vero Cells , Chlorocebus aethiops , Spike Glycoprotein, Coronavirus/metabolism , Animals , Virus Internalization/drug effects , Betacoronavirus/drug effects , Pandemics , COVID-19 Drug Treatment , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Angiotensin-Converting Enzyme 2/metabolism , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology
17.
BMC Complement Med Ther ; 24(1): 180, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698382

ABSTRACT

BACKGROUND: Dioscorea bulbifera Linn. has been used for wound care in Thailand. However, a comprehensive evaluation of its antibacterial activity is required. This study aimed to investigate the antibacterial efficacy of D. bulbifera extract against skin-associated bacteria and isolate and characterize its active antibacterial agent, flavanthrinin. METHODS: Air-dried bulbils of D. bulbifera were pulverised and extracted with hexane, dichloromethane, ethyl acetate, methanol, ethanol, and distilled water; vacuum filtered; concentrated; freeze-dried; and stored at -20 ºC. Antibacterial activity of the extracts was assessed using microdilution techniques against several skin-associated bacteria. Thin-layer chromatography (TLC) bioautography was used to identify the active compounds in the extract, which were fractionated by column chromatography and purified by preparative TLC. The chemical structures of the purified compounds were analysed using nuclear magnetic resonance (NMR). The cytotoxicity of the extract and its active compounds was evaluated in Vero cells. RESULTS: The ethyl acetate extract exhibited distinct inhibition zones against bacteria compared to other extracts. Therefore, the ethyl acetate extract of D. bulbifera in the ethyl acetate layer was used for subsequent analyses. D. bulbifera extract exhibited antibacterial activity, with minimum inhibitory concentrations (MICs) of 0.78-1.56 mg/mL. An active compound, identified through TLC-bioautography, demonstrated enhanced antibacterial activity, with MICs of 0.02-0.78 mg/mL. NMR analysis identified this bioactive compound as flavanthrinin. Both D. bulbifera extract and flavanthrinin-containing fraction demonstrated potent antibacterial activity against Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), and S. epidermidis. The flavanthrinin containing fraction demonstrated low cytotoxicity against Vero cells, showing CC50 values of 0.41 ± 0.03 mg/mL. These values are lower than the MIC value, indicating that this fraction is safer than the initial ethyl acetate extract. CONCLUSIONS: Dioscorea bulbifera extract and its bioactive component flavanthrinin demonstrated significant antibacterial activity against the skin-associated bacteria Staphylococci, including MRSA. Flavanthrinin has potential as a complementary therapeutic agent for managing skin infections owing to its potent antibacterial effects and low cytotoxicity.


Subject(s)
Anti-Bacterial Agents , Dioscorea , Microbial Sensitivity Tests , Plant Extracts , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Vero Cells , Chlorocebus aethiops , Animals , Dioscorea/chemistry , Thailand , Bacteria/drug effects
18.
BMC Biotechnol ; 24(1): 28, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38702622

ABSTRACT

Scientists know very little about the mechanisms underlying fish skin mucus, despite the fact that it is a component of the immune system. Fish skin mucus is an important component of defence against invasive infections. Recently, Fish skin and its mucus are gaining interest among immunologists. Characterization was done on the obtained silver nanoparticles Ag combined with Clarias gariepinus catfish epidermal mucus proteins (EMP-Ag-NPs) through UV-vis, FTIR, XRD, TEM, and SEM. Ag-NPs ranged in size from 4 to 20 nm, spherical in form and the angles were 38.10°, 44.20°, 64.40°, and 77.20°, Where wavelength change after formation of EMP-Ag-NPs as indicate of dark brown, the broad band recorded at wavelength at 391 nm. Additionally, the antimicrobial, antibiofilm and anticancer activities of EMP-Ag-NPs was assessed. The present results demonstrate high activity against unicellular fungi C. albicans, followed by E. faecalis. Antibiofilm results showed strong activity against both S. aureus and P. aeruginosa pathogens in a dose-dependent manner, without affecting planktonic cell growth. Also, cytotoxicity effect was investigated against normal cells (Vero), breast cancer cells (Mcf7) and hepatic carcinoma (HepG2) cell lines at concentrations (200-6.25 µg/mL) and current results showed highly anticancer effect of Ag-NPs at concentrations 100, 5 and 25 µg/mL exhibited rounding, shrinkage, deformation and granulation of Mcf7 and HepG2 with IC50 19.34 and 31.16 µg/mL respectively while Vero cells appeared rounded at concentration 50 µg/mL and normal shape at concentration 25, 12.5 and 6.25 µg/ml with IC50 35.85 µg/mL. This study evidence the potential efficacy of biologically generated Ag-NPs as a substitute medicinal agent against harmful microorganisms. Furthermore, it highlights their inhibitory effect on cancer cell lines.


Subject(s)
Biofilms , Catfishes , Metal Nanoparticles , Silver , Metal Nanoparticles/chemistry , Biofilms/drug effects , Biofilms/growth & development , Silver/chemistry , Silver/pharmacology , Animals , Humans , Mucus/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Vero Cells , Fish Proteins/pharmacology , Fish Proteins/chemistry , Fish Proteins/metabolism , Chlorocebus aethiops , Cell Line, Tumor , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Candida albicans/drug effects , Epidermis/metabolism
19.
Nat Commun ; 15(1): 4056, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744813

ABSTRACT

The fusion peptide of SARS-CoV-2 spike protein is functionally important for membrane fusion during virus entry and is part of a broadly neutralizing epitope. However, sequence determinants at the fusion peptide and its adjacent regions for pathogenicity and antigenicity remain elusive. In this study, we perform a series of deep mutational scanning (DMS) experiments on an S2 region spanning the fusion peptide of authentic SARS-CoV-2 in different cell lines and in the presence of broadly neutralizing antibodies. We identify mutations at residue 813 of the spike protein that reduced TMPRSS2-mediated entry with decreased virulence. In addition, we show that an F823Y mutation, present in bat betacoronavirus HKU9 spike protein, confers resistance to broadly neutralizing antibodies. Our findings provide mechanistic insights into SARS-CoV-2 pathogenicity and also highlight a potential challenge in developing broadly protective S2-based coronavirus vaccines.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Mutation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Internalization , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Neutralizing/immunology , COVID-19/virology , COVID-19/immunology , Animals , Antibodies, Viral/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism , Chlorocebus aethiops , HEK293 Cells , Vero Cells , Epitopes/immunology , Epitopes/genetics , Cell Line , Mice
20.
Arch Microbiol ; 206(6): 269, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38767708

ABSTRACT

Bacteriocins are ribosomally synthesized bacterial peptides endowed with antibacterial, antiprotozoal, anticancer and antiviral activities. In the present study, we evaluated the antiviral activities of two bacteriocins, enterocin DD14 (EntDD14) and lacticaseicin 30, against herpes simplex virus type 1 (HSV-1), human coronavirus 229E (HCoV-229E) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Vero, Huh7 and Vero E6 cells, respectively. In addition, the interactions of these bacteriocins with the envelope glycoprotein D of HSV-1 and the receptor binding domains of HCoV-229E and SARS-CoV-2 have been computationally evaluated using protein-protein docking and molecular dynamics simulations. HSV-1 replication in Vero cells was inhibited by EntDD14 and, to a lesser extent, by lacticaseicin 30 added to cells after virus inoculation. EntDD14 and lacticaseicin 30 had no apparent antiviral activity against HCoV-229E; however, EntDD14 was able to inhibit SARS-CoV-2 in Vero E6 cells. Further studies are needed to elucidate the antiviral mechanism of these bacteriocins.


Subject(s)
Antiviral Agents , Bacteriocins , SARS-CoV-2 , Bacteriocins/pharmacology , Chlorocebus aethiops , Animals , Antiviral Agents/pharmacology , Vero Cells , Humans , SARS-CoV-2/drug effects , Virus Replication/drug effects , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Molecular Docking Simulation , Molecular Dynamics Simulation , Bridged-Ring Compounds
SELECTION OF CITATIONS
SEARCH DETAIL
...