Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Front Immunol ; 12: 786617, 2021.
Article in English | MEDLINE | ID: mdl-34868073

ABSTRACT

Neuraminidase of influenza A and B viruses plays a critical role in the virus life cycle and is an important target of the host immune system. Here, we highlight the current understanding of influenza neuraminidase structure, function, antigenicity, immunogenicity, and immune protective potential. Neuraminidase inhibiting antibodies have been recognized as correlates of protection against disease caused by natural or experimental influenza A virus infection in humans. In the past years, we have witnessed an increasing interest in the use of influenza neuraminidase to improve the protective potential of currently used influenza vaccines. A number of well-characterized influenza neuraminidase-specific monoclonal antibodies have been described recently, most of which can protect in experimental challenge models by inhibiting the neuraminidase activity or by Fc receptor-dependent mechanisms. The relative instability of the neuraminidase poses a challenge for protein-based antigen design. We critically review the different solutions that have been proposed to solve this problem, ranging from the inclusion of stabilizing heterologous tetramerizing zippers to the introduction of inter-protomer stabilizing mutations. Computationally engineered neuraminidase antigens have been generated that offer broad, within subtype protection in animal challenge models. We also provide an overview of modern vaccine technology platforms that are compatible with the induction of robust neuraminidase-specific immune responses. In the near future, we will likely see the implementation of influenza vaccines that confront the influenza virus with a double punch: targeting both the hemagglutinin and the neuraminidase.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Viral Proteins/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigenic Drift and Shift , Antigens, Viral/immunology , Antigens, Viral/ultrastructure , Catalytic Domain/genetics , Catalytic Domain/immunology , Cross Protection , Evolution, Molecular , Humans , Immunogenicity, Vaccine , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Alphainfluenzavirus/enzymology , Alphainfluenzavirus/genetics , Alphainfluenzavirus/immunology , Betainfluenzavirus/enzymology , Betainfluenzavirus/genetics , Betainfluenzavirus/immunology , Mutation , Nanoparticles , Neuraminidase/administration & dosage , Neuraminidase/genetics , Neuraminidase/ultrastructure , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/ultrastructure , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/ultrastructure
2.
mBio ; 12(6): e0224121, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34809451

ABSTRACT

Current seasonal influenza virus vaccines do not induce robust immune responses to neuraminidase. Several factors, including immunodominance of hemagglutinin over neuraminidase, instability of neuraminidase in vaccine formulations, and variable, nonstandardized amounts of neuraminidase in the vaccines, may contribute to this effect. However, vaccines that induce strong antineuraminidase immune responses would be beneficial, as they are highly protective. Furthermore, antigenic drift is slower for neuraminidase than for hemagglutinin, potentially providing broader coverage. Here, we designed stabilized recombinant versions of neuraminidase by replacing the N-terminal cytoplasmic domain, transmembrane, and extracellular stalk with tetramerization domains from the measles or Sendai virus phosphoprotein or from an Arabidopsis thaliana transcription factor. The measles virus tetramerization domain-based construct, termed N1-MPP, was chosen for further evaluation, as it retained antigenicity, neuraminidase activity, and structural integrity and provided robust protection in vivo against lethal virus challenge in the mouse model. We tested N1-MPP as a standalone vaccine, admixed with seasonal influenza virus vaccines, or given with seasonal influenza virus vaccines but in the other leg of the mouse. Admixture with different formulations of seasonal vaccines led to a weak neuraminidase response, suggesting a dominant effect of hemagglutinin over neuraminidase when administered in the same formulation. However, administration of neuraminidase alone or with seasonal vaccine administered in the alternate leg of the mouse induced robust antibody responses. Thus, this recombinant neuraminidase construct is a promising vaccine antigen that may enhance and broaden protection against seasonal influenza viruses. IMPORTANCE Influenza virus infections remain a high risk to human health, causing up to 650,000 deaths worldwide every year, with an enormous burden on the health care system. Since currently available seasonal vaccines are only partially effective and often mismatched to the circulating strains, a broader protective influenza virus vaccine is needed. Here, we generated a recombinant influenza virus vaccine candidate based on the more conserved neuraminidase surface glycoprotein in order to induce a robust and broader protective immune response against a variety of circulating influenza virus strains.


Subject(s)
Influenza Vaccines/immunology , Influenza, Human/prevention & control , Measles virus/immunology , Neuraminidase/immunology , Phosphoproteins/immunology , Viral Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Antigenic Drift and Shift , Cross Reactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/chemistry , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Measles virus/chemistry , Measles virus/genetics , Mice , Mice, Inbred BALB C , Neuraminidase/administration & dosage , Neuraminidase/chemistry , Neuraminidase/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Domains , Sequence Alignment , Vaccination , Viral Proteins/administration & dosage , Viral Proteins/chemistry , Viral Proteins/genetics
3.
ACS Appl Bio Mater ; 4(6): 4953-4961, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34179728

ABSTRACT

We generated self-adjuvanted protein nanoparticles of conserved influenza antigens and immunized mice via skin vaccination with dissolvable microneedle patches (MNPs) to increase the strength and breadth of immune responses. We produced M2e nanoparticles via ethanol desolvation, and double-layered NA1/M2e (shell/core), NA1-FliC/M2e, NA2/M2e, and NA2-FliC/M2e protein nanoparticles by chemically crosslinking influenza NA and flagellin (FliC) onto the surfaces of the M2e nanoparticles. The resulting nanoparticles retained FliC TLR5 innate signaling activity and significantly increased antigen-uptake and dendritic cell maturation in vitro. We incorporated the nanoparticles into MNPs for skin vaccination in mice. The nanoparticle MNPs significantly increased M2e and NA-specific antibody levels, the numbers of germinal center B cells, and IL-4 positive splenocytes. Double-layered nanoparticle MNP skin vaccination protected mice against homologous and heterosubtypic influenza viruses. Our results demonstrated that MNP skin vaccination of NA-FliC/M2e nanoparticles could be developed into a standalone or synergistic component of a universal influenza vaccine strategy.


Subject(s)
Drug Delivery Systems , Flagellin/administration & dosage , Influenza Vaccines/administration & dosage , Nanoparticles/administration & dosage , Neuraminidase/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Viral Matrix Proteins/administration & dosage , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cytokines/immunology , Dendritic Cells/immunology , Flagellin/chemistry , Immunoglobulin G/blood , Influenza Vaccines/chemistry , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Microinjections , Nanoparticles/chemistry , Needles , Neuraminidase/chemistry , Neuraminidase/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology
4.
J Vet Sci ; 21(1): e12, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31940691

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus that causes diarrhea in piglets. However, the biological characteristics of PDCoV are unclear. In this study, the hemagglutination (HA) abilities of two PDCoV strains (CH-01 and HNZK-04) were investigated. Our results showed that PDCoV has the ability to agglutinate rabbit erythrocytes after virion pretreatment with trypsin or neuraminidase. Additionally, the HA assay results showed a significant positive correlation with the infectious viral titer. Our results suggest that assessing the HA activity of PDCoV may be a useful diagnostic method for investigating and surveilling PDCoV infections.


Subject(s)
Coronavirus Infections/veterinary , Coronavirus/physiology , Hemagglutination , Swine Diseases/immunology , Animals , Coronavirus Infections/immunology , Coronavirus Infections/virology , Diarrhea/immunology , Diarrhea/veterinary , Diarrhea/virology , Erythrocytes/immunology , Neuraminidase/administration & dosage , Rabbits , Swine , Swine Diseases/virology , Trypsin/administration & dosage , Virion/drug effects
5.
J Neuroinflammation ; 16(1): 245, 2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31791382

ABSTRACT

BACKGROUND: Neuraminidase (NA) is a sialidase present, among various locations, in the envelope/membrane of some bacteria/viruses (e.g., influenza virus), and is involved in infectiveness and/or dispersion. The administration of NA within the brain lateral ventricle represents a model of acute sterile inflammation. The relevance of the Toll-like receptors TLR2 and TLR4 (particularly those in microglial cells) in such process was investigated. METHODS: Mouse strains deficient in either TLR2 (TLR2-/-) or TLR4 (TLR4-/-) were used. NA was injected in the lateral ventricle, and the inflammatory reaction was studied by immunohistochemistry (IBA1 and IL-1ß) and qPCR (cytokine response). Also, microglia was isolated from those strains and in vitro stimulated with NA, or with TLR2/TLR4 agonists as positive controls (P3C and LPS respectively). The relevance of the sialidase activity of NA was investigated by stimulating microglia with heat-inactivated NA, or with native NA in the presence of sialidase inhibitors (oseltamivir phosphate and N-acetyl-2,3-dehydro-2-deoxyneuraminic acid). RESULTS: In septofimbria and hypothalamus, IBA1-positive and IL-1ß-positive cell counts increased after NA injection in wild type (WT) mice. In TLR4-/- mice, such increases were largely abolished, while were only slightly diminished in TLR2-/- mice. Similarly, the NA-induced expression of IL-1ß, TNFα, and IL-6 was completely blocked in TLR4-/- mice, and only partially reduced in TLR2-/- mice. In isolated cultured microglia, NA induced a cytokine response (IL-1ß, TNFα, and IL-6) in WT microglia, but was unable to do so in TLR4-/- microglia; TLR2 deficiency partially affected the NA-induced microglial response. When WT microglia was exposed in vitro to heat-inactivated NA or to native NA along with sialidase inhibitors, the NA-induced microglia activation was almost completely abrogated. CONCLUSIONS: NA is able to directly activate microglial cells, and it does so mostly acting through the TLR4 receptor, while TLR2 has a secondary role. Accordingly, the inflammatory reaction induced by NA in vivo is partially dependent on TLR2, while TLR4 plays a crucial role. Also, the sialidase activity of NA is critical for microglial activation. These results highlight the relevance of microbial NA in the neuroinflammation provoked by NA-bearing pathogens and the possibility of targeting its sialidase activity to ameliorate its impact.


Subject(s)
Microglia/metabolism , Neuraminidase/administration & dosage , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Neuraminidase/antagonists & inhibitors
6.
J Pharmacol Exp Ther ; 368(1): 11-20, 2019 01.
Article in English | MEDLINE | ID: mdl-30348750

ABSTRACT

Most patients acutely infected with Trypanosoma cruzi undergo short-term structural and functional cardiac alterations that heal without sequelae. By contrast, in patients whose disease progresses to chronic infection, irreversible degenerative chronic Chagas cardiomyopathy (CCC) may develop. To account for the contrast between cardiac regeneration in high-parasitism acute infection and progressive cardiomyopathy in low-parasitism CCC, we hypothesized that T. cruzi expresses repair factors that directly facilitate cardiac regeneration. We investigated, as one such repair factor, the T. cruzi parasite-derived neurotrophic factor (PDNF), known to trigger survival of cardiac myocytes and fibroblasts and upregulate chemokine chemokine C-C motif ligand 2, which promotes migration of regenerative cardiac progenitor cells (CPCs). Using in vivo and in vitro models of Chagas disease, we tested whether T. cruzi PDNF promotes cardiac repair. Quantitative PCR and flow cytometry of heart tissue revealed that stem-cell antigen-1 (Sca-1+) CPCs expand in acute infection in parallel to parasitism. Recombinant PDNF induced survival and expansion of ex vivo CPCs, and intravenous administration of PDNF into naïve mice upregulated mRNA of cardiac stem-cell marker Sca-1. Furthermore, in CCC mice, a 3-week intravenous administration of PDNF protocol induced CPC expansion and reversed left ventricular T-cell accumulation and cardiac remodeling including fibrosis. Compared with CCC vehicle-treated mice, which developed severe atrioventricular block, PDNF-treated mice exhibited reduced frequency and severity of conduction abnormalities. Our findings are in support of the novel concept that T. cruzi uses PDNF to promote mutually beneficial cardiac repair in Chagas disease. This could indicate a possible path to prevention or treatment of CCC.


Subject(s)
Atrioventricular Block/blood , Atrioventricular Block/therapy , Chagas Disease/blood , Chagas Disease/therapy , Glycoproteins/administration & dosage , Glycoproteins/blood , Neuraminidase/administration & dosage , Neuraminidase/blood , Administration, Intravenous , Animals , Atrioventricular Block/physiopathology , Chagas Disease/physiopathology , Chlorocebus aethiops , Chronic Disease , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Trypanosoma cruzi/metabolism , Vero Cells
7.
J Gene Med ; 20(9): e3049, 2018 09.
Article in English | MEDLINE | ID: mdl-30101537

ABSTRACT

BACKGROUND: Following systemic delivery, AAV9-mediated gene expression is significantly increased in ischemic versus non-ischemic muscle, suggesting that AAV9 is an attractive vector for treating peripheral arterial disease. Potential mechanisms underlying ischemia-augmented expression include: (i) increased vascular permeability and (ii) "unmasking" of endogenous AAV9 receptors. In the present study, we aimed to reconstitute the ischemic induction of AAV9 in vivo, using local injection of histamine (to increase vascular permeability) and neuraminidase (to desialylate cell surface glycans). METHODS: Bioassays were performed to optimize the effects of histamine and neuraminidase after intramuscular injection. Histamine and/or neuraminidase were then injected intramuscularly shortly before intravenous injection of an AAV9 vector expressing luciferase. Luciferase expression was serially assessed with bioluminescence imaging. At the end of the study, tissues were harvested for assays of luciferase activity and AAV9 genome copy number aiming to assess AAV-mediated gene expression and transduction, respectively. RESULTS: Intramuscular injection of either neuraminidase or neuraminidase plus histamine significantly increased both transduction and gene expression, whereas histamine alone had little effect. Pre-injection with neuraminidase increased AAV9-mediated gene delivery by four- to nine-fold and luciferase activity by 60-100-fold. Luciferase activity in neuraminidase-injected muscle was > 100-fold higher than in any off-target tissue (including heart, liver and brain). CONCLUSIONS: The ischemic induction of AAV9-mediated gene expression in muscle can largely be reconstituted by pre-injecting neuraminidase intranmuscularly. This strategy may prove useful in future human gene therapy protocols as a quick and efficient means to selectively target systemically injected AAV9 to localized regions of muscle, thus decreasing the potential for adverse effects in off-target tissues.


Subject(s)
Dependovirus/genetics , Gene Expression/genetics , Muscle, Skeletal/metabolism , Neuraminidase/metabolism , Transduction, Genetic/methods , Animals , Genetic Therapy/methods , Genetic Vectors/genetics , HEK293 Cells , Histamine/administration & dosage , Histamine/metabolism , Humans , Injections, Intramuscular , Ischemia , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements/methods , Mice, Inbred BALB C , N-Acetylneuraminic Acid/metabolism , Neuraminidase/administration & dosage , Neuraminidase/genetics , Reproducibility of Results
8.
Clin Respir J ; 12(11): 2539-2545, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30126042

ABSTRACT

OBJECTIVES: The prevention strategies for mother-to-fetus/infant transmission of H7N9 virus have not been well understood, and the study on this subject will provide further insights. METHODS: Reverse transcriptase polymerase chain reaction assay was undertaken to detect H7N9 virus in samples from a pregnant women, a postpartum woman, and their fetus/infant. Pathological features of tissues from the dead fetus were evaluated with hematoxylin and eosin staining. Hemagglutination inhibition assay was used to detect virus-specific antibodies. Furthermore, relevant literatures were reviewed and analyzed. RESULTS: A 28-year-old pregnant woman was hospitalized for H7N9 infection and prescribed with oseltamivir and peramivir for 2 days before admission. The fetal heart beating stopped on day 4, the dead fetus was delivered on day 13, and the woman expired on day 26. All fetal tissues were H7N9 virus-negative. A 28-year-old woman delivered a newborn on December 20, 2016. Five days later, she developed influenza-like symptoms and was confirmed with H7N9 infection. She had close contact with her infant for 9 days. Oseltamivir and peramivir were prescribed within 2 days after illness onset. A throat swab and a pair of serum samples from the infant were all negative for H7N9 virus during 4-week follow-up. In total, ten studies referring to transplacental transmission and four reports on maternal infection of H7N9 virus were reviewed and analyzed. CONCLUSION: No evidence showed H7N9 virus infection in both fetus and infant. The early administration of neuraminidase inhibitor seemed beneficial in preventing mother-to-fetus/infant transmission of H7N9 virus.


Subject(s)
Influenza A Virus, H7N9 Subtype/genetics , Influenza, Human/prevention & control , Influenza, Human/transmission , Neuraminidase/antagonists & inhibitors , Acids, Carbocyclic , Adult , Cyclopentanes/administration & dosage , Cyclopentanes/therapeutic use , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Fatal Outcome , Female , Fetal Death , Fetus , Guanidines/administration & dosage , Guanidines/therapeutic use , Humans , Infant , Influenza A Virus, H7N9 Subtype/isolation & purification , Influenza, Human/drug therapy , Influenza, Human/virology , Maternal-Fetal Exchange , Mothers , Neuraminidase/administration & dosage , Neuraminidase/therapeutic use , Oseltamivir/administration & dosage , Oseltamivir/therapeutic use , Pregnancy , Secondary Prevention/methods , Secondary Prevention/standards , Treatment Outcome
9.
Influenza Other Respir Viruses ; 11(6): 518-524, 2017 11.
Article in English | MEDLINE | ID: mdl-28752641

ABSTRACT

BACKGROUND: Avian influenza viruses represent a growing threat of an influenza pandemic. The co-circulation of multiple H9N2 genotypes over the past decade has been replaced by one predominant genotype-G57 genotype, which displays a changed antigenicity and improved adaptability in chickens. Effective H9N2 subtype avian influenza virus vaccines for poultry are urgently needed. OBJECTIVE: In this study, we constructed H9N2 subtype avian influenza virus-like particle (VLP) and evaluated its protective efficacy in specific pathogen-free (SPF) chickens to lay the foundation for developing an effective vaccine against influenza viruses. METHODS: Expression of influenza proteins in VLPs was confirmed by Western blot, hemagglutination inhibition (HI), and neuraminidase inhibition (NI). The morphology was observed by electron microscopy. A group of 15 three-week-old SPF chickens was divided into three subgroups of five chickens immunized with VLP, commercial vaccine, and PBS. Challenge study was performed to evaluate efficacy of VLP vaccine. RESULTS AND CONCLUSIONS: The hemagglutinin (HA) and neuraminidase (NA) proteins were co-expressed in the infected cells, self-assembled, and were released into the culture medium in the form of VLPs of diameter ~80 nm. The VLPs exhibited some functional characteristics of a full influenza virus, including hemagglutination and neuraminidase activity. In SPF chickens, the VLPs elicited serum antibodies specific for H9N2 and induced a higher HI titer (as detected by a homologous antigen) than did a commercial H9N2 vaccine (A/chicken/Shanghai/F/1998). Viral shedding from VLP vaccine subgroup was reduced compared with commercial vaccine subgroup and control subgroup.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H9N2 Subtype/chemistry , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Neuraminidase/chemistry , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Viral/blood , Chickens , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza in Birds/immunology , Neuraminidase/administration & dosage , Neuraminidase/genetics , Poultry Diseases/prevention & control , Poultry Diseases/virology , Specific Pathogen-Free Organisms , Vaccination/veterinary , Vaccines, Inactivated/immunology , Vaccines, Virus-Like Particle/administration & dosage , Virus Shedding
10.
Biomed Res Int ; 2017: 7212985, 2017.
Article in English | MEDLINE | ID: mdl-28337456

ABSTRACT

Previous studies showed the presence of Mycoplasma pneumoniae (M. pneumoniae) and membrane-shed microparticles (MPs) in vulnerable atherosclerotic plaques. H&S Science and Biotechnology developed PTCTS, composed by natural particles from medicinal plants (PTC) combined with trans-Sialidase (TS), to combat MPs and Mycoplasma pneumoniae. Our aim was to determine the effects of the different components of PTCTS in a rabbit model of atherosclerosis. Rabbits were fed with high cholesterol diet for 12 weeks and treated during the last 6 weeks with either vehicle, PTC, TS, or PTCTS. Lipid profile and quantification of MPs positive for Mycoplasma pneumoniae and oxidized LDL antigens were carried out. Aortas and organs were then histologically analyzed. PTCTS reduced circulating MPs positive for Mycoplasma pneumoniae and oxidized LDL antigens, reduced the plaque area in the abdominal aorta, and caused positive remodeling of the ascendant aorta. PTC caused positive remodeling and reduced plaque area in the abdominal aorta; however, TS had a lipid lowering effect. PTCTS components combined were more effective against atherosclerosis than individual components. Our data reinforce the infectious theory of atherosclerosis and underscore the potential role of circulating MPs. Therefore, the removal of Mycoplasma-derived MPs could be a new therapeutic approach in the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Glycoproteins/administration & dosage , Mycoplasma pneumoniae/drug effects , Neuraminidase/administration & dosage , Plaque, Atherosclerotic/drug therapy , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Atherosclerosis/metabolism , Atherosclerosis/microbiology , Atherosclerosis/pathology , Biological Products/administration & dosage , Biological Products/chemistry , Cholesterol, Dietary/pharmacology , Diet, High-Fat/adverse effects , Disease Models, Animal , Glycoproteins/chemistry , Humans , Lipoproteins, LDL/metabolism , Male , Mycoplasma pneumoniae/pathogenicity , Neuraminidase/chemistry , Plants, Medicinal/chemistry , Plaque, Atherosclerotic/microbiology , Plaque, Atherosclerotic/pathology , Rabbits
11.
J Pharmacol Sci ; 133(1): 49-52, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28089228

ABSTRACT

Sialic acids are highly charged glycoresidues that are attached to glycoproteins or glycosphingolipids, and they are associated with various biological functions. Gangliosides, sialic acid-containing glycosphingolipids, are abundant in neural tissues and play important roles in the nervous system. Previous studies revealed that peripheral gangliosides are involved in nociceptive behavior and hyperalgesia. These observations prompted us to determine whether the sialic acid-cleaving enzyme sialidase affects pain signaling. Intraplantar injection of sialidase reduced mechanical allodynia during complete Freund's adjuvant-induced inflammation. We also found that ganglioside induces mechanical allodynia in naïve mice. These results suggest that sialyl conjugates in subcutaneous tissues modify allodynia.


Subject(s)
Hyperalgesia/complications , Hyperalgesia/drug therapy , Inflammation/complications , Neuraminidase/administration & dosage , Neuraminidase/pharmacology , Animals , Foot/pathology , Freund's Adjuvant , Gangliosides/pharmacology , Hyperalgesia/prevention & control , Inflammation/chemically induced , Inflammation/pathology , Injections , Male , Mice , Neuraminidase/therapeutic use , Pain Management
12.
Virology ; 501: 176-182, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27936463

ABSTRACT

Avian influenza (AI) viruses circulating in wild birds pose a serious threat to public health. Human and veterinary vaccines against AI subtypes are needed. Here we prepared triple-subtype VLPs that co-localized H5, H7 and H9 antigens derived from H5N1, H7N3 and H9N2 viruses. VLPs also contained influenza N1 neuraminidase and retroviral gag protein. The H5/H7/H9/N1/gag VLPs were prepared using baculovirus expression. Biochemical, functional and antigenic characteristics were determined including hemagglutination and neuraminidase enzyme activities. VLPs were further evaluated in a chicken AI challenge model for safety, immunogenicity and protective efficacy against heterologous AI viruses including H5N2, H7N3 and H9N2 subtypes. All vaccinated birds survived challenges with H5N2 and H7N3 highly pathogenic AI (HPAI) viruses, while all controls died. Immune response was also detectable after challenge with low pathogenicity AI (LPAI) H9N2 virus suggesting that H5/H7/H9/N1/gag VLPs represent a promising approach for the development of broadly protective AI vaccine.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N2 Subtype/immunology , Influenza A Virus, H7N3 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Neuraminidase/immunology , Animals , Antibodies, Viral/immunology , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunity , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H7N3 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza in Birds/immunology , Influenza in Birds/virology , Neuraminidase/administration & dosage , Neuraminidase/genetics , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology
13.
Proc Natl Acad Sci U S A ; 113(37): 10304-9, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27551071

ABSTRACT

Cell surface sialosides constitute a central axis of immune modulation that is exploited by tumors to evade both innate and adaptive immune destruction. Therapeutic strategies that target tumor-associated sialosides may therefore potentiate antitumor immunity. Here, we report the development of antibody-sialidase conjugates that enhance tumor cell susceptibility to antibody-dependent cell-mediated cytotoxicity (ADCC) by selective desialylation of the tumor cell glycocalyx. We chemically fused a recombinant sialidase to the human epidermal growth factor receptor 2 (HER2)-specific antibody trastuzumab through a C-terminal aldehyde tag. The antibody-sialidase conjugate desialylated tumor cells in a HER2-dependent manner, reduced binding by natural killer (NK) cell inhibitory sialic acid-binding Ig-like lectin (Siglec) receptors, and enhanced binding to the NK-activating receptor natural killer group 2D (NKG2D). Sialidase conjugation to trastuzumab enhanced ADCC against tumor cells expressing moderate levels of HER2, suggesting a therapeutic strategy for cancer patients with lower HER2 levels or inherent trastuzumab resistance. Precision glycocalyx editing with antibody-enzyme conjugates is therefore a promising avenue for cancer immune therapy.


Subject(s)
Glycocalyx/genetics , Immunotherapy , Neoplasms/immunology , Receptor, ErbB-2/genetics , Sialic Acid Binding Ig-like Lectin 1/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm/immunology , Glycocalyx/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms/therapy , Neuraminidase/administration & dosage , Neuraminidase/chemistry , Receptor, ErbB-2/immunology , Sialic Acid Binding Ig-like Lectin 1/immunology , Trastuzumab/administration & dosage , Trastuzumab/adverse effects
14.
J Ethnopharmacol ; 193: 283-292, 2016 Dec 04.
Article in English | MEDLINE | ID: mdl-27553976

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: P. cuspidatum is a popular Chinese medicinal herb, having a long history of usage in traditional Chinese medicine for the treatment of several inflammatory diseases in the form of powders and decoctions. Similarly there are many reports that P. cuspidatum has antibacterial and anti-inflammatory effects, both of which are properties associated with compounds having activity against bacterial neuraminidase (BNA). AIM OF THE STUDY: We investigated whether P. cuspidatum's metabolites exhibited BNA inhibition. Consistent with our hypothesis, we found several inhibitors from the methanol extract of this plant, and then fully characterized their inhibitory mechanisms. MATERIALS AND METHODS: Activity guided separation of methanol extract led to isolation of individual constituents, and subsequently their structures were elucidated by spectroscopic analysis. Detailed kinetic behaviors of BNA inhibitors were explored by showing the changes of Km and Vmax, the ratios of KI/KIS and Kik/Kiv, and fluorescence quenching effect. RESULTS AND CONCLUSION: This study attempted to isolate the responsible metabolites and elucidate the BNA inhibitory mechanism. The principal BNA inhibitory compounds (2-6) were identified as emodin (2), physcion-8-O-ß-D-glucopyranoside (3), emodin-8-O-ß-D-glucopyranoside (4), emodin-1-O-ß-D-glucopyranoside (5), and 2-methoxy-6-acetyl-7-methyljuglone (6). Unexpectedly, anthraquinone glucosides (3-5) were much more potent than their corresponding aglycones (1 and 2). For example, emodin (2) had an IC50=5.4µM, whereas its glucosides (4 and 5) had IC50=0.85µM and 0.43µM respectively. A similar trend was observed with physcion (1, IC50>200µM) and its glucoside (3, IC50=6.2µM). The anthraquinone (2) was mixed type I inhibitor, whereas its glucosides (4 and 5) were noncompetitive. In addition, the fluorescence quenching study showed that the affinity constants (KSV) of inhibitors increased in proportion to their inhibitory potencies. Furthermore, we quantified the major and minor metabolites through UPLC-PDA-Q-TOF/MS, and revealed that the most potent inhibitors were the major constituents. This result contributes to our understanding of P. cuspidatum utility as functional food stuff and widely used herbal medicine.


Subject(s)
Bacteria/chemistry , Enzyme Inhibitors/pharmacology , Fallopia japonica/chemistry , Glucosides/chemistry , Neuraminidase/administration & dosage , Quinones/pharmacology , Kinetics , Quinones/chemistry , Spectrometry, Fluorescence
15.
Arch Virol ; 161(8): 2087-94, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27255748

ABSTRACT

Influenza, the most common infectious disease, poses a great threat to human health because of its highly contagious nature and fast transmissibility, often leading to high morbidity and mortality. Effective vaccination strategies may aid in the prevention and control of recurring epidemics and pandemics associated with this infectious disease. However, antigenic shifts and drifts are major concerns with influenza virus, requiring effective global monitoring and updating of vaccines. Current vaccines are standardized primarily based on the amount of hemagglutinin, a major surface antigen, which chiefly constitutes these preparations along with the varying amounts of neuraminidase (NA). Anti-influenza drugs targeting the active site of NA have been in use for more than a decade now. However, NA has not been approved as an effective antigenic component of the influenza vaccine because of standardization issues. Although some studies have suggested that NA antibodies are able to reduce the severity of the disease and induce a long-term and cross-protective immunity, a few major scientific issues need to be addressed prior to launching NA-based vaccines. Interestingly, an increasing number of studies have shown NA to be a promising target for future influenza vaccines. This review is an attempt to consolidate studies that reflect the strength of NA as a suitable vaccine target. The studies discussed in this article highlight NA as a potential influenza vaccine candidate and support taking the process of developing NA vaccines to the next stage.


Subject(s)
Influenza A virus/enzymology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Viral Proteins/immunology , Animals , Humans , Influenza A virus/genetics , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Neuraminidase/administration & dosage , Neuraminidase/genetics , Viral Proteins/administration & dosage , Viral Proteins/genetics
16.
PLoS One ; 10(12): e0143351, 2015.
Article in English | MEDLINE | ID: mdl-26629687

ABSTRACT

BACKGROUND: Preclinical and clinical studies have previously shown that systemic administration of GM1 ganglioside has neuroprotective and neurorestorative properties in Parkinson's disease (PD) models and in PD patients. However, the clinical development of GM1 for PD has been hampered by its animal origin (GM1 used in previous studies was extracted from bovine brains), limited bioavailability, and limited blood brain barrier penetrance following systemic administration. OBJECTIVE: To assess an alternative therapeutic approach to systemic administration of brain-derived GM1 to enhance GM1 levels in the brain via enzymatic conversion of polysialogangliosides into GM1 and to assess the neuroprotective potential of this approach. METHODS: We used sialidase from Vibrio cholerae (VCS) to convert GD1a, GD1b and GT1b gangliosides to GM1. VCS was infused by osmotic minipump into the dorsal third ventricle in mice over a 4-week period. After the first week of infusion, animals received MPTP injections (20 mg/kg, s.c., twice daily, 4 hours apart, for 5 consecutive days) and were euthanized 2 weeks after the last injection. RESULTS: VCS infusion resulted in the expected change in ganglioside expression with a significant increase in GM1 levels. VCS-treated animals showed significant sparing of striatal dopamine (DA) levels and substantia nigra DA neurons following MPTP administration, with the extent of sparing of DA neurons similar to that achieved with systemic GM1 administration. CONCLUSION: The results suggest that enzymatic conversion of polysialogangliosides to GM1 may be a viable treatment strategy for increasing GM1 levels in the brain and exerting a neuroprotective effect on the damaged nigrostriatal DA system.


Subject(s)
G(M1) Ganglioside/metabolism , Gene Expression Regulation/drug effects , Neuraminidase/administration & dosage , Neuraminidase/pharmacology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Parkinson Disease/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Neuraminidase/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Pars Compacta/drug effects , Pars Compacta/pathology , Vibrio cholerae/enzymology
17.
Antiviral Res ; 123: 114-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26391974

ABSTRACT

DAS181, (study drug, Fludase®) was developed for treatment of influenza and parainfluenza infections. Delivered by inhalation, DAS181 cleaves sialic acid receptors from respiratory epithelial cells. Treatment of influenza for three days with DAS181 reduced viral shedding. To increase deposition in the upper airways and decrease systemic absorption, the particle size was increased to 10µm. We conducted two Phase I trials with three cohorts, randomized 2:1, active drug to placebo. The initial cohort got a single 20mg dose of DAS181, or placebo; the second, 20mg DAS181 or placebo for 10days, and the third got 20mg of DAS181 or placebo for 3days. Formulations differed slightly in their excipients. Subjects in the 1- and 3-day cohorts completed dosing without serious adverse events. Two subjects in the 10-day cohort stopped at Day 9 after developing respiratory and systemic symptoms, and a third experienced a decrease in FEV1 (Forced Expiratory Volume in 1s) after the 9th dose and a further decline after the 10th dose. Plasma DAS181, in the 10-day cohort, peaked and began falling before the last dose. Antibodies, predominately IgG with neutralizing activity, were detected in 15/18 subjects by Day 30. The highest IgG concentrations were in the 10-day cohort. The respiratory adverse events occurring after seven days and rapid drug clearance during continued dosing are consistent with the induction of DAS181 antibodies. This could preclude use of this medication for longer than seven days or for repeated courses. (These studies have been registered at ClinicalTrials.gov under registration Nos. NCT 00527865 and NCT 01651494.).


Subject(s)
Antiviral Agents/administration & dosage , Neuraminidase/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Administration, Inhalation , Adult , Antibodies/blood , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Healthy Volunteers , Humans , Immunoglobulin G/blood , Male , Neuraminidase/adverse effects , Placebos/administration & dosage , Recombinant Fusion Proteins/adverse effects
18.
J Virol ; 89(3): 1550-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25392225

ABSTRACT

UNLABELLED: The envelope of influenza A viruses contains two large antigens, hemagglutinin (HA) and neuraminidase (NA). Conventional influenza virus vaccines induce neutralizing antibodies that are predominantly directed to the HA globular head, a domain that is subject to extensive antigenic drift. Antibodies directed to NA are induced at much lower levels, probably as a consequence of the immunodominance of the HA antigen. Although antibodies to NA may affect virus release by inhibiting the sialidase function of the glycoprotein, the antigen has been largely neglected in past vaccine design. In this study, we characterized the protective properties of monospecific immune sera that were generated by vaccination with recombinant RNA replicon particles encoding NA. These immune sera inhibited hemagglutination in an NA subtype-specific and HA subtype-independent manner and interfered with infection of MDCK cells. In addition, they inhibited the sialidase activities of various influenza viruses of the same and even different NA subtypes. With this, the anti-NA immune sera inhibited the spread of H5N1 highly pathogenic avian influenza virus and HA/NA-pseudotyped viruses in MDCK cells in a concentration-dependent manner. When chickens were immunized with NA recombinant replicon particles and subsequently infected with low-pathogenic avian influenza virus, inflammatory serum markers were significantly reduced and virus shedding was limited or eliminated. These findings suggest that NA antibodies can inhibit virus dissemination by interfering with both virus attachment and egress. Our results underline the potential of high-quality NA antibodies for controlling influenza virus replication and place emphasis on NA as a vaccine antigen. IMPORTANCE: The neuraminidase of influenza A viruses is a sialidase that acts as a receptor-destroying enzyme facilitating the release of progeny virus from infected cells. Here, we demonstrate that monospecific anti-NA immune sera inhibited not only sialidase activity, but also influenza virus hemagglutination and infection of MDCK cells, suggesting that NA antibodies can interfere with virus attachment. Inhibition of both processes, virus release and virus binding, may explain why NA antibodies efficiently blocked virus dissemination in vitro and in vivo. Anti-NA immune sera showed broader reactivity than anti-HA sera in hemagglutination inhibition tests and demonstrated cross-subtype activity in sialidase inhibition tests. These remarkable features of NA antibodies highlight the importance of the NA antigen for the development of next-generation influenza virus vaccines.


Subject(s)
Immune Sera/immunology , Influenza A virus/immunology , Neuraminidase/immunology , Viral Proteins/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Chickens , Dogs , Influenza in Birds/prevention & control , Neuraminidase/administration & dosage , Swine , Viral Proteins/administration & dosage , Virus Internalization , Virus Release/immunology , Virus Shedding
19.
Virology ; 476: 189-195, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25546254

ABSTRACT

Influenza A viruses pose a serious threat to public health. Current influenza A vaccines predominantly focus on hemagglutinin (HA) and show strain-specific protection. Neuraminidase (NA) is much less studied in the context of humoral immunity against influenza A viruses. The purpose of this study is to evaluate the cross protective immunity of NA presented on Lactococcus lactis (L.lactis) surface against homologous and heterologous influenza A viruses in the mouse model. L.lactis/pNZ8110-pgsA-NA was constructed in which pgsA was used as an anchor protein. Mice vaccinated orally with L.lactis/pNZ8110-pgsA-NA could elicit significant NA-specific serum IgG and mucosa IgA antibodies, as well as neuraminidase inhibition (NI) titers. Importantly, L.lactis/pNZ8110-pgsA-NA provided 80% protection against H5N1, 60% protection against H3N2 and H1N1, respectively. These findings suggest that recombinant L.lactis/pNZ110-pgsA-NA in the absence of adjuvant via oral administration can be served as an effective vaccine candidate against diverse strains of influenza A viruses.


Subject(s)
Cross Protection , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Lactococcus lactis/genetics , Neuraminidase/immunology , Viral Proteins/immunology , Animals , Antibodies, Viral/immunology , Female , Gene Expression , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/virology , Lactococcus lactis/metabolism , Mice , Mice, Inbred BALB C , Neuraminidase/administration & dosage , Neuraminidase/genetics , Viral Proteins/administration & dosage , Viral Proteins/genetics
20.
J Virol ; 88(12): 7016-23, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24719414

ABSTRACT

UNLABELLED: Live attenuated H7N9 influenza vaccine viruses that possess the hemagglutinin (HA) and neuraminidase (NA) gene segments from the newly emerged wild-type (wt) A/Anhui/1/2013 (H7N9) and six internal protein gene segments from the cold-adapted influenza virus A/Ann Arbor/6/60 (AA ca) were generated by reverse genetics. The reassortant virus containing the original wt A/Anhui/1/2013 HA and NA sequences replicated poorly in eggs. Multiple variants with amino acid substitutions in the HA head domain that improved viral growth were identified by viral passage in eggs and MDCK cells. The selected vaccine virus containing two amino acid changes (N133D/G198E) in the HA improved viral titer by more than 10-fold (reached a titer of 10(8.6) fluorescent focus units/ml) without affecting viral antigenicity. Introduction of these amino acid changes into an H7N9 PR8 reassortant virus also significantly improved viral titers and HA protein yield in eggs. The H7N9 ca vaccine virus was immunogenic in ferrets. A single dose of vaccine conferred complete protection of ferrets from homologous wt A/Anhui/1/2013 (H7N9) and nearly complete protection from heterologous wt A/Netherlands/219/2003 (H7N7) challenge infection. Therefore, this H7N9 live attenuated influenza vaccine (LAIV) candidate has been selected for vaccine manufacture and clinical evaluation to protect humans from wt H7N9 virus infection. IMPORTANCE: In response to the recent avian H7N9 influenza virus infection in humans, we developed a live attenuated H7N9 influenza vaccine (LAIV) with two amino acid substitutions in the viral HA protein that improved vaccine yield by 10-fold in chicken embryonated eggs, the substrate for vaccine manufacture. The two amino acids also improved the antigen yield for inactivated H7N9 vaccines, demonstrating that this finding could great facilitate the efficiency of H7N9 vaccine manufacture. The candidate H7N9 LAIV was immunogenic and protected ferrets against homologous and heterologous wild-type H7 virus challenge, making it suitable for use in protecting humans from H7 infection.


Subject(s)
Influenza A Virus, H7N7 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Animals , Cross Protection , Disease Models, Animal , Female , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H7N7 Subtype/genetics , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/virology , Male , Neuraminidase/administration & dosage , Neuraminidase/genetics , Neuraminidase/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...