Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Pharmacol ; 180(11): 1460-1481, 2023 06.
Article in English | MEDLINE | ID: mdl-36526272

ABSTRACT

BACKGROUND AND PURPOSE: Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH: The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS: Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS: These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.


Subject(s)
COVID-19 , Sepsis , Humans , Mice , Animals , Oseltamivir/adverse effects , Zanamivir/adverse effects , Neuraminidase/metabolism , Neuraminidase/pharmacology , Neutrophils , Matrix Metalloproteinase 9/metabolism , Reactive Oxygen Species , Lipopolysaccharides/pharmacology , Sepsis/chemically induced
2.
Mar Drugs ; 20(11)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36354999

ABSTRACT

Acute and chronic dermatological injuries need rapid tissue repair due to the susceptibility to infections. To effectively promote cutaneous wound recovery, it is essential to develop safe, low-cost, and affordable regenerative tools. Therefore, we aimed to identify the biological mechanisms involved in the wound healing properties of the glycosaminoglycan dermatan sulfate (DS), obtained from ascidian Styela plicata, a marine invertebrate, which in preliminary work from our group showed no toxicity and promoted a remarkable fibroblast proliferation and migration. In this study, 2,4-DS (50 µg/mL)-treated and control groups had the relative gene expression of 84 genes participating in the healing pathway evaluated. The results showed that 57% of the genes were overexpressed during treatment, 16% were underexpressed, and 9.52% were not detected. In silico analysis of metabolic interactions exhibited overexpression of genes related to: extracellular matrix organization, hemostasis, secretion of inflammatory mediators, and regulation of insulin-like growth factor transport and uptake. Furthermore, in C57BL/6 mice subjected to experimental wounds treated with 0.25% 2,4-DS, the histological parameters demonstrated a great capacity for vascular recovery. Additionally, this study confirmed that DS is a potent inducer of wound-healing cellular pathways and a promoter of neovascularization, being a natural ally in the tissue regeneration strategy.


Subject(s)
Dermatan Sulfate , Urochordata , Animals , Mice , Dermatan Sulfate/metabolism , Dermatan Sulfate/pharmacology , Mice, Inbred C57BL , Urochordata/metabolism , Wound Healing , Natural Resources
3.
bioRxiv ; 2022 Oct 14.
Article in English | MEDLINE | ID: mdl-33200130

ABSTRACT

Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Neuraminidase (NEU)-mediated cleavage of surface sialic acid has been demonstrated to regulate leukocyte responses. Here, we report that antiviral NEU inhibitors constrain host NEU activity, surface sialic acid release, ROS production, and NETs released by microbial-activated human neutrophils. In vivo, treatment with Oseltamivir results in infection control and host survival in peritonitis and pneumonia models of sepsis. Single-cell RNA sequencing re-analysis of publicly data sets of respiratory tract samples from critical COVID-19 patients revealed an overexpression of NEU1 in infiltrated neutrophils. Moreover, Oseltamivir or Zanamivir treatment of whole blood cells from severe COVID-19 patients reduces host NEU-mediated shedding of cell surface sialic acid and neutrophil overactivation. These findings suggest that neuraminidase inhibitors can serve as host-directed interventions to dampen neutrophil dysfunction in severe infections.

4.
Biophys Chem ; 231: 116-124, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28410940

ABSTRACT

BACKGROUND: Avian influenza A viruses can cross naturally into mammals and cause severe diseases, as observed for H5N1. The high lethality of human infections causes major concerns about the real risk of a possible pandemic of severe diseases to which human susceptibility may be high and universal. High hydrostatic pressure (HHP) is a valuable tool for studies regarding the folding of proteins and the assembly of macromolecular structures such as viruses; furthermore, HHP has already been demonstrated to promote viral inactivation. METHODS: Here, we investigated the structural stability of avian and human influenza viruses using spectroscopic and light-scattering techniques. We found that both particles have similar structural stabilities and that HHP promotes structural changes. RESULTS: HHP induced slight structural changes to both human and avian influenza viruses, and these changes were largely reversible when the pressure returned to its initial level. The spectroscopic data showed that H3N2 was more pressure-sensitive than H3N8. Structural changes did not predict changes in protein function, as H3N2 fusion activity was not affected, while H3N8 fusion activity drastically decreased. The fusion activity of H1N1 was also strongly affected by HHP. In all cases, HHP caused inactivation of the different influenza viruses. CONCLUSIONS: HHP may be a useful tool for vaccine development, as it induces minor and reversible structural changes that may be associated with partial preservation of viral biological activities and may potentiate their immunogenic response while abolishing their infectivity. We also confirmed that, although pressure does not promote drastic changes in viral particle structure, it can distinctly affect viral fusion activity.


Subject(s)
Influenza A virus/chemistry , Animals , Guanidine/chemistry , Humans , Hydrostatic Pressure , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H3N8 Subtype/chemistry , Influenza A Virus, H3N8 Subtype/physiology , Influenza A virus/physiology , Temperature , Urea/chemistry , Vaccines/immunology , Virus Inactivation
5.
Subcell Biochem ; 72: 301-18, 2015.
Article in English | MEDLINE | ID: mdl-26174388

ABSTRACT

In recent years, many applications in diverse scientific fields with various purposes have examined pressure as a thermodynamic parameter. Pressure studies on viruses have direct biotechnological applications. Currently, most studies that involve viral inactivation by HHP are found in the area of food engineering and focus on the inactivation of foodborne viruses. Nevertheless, studies of viral inactivation for other purposes have also been conducted. HHP has been shown to be efficient in the inactivation of many viruses of clinical importance and the use of HHP approach has been proposed for the development of animal and human vaccines. Several studies have demonstrated that pressure can result in virus inactivation while preserving immunogenic properties. Viruses contain several components that can be susceptible to the effects of pressure. HHP has been a valuable tool for assessing viral structure function relationships because the viral structure is highly dependent on protein-protein interactions. In the case of small icosahedral viruses, incremental increases in pressure produce a progressive decrease in the folding structure when moving from assembled capsids to ribonucleoprotein intermediates (in RNA viruses), free dissociated units (dimers and/or monomers) and denatured monomers. High pressure inactivates enveloped viruses by trapping their particles in a fusion-like intermediate state. The fusogenic state, which is characterized by a smaller viral volume, is the final conformation promoted by HHP, in contrast with the metastable native state, which is characterized by a larger volume. The combined effects of high pressure with other factors, such as low or subzero temperature, pH and agents in sub-denaturing conditions (urea), have been a formidable tool in the assessment of the component's structure, as well as pathogen inactivation. HHP is a technology for the production of inactivated vaccines that are free of chemicals, safe and capable of inducing strong humoral and cellular immune responses. Here we present a current overview about the pressure-induced viral inactivation and the production of inactivated viral vaccines.


Subject(s)
Hydrostatic Pressure , Viral Vaccines/biosynthesis , Virus Inactivation , Temperature , Viruses/immunology
6.
PLoS One ; 10(6): e0128785, 2015.
Article in English | MEDLINE | ID: mdl-26056825

ABSTRACT

Influenza viruses pose a serious global health threat, particularly in light of newly emerging strains, such as the avian influenza H5N1 and H7N9 viruses. Vaccination remains the primary method for preventing acquiring influenza or for avoiding developing serious complications related to the disease. Vaccinations based on inactivated split virus vaccines or on chemically inactivated whole virus have some important drawbacks, including changes in the immunogenic properties of the virus. To induce a greater mucosal immune response, intranasally administered vaccines are highly desired as they not only prevent disease but can also block the infection at its primary site. To avoid these drawbacks, hydrostatic pressure has been used as a potential method for viral inactivation and vaccine production. In this study, we show that hydrostatic pressure inactivates the avian influenza A H3N8 virus, while still maintaining hemagglutinin and neuraminidase functionalities. Challenged vaccinated animals showed no disease signs (ruffled fur, lethargy, weight loss, and huddling). Similarly, these animals showed less Evans Blue dye leakage and lower cell counts in their bronchoalveolar lavage fluid compared with the challenged non-vaccinated group. We found that the whole inactivated particles were capable of generating a neutralizing antibody response in serum, and IgA was also found in nasal mucosa and feces. After the vaccination and challenge we observed Th1/Th2 cytokine secretion with a prevalence of IFN-γ. Our data indicate that the animals present a satisfactory immune response after vaccination and are protected against infection. Our results may pave the way for the development of a novel pressure-based vaccine against influenza virus.


Subject(s)
Administration, Intranasal/methods , Orthomyxoviridae Infections/prevention & control , Vaccines, Inactivated/immunology , Administration, Intranasal/adverse effects , Animals , Cytokines/genetics , Cytokines/metabolism , Dogs , Female , Influenza A Virus, H3N8 Subtype/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Pressure , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects
7.
PLoS One ; 8(11): e80785, 2013.
Article in English | MEDLINE | ID: mdl-24282553

ABSTRACT

Whole inactivated vaccines (WIVs) possess greater immunogenicity than split or subunit vaccines, and recent studies have demonstrated that WIVs with preserved fusogenic activity are more protective than non-fusogenic WIVs. In this work, we describe the inactivation of human influenza virus X-31 by high hydrostatic pressure (HHP) and analyze the effects on the structure by spectroscopic measurements, light scattering, and electron microscopy. We also investigated the effects of HHP on the glycoprotein activity and fusogenic activity of the viral particles. The electron microscopy data showed pore formation on the viral envelope, but the general morphology was preserved, and small variations were seen in the particle structure. The activity of hemagglutinin (HA) during the process of binding and fusion was affected in a time-dependent manner, but neuraminidase (NA) activity was not affected. Infectious activity ceased after 3 hours of pressurization, and mice were protected from infection after being vaccinated. Our results revealed full viral inactivation with overall preservation of viral structure and maintenance of fusogenic activity, thereby conferring protection against infection. A strong response consisting of serum immunoglobulin IgG1, IgG2a, and serum and mucosal IgA was also detected after vaccination. Thus, our data strongly suggest that applying hydrostatic pressure may be an effective method for developing new vaccines against influenza A as well as other viruses.


Subject(s)
Hydrostatic Pressure , Influenza, Human/virology , Membrane Fusion , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae/physiology , Animals , Antibodies, Viral/biosynthesis , Humans , Mice , Microscopy, Electron , Orthomyxoviridae/immunology , Orthomyxoviridae/ultrastructure , Orthomyxoviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...