Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(18)2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37762571

ABSTRACT

Effective therapy against the influenza virus is still an unmet goal. Drugs with antiviral effects exist, but the appearance of resistant viruses pushes towards the discovery of drugs with different mechanisms of action. New anti-influenza molecules should target a good candidate, as a new anti-influenza molecule could be an inhibitor of the influenza A virus hemagglutinin (HA), which plays a key role during the early phases of infection. In previous work, we identified two tetrapeptide sequences, SLDC (1) and SKHS (2), derived from bovine lactoferrin (bLf) C-lobe fragment 418-429, which were able to bind HA and inhibit cell infection at picomolar concentration. Considering the above, the aim of this study was to synthesize a new library of peptidomimetics active against the influenza virus. In order to test their ability to bind HA, we carried out a preliminary screening using biophysical assays such as surface plasmon resonance (SPR) and orthogonal immobilization-free microscale thermophoresis (MST). Biological and computational studies on the most interesting compounds were carried out. The methods applied allowed for the identification of a N-methyl peptide, S(N-Me)LDC, which, through high affinity binding of influenza virus hemagglutinin, was able to inhibit virus-induced hemagglutination and cell infection at picomolar concentration. This small sequence, with high activity, represents a good starting point for the design of new peptidomimetics and small molecules.


Subject(s)
Influenza A virus , Peptidomimetics , Peptidomimetics/pharmacology , Hemagglutinins , Antiviral Agents/pharmacology , Biological Assay
2.
Molecules ; 28(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37241786

ABSTRACT

Respiratory viral diseases are among the most important causes of disability, morbidity, and death worldwide. Due to the limited efficacy or side effects of many current therapies and the increase in antiviral-resistant viral strains, the need to find new compounds to counteract these infections is growing. Since the development of new drugs is a time-consuming and expensive process, numerous studies have focused on the reuse of commercially available compounds, such as natural molecules with therapeutic properties. This phenomenon is generally called drug repurposing or repositioning and represents a valid emerging strategy in the drug discovery field. Unfortunately, the use of natural compounds in therapy has some limitations, due to their poor kinetic performance and consequently reduced therapeutic effect. The advent of nanotechnology in biomedicine has allowed this limitation to be overcome, showing that natural compounds in nanoform may represent a promising strategy against respiratory viral infections. In this narrative review, the beneficial effects of some promising natural molecules, curcumin, resveratrol, quercetin, and vitamin C, which have been already studied both in native form and in nanoform, against respiratory viral infections are presented and discussed. The review focuses on the ability of these natural compounds, analyzed in in vitro and in vivo studies, to counteract inflammation and cellular damage induced by viral infection and provide scientific evidence of the benefits of nanoformulations in increasing the therapeutic potential of these molecules.


Subject(s)
Virus Diseases , Humans , Virus Diseases/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Resveratrol/pharmacology , Inflammation/drug therapy
3.
Int J Mol Sci ; 24(6)2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36982205

ABSTRACT

Viral respiratory tract infections (RTIs) are responsible for significant morbidity and mortality worldwide. A prominent feature of severe respiratory infections, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, is the cytokine release syndrome. Therefore, there is an urgent need to develop different approaches both against viral replication and against the consequent inflammation. N-acetylglucosamine (GlcNAc), a glucosamine (GlcN) derivative, has been developed as an immunomodulatory and anti-inflammatory inexpensive and non-toxic drug for non-communicable disease treatment and/or prevention. Recent studies have suggested that GlcN, due to its anti-inflammatory activity, could be potentially useful for the control of respiratory virus infections. Our present study aimed to evaluate in two different immortalized cell lines whether GlcNAc could inhibit or reduce both viral infectivity and the inflammatory response to viral infection. Two different viruses, frequent cause of upper and lower respiratory tract infections, were used: the H1N1 Influenza A virus (IAV) (as model of enveloped RNA virus) and the Human adenovirus type 2 (Adv) (as model of naked DNA virus). Two forms of GlcNAc have been considered, bulk GlcNAc and GlcNAc in nanoform to overcome the possible pharmacokinetic limitations of GlcNAc. Our study suggests that GlcNAc restricts IAV replication but not Adv infection, whereas nano-GlcNAc inhibits both viruses. Moreover, GlcNAc and mainly its nanoformulation were able to reduce the pro-inflammatory cytokine secretion stimulated by viral infection. The correlation between inflammatory and infection inhibition is discussed.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza A virus , Pneumonia , Respiratory Tract Infections , Virus Diseases , Humans , Antiviral Agents/pharmacology , Acetylglucosamine/pharmacology , SARS-CoV-2 , Respiratory Tract Infections/drug therapy , Anti-Inflammatory Agents/pharmacology , Glucosamine/pharmacology , Adenoviridae
4.
Molecules ; 27(12)2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35744845

ABSTRACT

Influenza viruses are transmitted from human to human via airborne droplets and can be transferred through contaminated environmental surfaces. Some works have demonstrated the efficacy of essential oils (EOs) as antimicrobial and antiviral agents, but most of them examined the liquid phases, which are generally toxic for oral applications. In our study, we describe the antiviral activity of Citrus bergamia, Melaleuca alternifolia, Illicium verum and Eucalyptus globulus vapor EOs against influenza virus type A. In the vapor phase, C. bergamia and M. alternifolia strongly reduced viral cytopathic effect without exerting any cytotoxicity. The E. globulus vapor EO reduced viral infection by 78% with no cytotoxicity, while I. verum was not effective. Furthermore, we characterized the EOs and their vapor phase by the head-space gas chromatography-mass spectrometry technique, observing that the major component found in each liquid EO is the same one of the corresponding vapor phases, with the exception of M. alternifolia. To deepen the mechanism of action, the morphological integrity of virus particles was checked by negative staining transmission electron microscopy, showing that they interfere with the lipid bilayer of the viral envelope, leading to the decomposition of membranes. We speculated that the most abundant components of the vapor EOs might directly interfere with influenza virus envelope structures or mask viral structures important for early steps of viral infection.


Subject(s)
Anti-Infective Agents , Eucalyptus , Influenza A Virus, H1N1 Subtype , Melaleuca , Oils, Volatile , Anti-Infective Agents/pharmacology , Antiviral Agents/pharmacology , Eucalyptus/chemistry , Melaleuca/chemistry , Oils, Volatile/chemistry , Oils, Volatile/pharmacology
5.
J Appl Toxicol ; 36(3): 394-403, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26370214

ABSTRACT

Toxic effects were reported for pristine-multi-wall carbon nanotubes (p-MWCNTs) while the role of the functionalization on MWCNT-induced toxicity is not yet well defined. We evaluated on human alveolar (A549) epithelial cells and normal bronchial (BEAS-2B) cells exposed to p-MWCNTs, MWCNTs-OH and MWCNTs-COOH: uptake by TEM, cell viability by different assays, membrane damage by the LDH assay and cytokine release by ELISA. The aims of the present study were to: (i) confirm MWCNT cytotoxicity mechanisms hypothesized in our previous studies; (ii) identify the most reliable viability assay to screen MWCNT toxicity; and (iii) to test our model to clarify the role of functionalization on MWCNT-induced toxicity. In A549 cells, p-MWCNTs and MWCNTs-OH were localized free in the cytoplasm and inside vacuoles whereas MWCNTs-COOH were confined inside filled cytoplasmic vesicles. WST-1 and Trypan blue assays showed in A549 cells a similar slight viability reduction for all MWCNTs whereas in BEAS-2B cells WST1 showed a high viability reduction at the highest concentrations, particularly for MWCNTs-COOH. The MTT assay showed a false cytotoxicity as a result of MWCNTs-interference. Pristine and MWCNTs-COOH induced membrane damage, particularly in BEAS-2B cells. MWCNTs-COOH induced interleukin-6 (IL-6) and IL-8 release in A549 cells whereas p-MWCNTs induced IL-8 release in BEAS-2B cells. MWCNTs intracellular localization in A549 cells confirms the toxicity mechanisms previously hypothesized, with p-MWCNTs disrupting the membrane and vesicle-confined MWCNTs-COOH inducing inflammation. WST-1 was more reliable than MTT to test MWCNT-toxicity. BEAS-2B cells were more susceptible then A549 cells, particularly to MWCNT-COOH cytotoxicity. Our results confirm the toxicity of p-MWCNTs and demonstrate, also for the two kinds of tested functionalized MWCNTs toxic effects with a different mechanism of action.


Subject(s)
Carboxylic Acids/toxicity , Epithelial Cells/drug effects , Lung/drug effects , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Biological Assay , Carboxylic Acids/chemistry , Carboxylic Acids/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endocytosis , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Humans , Hydroxylation , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , L-Lactate Dehydrogenase/metabolism , Lung/metabolism , Lung/ultrastructure , Microscopy, Electron, Transmission , Nanotubes, Carbon/chemistry , Pneumonia/metabolism , Pneumonia/pathology , Reproducibility of Results , Risk Assessment , Tumor Necrosis Factor-alpha/metabolism
6.
J Virol ; 89(21): 11107-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311877

ABSTRACT

UNLABELLED: Viruses modulate cellular processes and metabolism in diverse ways, but these are almost universally studied in the infected cell itself. Here, we study spatial organization of DNA synthesis during multiround transmission of herpes simplex virus (HSV) using pulse-labeling with ethynyl nucleotides and cycloaddition of azide fluorophores. We report a hitherto unknown and unexpected outcome of virus-host interaction. Consistent with the current understanding of the single-step growth cycle, HSV suppresses host DNA synthesis and promotes viral DNA synthesis in spatially segregated compartments within the cell. In striking contrast, during progressive rounds of infection initiated at a single cell, we observe that infection induces a clear and pronounced stimulation of cellular DNA replication in remote uninfected cells. This induced DNA synthesis was observed in hundreds of uninfected cells at the extended border, outside the perimeter of the progressing infection. Moreover, using pulse-chase analysis, we show that this activation is maintained, resulting in a propagating wave of host DNA synthesis continually in advance of infection. As the virus reaches and infects these activated cells, host DNA synthesis is then shut off and replaced with virus DNA synthesis. Using nonpropagating viruses or conditioned medium, we demonstrate a paracrine effector of uninfected cell DNA synthesis in remote cells continually in advance of infection. These findings have significant implications, likely with broad applicability, for our understanding of the ways in which virus infection manipulates cell processes not only in the infected cell itself but also now in remote uninfected cells, as well as of mechanisms governing host DNA synthesis. IMPORTANCE: We show that during infection initiated by a single particle with progressive cell-cell virus transmission (i.e., the normal situation), HSV induces host DNA synthesis in uninfected cells, mediated by a virus-induced paracrine effector. The field has had no conception that this process occurs, and the work changes our interpretation of virus-host interaction during advancing infection and has implications for understanding controls of host DNA synthesis. Our findings demonstrate the utility of chemical biology techniques in analysis of infection processes, reveal distinct processes when infection is examined in multiround transmission versus single-step growth curves, and reveal a hitherto-unknown process in virus infection, likely relevant for other viruses (and other infectious agents) and for remote signaling of other processes, including transcription and protein synthesis.


Subject(s)
DNA Replication/physiology , Herpes Simplex/physiopathology , Host-Pathogen Interactions/physiology , Paracrine Communication/physiology , Simplexvirus/physiology , Animals , Azides , Bacterial Proteins , Cell Line , Chlorocebus aethiops , Humans , Luminescent Proteins , Phosphonoacetic Acid , Vero Cells , Viral Plaque Assay , Virus Internalization
7.
Philos Trans R Soc Lond B Biol Sci ; 370(1661): 20140038, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25533095

ABSTRACT

The alveolar respiratory unit constitutes one of the main targets of inhaled nanoparticles; the effect of engineered nanomaterials (NMs) on human health is largely unknown. Surfactant protein D (SP-D) is synthesized by alveolar type II epithelial cells and released into respiratory secretions; its main function is in immune defence, notably against inhaled microbes. SP-D also plays an important role in modulating an appropriate inflammatory response in the lung, and reduced SP-D is associated with a number of inflammatory lung diseases. Adsorption of SP-D to inhaled NMs may facilitate their removal via macrophage phagocytosis. This study addresses the hypothesis that the chemistry, size and surface modification of engineered NMs will impact on their interaction with, and adsorption of, SP-D. To this purpose, we have examined the interactions between SP-D in human lung lavage and two NMs, carbon nanotubes and polystyrene nanoparticles, with different surface functionalization. We have demonstrated that particle size, functionalization and concentration affect the adsorption of SP-D from human lung lavage. Functionalization with negatively charged groups enhanced the amount of SP-D binding. While SP-D binding would be expected to enhance macrophage phagocytosis, these results suggest that the degree of binding is markedly affected by the physicochemistry of the NM and that deposition of high levels of some nanoparticles within the alveolar unit might deplete SP-D levels and affect alveolar immune defence mechanisms.


Subject(s)
Bronchoalveolar Lavage Fluid , Nanoparticles/chemistry , Nanotubes, Carbon/chemistry , Polystyrenes/chemistry , Pulmonary Surfactant-Associated Protein D/chemistry , Adsorption , Humans , Particle Size , Protein Binding , Surface Properties
8.
BMC Microbiol ; 11: 25, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21284853

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is an important human pathogen representing a major cause of morbidity and mortality worldwide. We sequenced the genome of a serotype 11A, ST62 S. pneumoniae invasive isolate (AP200), that was erythromycin-resistant due to the presence of the erm(TR) determinant, and carried out analysis of the genome organization and comparison with other pneumococcal genomes. RESULTS: The genome sequence of S. pneumoniae AP200 is 2,130,580 base pair in length. The genome carries 2216 coding sequences (CDS), 56 tRNA, and 12 rRNA genes. Of the CDSs, 72.9% have a predicted biological known function. AP200 contains the pilus islet 2 and, although its phenotype corresponds to serotype 11A, it contains an 11D capsular locus. Chromosomal rearrangements resulting from a large inversion across the replication axis, and horizontal gene transfer events were observed. The chromosomal inversion is likely implicated in the rebalance of the chromosomal architecture affected by the insertions of two large exogenous elements, the erm(TR)-carrying Tn1806 and a functional prophage designated φSpn_200. Tn1806 is 52,457 bp in size and comprises 49 ORFs. Comparative analysis of Tn1806 revealed the presence of a similar genetic element or part of it in related species such as Streptococcus pyogenes and also in the anaerobic species Finegoldia magna, Anaerococcus prevotii and Clostridium difficile. The genome of φSpn_200 is 35,989 bp in size and is organized in 47 ORFs grouped into five functional modules. Prophages similar to φSpn_200 were found in pneumococci and in other streptococcal species, showing a high degree of exchange of functional modules. φSpn_200 viral particles have morphologic characteristics typical of the Siphoviridae family and are capable of infecting a pneumococcal recipient strain. CONCLUSIONS: The sequence of S. pneumoniae AP200 chromosome revealed a dynamic genome, characterized by chromosomal rearrangements and horizontal gene transfers. The overall diversity of AP200 is driven mainly by the presence of the exogenous elements Tn1806 and φSpn_200 that show large gene exchanges with other genetic elements of different bacterial species. These genetic elements likely provide AP200 with additional genes, such as those conferring antibiotic-resistance, promoting its adaptation to the environment.


Subject(s)
Genome, Bacterial , Streptococcus pneumoniae/genetics , Chromosomes, Bacterial/genetics , DNA Transposable Elements , DNA, Bacterial/genetics , Molecular Sequence Annotation , Molecular Sequence Data , Prophages/genetics , Sequence Analysis, DNA , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/isolation & purification
9.
Antiviral Res ; 76(3): 252-62, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881064

ABSTRACT

Lactoferrin (Lf) is a multifunctional glycoprotein that plays an important role in immune regulation and defence mechanisms against bacteria, fungi and viruses. Bovine lactoferrin (bLf) has been recognized as a potent inhibitor of human herpetic viruses, such as cytomegalovirus and herpes simplex virus type 1 and 2. BLf has been found to prevent viral infection by binding to heparan sulphate containing proteoglycans that also act as cell receptors for herpetic viruses. In this study we further investigated the inhibiting activity of bLf against herpes simplex virus type 1 (HSV-1) in Green Monkey Kidney (GMK) cells and found that, in addition to the viral adsorption step, bLf also targets the HSV-1 entry process and cell-to-cell viral spread. Our study showed that the inhibition of HSV-1 infectivity by bLf is dependent on its interaction with specific structural viral proteins. Apart from the prevention of early phases of viral infection, cell-to-cell spread inhibition activity of HSV-1 by bLf confirmed that this protein is an outstanding candidate for the treatment of herpetic infections since it would offer the advantage to prevent also viral infections caused by cell-associated virus.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/growth & development , Lactoferrin/pharmacology , Virus Internalization/drug effects , Animals , Cattle , Cell Line , Chlorocebus aethiops , Lactoferrin/metabolism , Protein Binding , Viral Plaque Assay , Viral Proteins/metabolism , Virus Attachment/drug effects
10.
Antiviral Res ; 72(2): 145-52, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16774792

ABSTRACT

Lactoferrin, a member of the transferrin family, is a bi-globular iron binding glycoprotein, found in milk, exocrine secretions of mammals, and in secondary granules of polymorphonuclear neutrophiles that plays an important role in the defence against various pathogenic microorganisms. Previous studies in different virus-cell systems showed that lactoferrin is a potent inhibitor of different enveloped and naked virus infection. In this research we studied the effect of lactoferrin on BK polyomavirus, a human naked double-stranded DNA virus responsible for productive, persistent, and latent infections of the urinary tract. Results obtained demonstrate that lactoferrin treatment prevents early steps of BK virus infection in Vero cells, at the level of the adsorption phase, probably through the interaction with capsidic structures, although a lactoferrin-BK virus competition for cell plasma-membrane receptors cannot be ruled out.


Subject(s)
Antiviral Agents/pharmacology , BK Virus/drug effects , Lactoferrin/pharmacology , Animals , BK Virus/physiology , Cell Survival , Chlorocebus aethiops , DNA, Viral/analysis , Dose-Response Relationship, Drug , Neutralization Tests , Vero Cells , Virus Attachment/drug effects , Virus Replication/drug effects
11.
Biometals ; 17(3): 295-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15222481

ABSTRACT

It is well known that lactoferrin (Lf) is a potent inhibitor towards several enveloped and naked viruses, such as rotavirus, enterovirus and adenovirus. Lf is resistant to tryptic digestion and breast-fed infants excrete high levels of faecal Lf, so that its effect on viruses replicating in the gastrointestinal tract is of great interest. In this report, we analysed the mechanism of the antiviral action of this protein in three viral models which, despite representing different genoma and replication strategies, share the ability to infect the gut. Concerning the mechanism of action against rotavirus, Lf from bovine milk (BLf) possesses a dual role, preventing virus attachment to intestinal cells by binding to viral particles, and inhibiting a post adsorption step. The BLf effect towards poliovirus is due to the interference with an early infection step but, when the BLf molecule is saturated with Zn+2 ions, it is also capable of inhibiting viral replication after the viral adsorption phase. The anti-adenovirus action of BLf takes place on virus attachment to cell membranes through competition for common glycosaminoglycan receptors and a specific interaction with viral structural polypeptides. Taken together, these findings provide further evidence that Lf is an excellent candidate in the search of natural agents against viral enteric diseases, as it mainly acts by hindering adsorption and internalisation into cells through specific binding to cell receptors and/or viral particles.


Subject(s)
Antiviral Agents/metabolism , Lactoferrin/metabolism , Adenoviridae/metabolism , Animals , Enterovirus/metabolism , Humans , Rotavirus/metabolism
12.
Virology ; 318(1): 405-13, 2004 Jan 05.
Article in English | MEDLINE | ID: mdl-14972565

ABSTRACT

Previous reports have indicated that lactoferrin inhibits herpes simplex virus (HSV) infection during the very early phases of the viral replicative cycle. In the present work we investigated the mechanism of the antiviral activity of lactoferrin in mutant glycosaminoglycan (GAG)-deficient cells. Bovine lactoferrin (BLf) was a strong inhibitor of HSV-1 infection in cells expressing either heparan sulfate (HS) or chondroitin sulfate (CS) or both, but was ineffective or less efficient in GAG-deficient cells or in cells treated with GAG-degrading enzymes. In contrast to wild-type HSV-1, virus mutants devoid of glycoprotein C (gC) were significantly less inhibited by lactoferrin in GAG-expressing cells, indicating that lactoferrin interfered with the binding of viral gC to cell surface HS and/or CS. Finally, we demonstrated that lactoferrin bound directly to both HS and CS isolated from surfaces of the studied cells, as well as to commercial preparations of GAG chains. The results support the hypothesis that the inhibition of HSV-1 infectivity by lactoferrin is dependent on its interaction with cell surface GAG chains of HS and CS.


Subject(s)
Antiviral Agents/pharmacology , Fibroblasts/virology , Glycosaminoglycans/metabolism , Herpesvirus 1, Human/pathogenicity , Lactoferrin/pharmacology , Animals , Antiviral Agents/metabolism , Cattle , Cells, Cultured , Chondroitin Sulfates/metabolism , Fibroblasts/drug effects , Glycosaminoglycans/chemistry , Heparitin Sulfate/metabolism , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/metabolism , Humans , L Cells , Lactoferrin/metabolism , Mice , Viral Envelope Proteins/metabolism
13.
Antimicrob Agents Chemother ; 47(8): 2688-91, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12878543

ABSTRACT

We recently demonstrated that lactoferrin, an antimicrobial glycoprotein, can inhibit adenovirus infection by competing for common glycosaminoglycan receptors. This study further characterizes the antiadenovirus activity of the protein, thus demonstrating that lactoferrin neutralizes infection by binding to adenovirus particles and that its targets are viral III and IIIa structural polypeptides.


Subject(s)
Adenoviridae/drug effects , Adenovirus Infections, Human/drug therapy , Lactoferrin/therapeutic use , Peptides/drug effects , Adenoviridae/chemistry , Adenoviridae/ultrastructure , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Biotin/chemistry , Cattle , Electrophoresis, Polyacrylamide Gel , Enterovirus/chemistry , Enterovirus/drug effects , Enterovirus/ultrastructure , Lactoferrin/pharmacology , Microscopy, Electron , Peptides/chemistry , Tumor Cells, Cultured
14.
Antiviral Res ; 53(2): 153-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11750941

ABSTRACT

Different milk proteins were analysed for their inhibitory effect on adenovirus infection in vitro. Proteins investigated were mucin, alpha-lactalbumin, beta-lactoglobulin, bovine lactoferrin, and human lactoferrin. Results obtained demonstrated that mucin, alpha-lactalbumin, and beta-lactoglobulin did not prevent the viral cytopathic effect, whereas lactoferrin was able to inhibit adenovirus replication in a dose-dependent manner. Further experiments were carried out in which lactoferrin was added to the cells during different phases of viral infection. Results obtained showed that lactoferrin was able to prevent viral replication when added both before, or during the viral adsorption step, or when present during the entire replicative cycle of adenovirus, demonstrating that its action takes place on an early phase of viral replication.


Subject(s)
Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Lactoferrin/pharmacology , Milk Proteins/pharmacology , Adenovirus Infections, Human/virology , Adenoviruses, Human/pathogenicity , Adenoviruses, Human/physiology , Animals , Cattle , Cytopathogenic Effect, Viral/drug effects , Dose-Response Relationship, Drug , Humans , Tumor Cells, Cultured , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...