Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Front Immunol ; 12: 584299, 2021.
Article in English | MEDLINE | ID: mdl-33746943

ABSTRACT

Parenteral administration of killed/inactivated swine influenza A virus (SwIAV) vaccine in weaned piglets provides variable levels of immunity due to the presence of preexisting virus specific maternal derived antibodies (MDA). To overcome the effect of MDA on SwIAV vaccine in piglets, we developed an intranasal deliverable killed SwIAV antigen (KAg) encapsulated chitosan nanoparticles called chitosan-based NPs encapsulating KAg (CS NPs-KAg) vaccine. Further, to target the candidate vaccine to dendritic cells and macrophages which express mannose receptor, we conjugated mannose to chitosan (mCS) and formulated KAg encapsulated mCS nanoparticles called mannosylated chitosan-based NPs encapsulating KAg (mCS NPs-KAg) vaccine. In MDA-positive piglets, prime-boost intranasal inoculation of mCS NPs-KAg vaccine elicited enhanced homologous (H1N2-OH10), heterologous (H1N1-OH7), and heterosubtypic (H3N2-OH4) influenza virus-specific secretory IgA (sIgA) antibody response in nasal passage compared to CS NPs-KAg vaccinates. In vaccinated upon challenged with a heterologous SwIAV H1N1, both mCS NPs-KAg and CS NPs-KAg vaccinates augmented H1N2-OH10, H1N1-OH7, and H3N2-OH4 virus-specific sIgA antibody responses in nasal swab, lung lysate, and bronchoalveolar lavage (BAL) fluid; and IgG antibody levels in lung lysate and BAL fluid samples. Whereas, the multivalent commercial inactivated SwIAV vaccine delivered intramuscularly increased serum IgG antibody response. In mCS NPs-KAg and CS NPs-KAg vaccinates increased H1N2-OH10 but not H1N1-OH7 and H3N2-OH4-specific serum hemagglutination inhibition titers were observed. Additionally, mCS NPs-KAg vaccine increased specific recall lymphocyte proliferation and cytokines IL-4, IL-10, and IFNγ gene expression compared to CS NPs-KAg and commercial SwIAV vaccinates in tracheobronchial lymph nodes. Consistent with the immune response both mCS NPs-KAg and CS NPs-KAg vaccinates cleared the challenge H1N1-OH7 virus load in upper and lower respiratory tract more efficiently when compared to commercial vaccine. The virus clearance was associated with reduced gross lung lesions. Overall, mCS NP-KAg vaccine intranasal immunization in MDA-positive pigs induced a robust cross-reactive immunity and offered protection against influenza virus.


Subject(s)
Chitosan/immunology , Immunity/immunology , Influenza Vaccines/immunology , Mannose/immunology , Orthomyxoviridae Infections/immunology , Swine Diseases/immunology , Animals , Antibodies, Viral/immunology , Cells, Cultured , Chitosan/metabolism , Dogs , Female , Immunity/drug effects , Influenza Vaccines/administration & dosage , Madin Darby Canine Kidney Cells , Mannose/metabolism , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Pregnancy , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
2.
mSphere ; 5(3)2020 05 13.
Article in English | MEDLINE | ID: mdl-32404512

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS coronavirus 2, or SARS-CoV-2) is the cause of the respiratory infection known as COVID-19. From an immunopathological standpoint, coronaviruses such as SARS-CoV-2 induce increased levels of a variety of T-helper 1 (Th1) and inflammatory cytokines and chemokines, including interleukin-1 (IL-1), IL-6, CCL2 protein, and CXCL10 protein. In the absence of proven antiviral agents or an effective vaccine, substances with immunomodulatory activity may be able to inhibit inflammatory and Th1 cytokines and/or yield an anti-inflammatory and/or Th2 immune response to counteract COVID-19 symptoms and severity. This report briefly describes the following four unconventional but commercially accessible immunomodulatory agents that can be employed in clinical trials to evaluate their effectiveness at alleviating disease symptoms and severity: low-dose oral interferon alpha, microdose DNA, low-dose thimerosal, and phytocannabinoids.


Subject(s)
Cannabinoids/therapeutic use , Coronavirus Infections/drug therapy , DNA/therapeutic use , Immunomodulation , Interferon-alpha/therapeutic use , Pneumonia, Viral/drug therapy , Thimerosal/therapeutic use , Betacoronavirus , COVID-19 , Cytokines/immunology , Humans , Pandemics , Phytochemicals/therapeutic use , SARS-CoV-2 , COVID-19 Drug Treatment
3.
Vaccines (Basel) ; 8(2)2020 May 18.
Article in English | MEDLINE | ID: mdl-32443416

ABSTRACT

Intranasal vaccination elicits secretory IgA (SIgA) antibodies in the airways, which is required for cross-protection against influenza. To enhance the breadth of immunity induced by a killed swine influenza virus antigen (KAg) or conserved T cell and B cell peptides, we adsorbed the antigens together with the TLR3 agonist poly(I:C) electrostatically onto cationic alpha-D-glucan nanoparticles (Nano-11) resulting in Nano-11-KAg-poly(I:C) and Nano-11-peptides-poly(I:C) vaccines. In vitro, increased TNF-α and IL-1ß cytokine mRNA expression was observed in Nano-11-KAg-poly(I:C)-treated porcine monocyte-derived dendritic cells. Nano-11-KAg-poly(I:C), but not Nano-11-peptides-poly(I:C), delivered intranasally in pigs induced high levels of cross-reactive virus-specific SIgA antibodies secretion in the nasal passage and lungs compared to a multivalent commercial influenza virus vaccine administered intramuscularly. The commercial and Nano-11-KAg-poly(I:C) vaccinations increased the frequency of IFNγ secreting T cells. The poly(I:C) adjuvanted Nano-11-based vaccines increased various cytokine mRNA expressions in lymph nodes compared to the commercial vaccine. In addition, Nano-11-KAg-poly(I:C) vaccine elicited high levels of virus neutralizing antibodies in bronchoalveolar lavage fluid. Microscopic lung lesions and challenge virus load were partially reduced in poly(I:C) adjuvanted Nano-11 and commercial influenza vaccinates. In conclusion, compared to our earlier study with Nano-11-KAg vaccine, addition of poly(I:C) to the formulation improved cross-protective antibody and cytokine response.

4.
Vet Microbiol ; 242: 108611, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32122615

ABSTRACT

To improve the innate and adaptive immune responses elicited by a killed/inactivated swine influenza virus antigen (KAg)-loaded chitosan nanoparticles (CS NPs-KAg), we used the adjuvant, poly(I:C). The formulated CS NPs-KAg and CS NPs-poly(I:C) had a net surface charge of +30.7 mV and +25.1 mV, respectively. The CS NPs-KAg was coadministered with CS NPs-poly(I:C) (chitosan nanovaccine) as intranasal mist. Vaccinations enhanced homologous (H1N2-OH10) and heterologous (H1N1-OH7) hemagglutination inhibition (HI) titers in both vaccinated and virus-challenged animals compared to the control soluble poly(I:C) vaccinated pigs. In addition, the chitosan nanovaccine induced the proliferation of antigen-specific IFNγ secreting T-helper/memory and γδ T cells compared to control poly(I:C) group; and an increased Th1 (IFNγ, IL-6 and IL-2) and Th2 (IL-10 and IL-13) cytokines mRNA expression in the tracheobronchial lymph nodes compared to lymphoid tissues obtained from pigs given commercial influenza vaccine. The virus load in nasal passages and microscopic lung lesions were partially reduced by both chitosan nanovaccine and commercial vaccine. The HA gene homology between the vaccine and challenge viruses indicated that the chitosan nanovaccine induced a cross-protective immune response. In conclusion, coadministration of CS NPs-poly(I:C) with CS NPs-KAg augmented the cross-reactive specific HI titers and the cell-mediated immune responses in pigs.


Subject(s)
Immunity, Cellular , Influenza Vaccines/immunology , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/prevention & control , Poly I-C/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/blood , Chitosan , Cytokines/genetics , Cytokines/immunology , Hemagglutination Inhibition Tests , Immunity, Innate , Influenza Vaccines/administration & dosage , Poly I-C/immunology , Swine , Swine Diseases/prevention & control , Th1 Cells/immunology , Th2 Cells/immunology , Viral Load
5.
Front Immunol ; 10: 1509, 2019.
Article in English | MEDLINE | ID: mdl-31379808

ABSTRACT

The gut microbiome plays an important role in the immune system development, maintenance of normal health status, and in disease progression. In this study, we comparatively examined the fecal microbiomes of Amish (rural) and non-Amish (urban) infants and investigated how they could affect the mucosal immune maturation in germ-free piglets that were inoculated with the two types of infant fecal microbiota (IFM). Differences in microbiome diversity and structure were noted between the two types of fecal microbiotas. The fecal microbiota of the non-Amish (urban) infants had a greater relative abundance of Actinobacteria and Bacteroidetes phyla, while that of the Amish (rural) counterparts was dominated by Firmicutes. Amish infants had greater species richness compared with the non-Amish infants' microbiota. The fecal microbiotas of the Amish and the non-Amish infants were successfully transplanted into germ-free piglets, and the diversity and structure of the microbiota in the transplanted piglets remained similar at phylum level but not at the genus level. Principal coordinates analysis (PCoA) based on Weighted-UniFrac distance revealed distinct microbiota structure in the intestines of the transplanted piglets. Shotgun metagenomic analysis also revealed clear differences in functional diversity of fecal microbiome between Amish and non-Amish donors as well as microbiota transplanted piglets. Specific functional features were enriched in either of the microbiota transplanted piglet groups directly corresponding to the predominance of certain bacterial populations in their gut environment. Some of the colonized bacterial genera were correlated with the frequency of important lymphoid and myeloid immune cells in the ileal submucosa and mesenteric lymph nodes (MLN), both important for mucosal immune maturation. Overall, this study demonstrated that transplantation of diverse IFM into germ-free piglets largely recapitulates the differences in gut microbiota structure between rural (Amish) and urban (non-Amish) infants. Thus, fecal microbiota transplantation to germ-free piglets could be a useful large animal model system for elucidating the impact of gut microbiota on the mucosal immune system development. Future studies can focus on determining the additional advantages of the pig model over the rodent model.


Subject(s)
Feces/microbiology , Gastrointestinal Microbiome/immunology , Microbiota/immunology , Mucous Membrane/immunology , Mucous Membrane/microbiology , Amish , Animals , Fecal Microbiota Transplantation/methods , Firmicutes/immunology , Humans , Infant , Metagenome/immunology , Swine
6.
Int J Nanomedicine ; 13: 6699-6715, 2018.
Article in English | MEDLINE | ID: mdl-30425484

ABSTRACT

BACKGROUND: Influenza (flu) is a constant threat to humans and animals, and vaccination is one of the most effective ways to mitigate the disease. Due to incomplete protection induced by current flu vaccines, development of novel flu vaccine candidates is warranted to achieve greater efficacy against constantly evolving flu viruses. METHODS: In the present study, we used liposome nanoparticle (<200 nm diameter)-based subunit flu vaccine containing ten encapsulated highly conserved B and T cell epitope peptides to induce protective immune response against a zoonotic swine influenza A virus (SwIAV) H1N1 challenge infection in a pig model. Furthermore, we used monosodium urate (MSU) crystals as an adjuvant and co-administered the vaccine formulation as an intranasal mist to flu-free nursery pigs, twice at 3-week intervals. RESULTS: Liposome peptides flu vaccine delivered with MSU adjuvant improved the hemagglutination inhibition antibody titer and mucosal IgA response against the SwIAV challenge and also against two other highly genetically variant IAVs. Liposomal vaccines also enhanced the frequency of peptides and virus-specific T-helper/memory cells and IFN-γ response. The improved specific cellular and mucosal humoral immune responses in adjuvanted liposomal peptides flu vaccine partially protected pigs from flu-induced fever and pneumonic lesions, and reduced the nasal virus shedding and viral load in the lungs. CONCLUSION: Overall, our study shows great promise for using liposome and MSU adjuvant- based subunit flu vaccine through the intranasal route, and provides scope for future, pre-clinical investigations in a pig model for developing potent human intranasal subunit flu vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity , Influenza Vaccines/immunology , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology , Peptides/immunology , Uric Acid/pharmacology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Amino Acid Sequence , Animals , Antibody Formation/drug effects , Body Temperature/drug effects , Cytokines/biosynthesis , Dogs , Immunity/drug effects , Immunity, Mucosal/drug effects , Immunologic Memory/drug effects , Influenza A Virus, H1N1 Subtype , Liposomes , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Nanoparticles/ultrastructure , Orthomyxoviridae Infections/virology , Peptides/chemistry , Sus scrofa , Th1 Cells/drug effects , Th1 Cells/immunology , Vaccination , Viral Load/drug effects
7.
Front Immunol ; 9: 934, 2018.
Article in English | MEDLINE | ID: mdl-29770135

ABSTRACT

Annually, swine influenza A virus (SwIAV) causes severe economic loss to swine industry. Currently used inactivated SwIAV vaccines administered by intramuscular injection provide homologous protection, but limited heterologous protection against constantly evolving field viruses, attributable to the induction of inadequate levels of mucosal IgA and cellular immune responses in the respiratory tract. A novel vaccine delivery platform using mucoadhesive chitosan nanoparticles (CNPs) administered through intranasal (IN) route has the potential to elicit strong mucosal and systemic immune responses in pigs. In this study, we evaluated the immune responses and cross-protective efficacy of IN chitosan encapsulated inactivated SwIAV vaccine in pigs. Killed SwIAV H1N2 (δ-lineage) antigens (KAg) were encapsulated in chitosan polymer-based nanoparticles (CNPs-KAg). The candidate vaccine was administered twice IN as mist to nursery pigs. Vaccinates and controls were then challenged with a zoonotic and virulent heterologous SwIAV H1N1 (γ-lineage). Pigs vaccinated with CNPs-KAg exhibited an enhanced IgG serum antibody and mucosal secretory IgA antibody responses in nasal swabs, bronchoalveolar lavage (BAL) fluids, and lung lysates that were reactive against homologous (H1N2), heterologous (H1N1), and heterosubtypic (H3N2) influenza A virus strains. Prior to challenge, an increased frequency of cytotoxic T lymphocytes, antigen-specific lymphocyte proliferation, and recall IFN-γ secretion by restimulated peripheral blood mononuclear cells in CNPs-KAg compared to control KAg vaccinates were observed. In CNPs-KAg vaccinated pigs challenged with heterologous virus reduced severity of macroscopic and microscopic influenza-associated pulmonary lesions were observed. Importantly, the infectious SwIAV titers in nasal swabs [days post-challenge (DPC) 4] and BAL fluid (DPC 6) were significantly (p < 0.05) reduced in CNPs-KAg vaccinates but not in KAg vaccinates when compared to the unvaccinated challenge controls. As well, an increased frequency of T helper memory cells and increased levels of recall IFNγ secretion by tracheobronchial lymph nodes cells were observed. In summary, chitosan SwIAV nanovaccine delivered by IN route elicited strong cross-reactive mucosal IgA and cellular immune responses in the respiratory tract that resulted in a reduced nasal viral shedding and lung virus titers in pigs. Thus, chitosan-based influenza nanovaccine may be an ideal candidate vaccine for use in pigs, and pig is a useful animal model for preclinical testing of particulate IN human influenza vaccines.


Subject(s)
Chitosan , Immunity, Mucosal , Influenza Vaccines/immunology , Nanoparticles , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , Vaccines, Inactivated/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Antibody Specificity/immunology , Chitosan/chemistry , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Cellular , Influenza Vaccines/administration & dosage , Lymphocyte Activation/immunology , Nanoparticles/chemistry , Swine , Swine Diseases/immunology , Swine Diseases/metabolism , Swine Diseases/pathology , Vaccines, Inactivated/administration & dosage , Virus Shedding
8.
Vaccine ; 35(8): 1124-1131, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28117173

ABSTRACT

We have recently demonstrated the effectiveness of an influenza A virus (IAV) subunit vaccine based on biodegradable polyanhydride nanoparticles delivery in mice. In the present study, we evaluated the efficacy of ∼200nm polyanhydride nanoparticles encapsulating inactivated swine influenza A virus (SwIAV) as a vaccine to induce protective immunity against a heterologous IAV challenge in pigs. Nursery pigs were vaccinated intranasally twice with inactivated SwIAV H1N2 (KAg) or polyanhydride nanoparticle-encapsulated KAg (KAg nanovaccine), and efficacy was evaluated against a heterologous zoonotic virulent SwIAV H1N1 challenge. Pigs were monitored for fever daily. Local and systemic antibody responses, antigen-specific proliferation of peripheral blood mononuclear cells, gross and microscopic lung lesions, and virus load in the respiratory tract were compared among the groups of animals. Our pre-challenge results indicated that KAg nanovaccine induced virus-specific lymphocyte proliferation and increased the frequency of CD4+CD8αα+ T helper and CD8+ cytotoxic T cells in peripheral blood mononuclear cells. KAg nanovaccine-immunized pigs were protected from fever following SwIAV challenge. In addition, pigs immunized with the KAg nanovaccine presented with lower viral antigens in lung sections and had 6 to 8-fold reduction in nasal shedding of SwIAV four days post-challenge compared to control animals. Immunologically, increased IFN-γ secreting T lymphocyte populations against both the vaccine and challenge viruses were detected in KAg nanovaccine-immunized pigs compared to the animals immunized with KAg alone. However, in the KAg nanovaccine-immunized pigs, hemagglutination inhibition, IgG and IgA antibody responses, and virus neutralization titers were comparable to that in the animals immunized with KAg alone. Overall, our data indicated that intranasal delivery of polyanhydride-based SwIAV nanovaccine augmented antigen-specific cellular immune response in pigs, with promise to induce cross-protective immunity.


Subject(s)
Antibodies, Viral/biosynthesis , Influenza Vaccines/administration & dosage , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , Administration, Intranasal , Animals , Cell Proliferation/drug effects , Hemagglutination Inhibition Tests , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/drug effects , Influenza A Virus, H1N2 Subtype/immunology , Influenza Vaccines/chemistry , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Lung/drug effects , Lung/immunology , Lung/virology , Nanoparticles/chemistry , Nanoparticles/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Polyanhydrides/chemistry , Polyanhydrides/metabolism , Swine , Swine Diseases/immunology , Swine Diseases/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Vaccines, Inactivated , Viral Load/drug effects
9.
J Control Release ; 247: 194-205, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28057521

ABSTRACT

Swine influenza virus (SwIV) is one of the important zoonotic pathogens. Current flu vaccines have failed to provide cross-protection against evolving viruses in the field. Poly(lactic-co-glycolic acid) (PLGA) is a biodegradable FDA approved polymer and widely used in drug and vaccine delivery. In this study, inactivated SwIV H1N2 antigens (KAg) encapsulated in PLGA nanoparticles (PLGA-KAg) were prepared, which were spherical in shape with 200 to 300nm diameter, and induced maturation of antigen presenting cells in vitro. Pigs vaccinated twice with PLGA-KAg via intranasal route showed increased antigen specific lymphocyte proliferation and enhanced the frequency of T-helper/memory and cytotoxic T cells (CTLs) in peripheral blood mononuclear cells (PBMCs). In PLGA-KAg vaccinated and heterologous SwIV H1N1 challenged pigs, clinical flu symptoms were absent, while the control pigs had fever for four days. Grossly and microscopically, reduced lung pathology and viral antigenic mass in the lung sections with clearance of infectious challenge virus in most of the PLGA-KAg vaccinated pig lung airways were observed. Immunologically, PLGA-KAg vaccine irrespective of not significantly boosting the mucosal antibody response, it augmented the frequency of IFN-γ secreting total T cells, T-helper and CTLs against both H1N2 and H1N1 SwIV. In summary, inactivated influenza virus delivered through PLGA-NPs reduced the clinical disease and induced cross-protective cell-mediated immune response in a pig model. Our data confirmed the utility of a pig model for intranasal particulate flu vaccine delivery platform to control flu in humans.


Subject(s)
Antigens, Viral/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Lactic Acid/chemistry , Nanoparticles/chemistry , Orthomyxoviridae Infections/prevention & control , Polyglycolic Acid/chemistry , Vaccines, Inactivated/administration & dosage , Administration, Intranasal , Animals , Antigens, Viral/immunology , Antigens, Viral/therapeutic use , Cells, Cultured , Dogs , Immunity, Cellular , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/immunology , Polylactic Acid-Polyglycolic Acid Copolymer , Swine , Vaccines, Inactivated/immunology , Vaccines, Inactivated/therapeutic use
10.
Vet Immunol Immunopathol ; 172: 64-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27032505

ABSTRACT

While the safety and efficacy profiles of orally administered bovine interferon (IFN) alpha have been documented, the mechanism(s) that result in clinical benefits remain elusive. One approach to delineating the molecular pathways of IFN efficacy is through the use of gene expression profiling technologies. In this proof-of-concept study, different (0, 50, 200 and 800 units) oral doses of natural bovine IFN (type I) were tested in cattle to determine if oral IFN altered the expression of genes that may be pivotal to the development of systemic resistance to viral infections such as foot-and-mouth disease (FMD). Oral IFN was administered twice: Time 0 and 8h later. Blood was collected at 0, 8 and 24h after the first IFN administration, and DNA isolated from peripheral blood mononuclear cells (PBMCs) was employed in quantitative polymerase chain reaction (qPCR) microarray assays. Within 8h, 50 and 200 units of oral IFN induced significant (P<0.05) changes in expression of 41 of 92 tested autoimmune and inflammatory response-associated genes. These data suggest that orally administered IFN is a viable approach for providing short-term antiviral immunity to livestock exposed to viruses such as FMD virus (FMDV) until such a time that an effective vaccine can be produced and distributed to producers.


Subject(s)
Autoimmunity/drug effects , Cattle , Gene Expression/drug effects , Interferon-alpha/therapeutic use , Animals , Autoimmunity/genetics , Creatine Kinase/blood , Cytokines/genetics , Dose-Response Relationship, Drug , Interferon-alpha/administration & dosage , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Receptors, Cytokine/genetics
11.
Vet Microbiol ; 186: 90-6, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27016762

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PdCV) cause indistinguishable clinical signs and pathological changes in swine. Here we investigated the antigenic relationship between PEDV and PdCV. We provide the first evidence that conserved epitope(s) on the respective viral nucleocapsid proteins cross-react with each other although virus neutralization cross-reactivity was not observed. As a practical matter, prevention of these two very similar diseases of swine will require the development of separate virus-specific vaccine products.


Subject(s)
Coronavirus Infections/veterinary , Coronavirus/immunology , Porcine epidemic diarrhea virus/immunology , Swine Diseases/immunology , Swine Diseases/virology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cell Line , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cross Reactions/immunology , Intestines/immunology , Intestines/virology , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Specific Pathogen-Free Organisms , Swine , Vero Cells
12.
Appl Environ Microbiol ; 81(19): 6679-87, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26187968

ABSTRACT

Human norovirus (NoV) is responsible for over 90% of outbreaks of acute nonbacterial gastroenteritis worldwide and accounts for 60% of cases of foodborne illness in the United States. Currently, the infectivity of human NoVs is poorly understood due to the lack of a cell culture system. In this study, we determined the survival of a human NoV genogroup II, genotype 4 (GII.4) strain in seeded oyster homogenates after high-pressure processing (HPP) using a novel receptor binding assay and a gnotobiotic pig model. Pressure conditions of 350 MPa at 0°C for 2 min led to a 3.7-log10 reduction in the number of viral RNA copies in oysters, as measured by the porcine gastric mucin-conjugated magnetic bead (PGM-MB) binding assay and real-time RT-PCR, whereas pressure conditions of 350 MPa at 35°C for 2 min achieved only a 1-log10 reduction in the number of RNA copies. Newborn gnotobiotic piglets orally fed oyster homogenate treated at 350 MPa and 0°C for 2 min did not have viral RNA shedding in feces, histologic lesions, or viral replication in the small intestine. In contrast, gnotobiotic piglets fed oysters treated at 350 MPa and 35°C for 2 min had high levels of viral shedding in feces and exhibited significant histologic lesions and viral replication in the small intestine. Collectively, these data demonstrate that (i) human NoV survival estimated by an in vitro PGM-MB virus binding assay is consistent with the infectivity determined by an in vivo gnotobiotic piglet model and (ii) HPP is capable of inactivating a human NoV GII.4 strain at commercially acceptable pressure levels.


Subject(s)
Caliciviridae Infections/virology , Food Handling/methods , Foodborne Diseases/virology , Norovirus/physiology , Ostreidae/virology , Shellfish/virology , Virus Inactivation , Animals , Disease Models, Animal , Food Contamination/prevention & control , Food Handling/instrumentation , Germ-Free Life , Humans , Norovirus/chemistry , Pressure , Swine
13.
mBio ; 6(2): e00064, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25759498

ABSTRACT

UNLABELLED: A novel porcine deltacoronavirus (PdCV) was first discovered in Ohio and Indiana in February 2014, rapidly spread to other states in the United States and Canada, and caused significant economic loss in the swine industry. The origin and virulence of this novel porcine coronavirus are not known. Here, we characterized U.S. PdCV isolates and determined their virulence in gnotobiotic and conventional piglets. Genome analyses revealed that U.S. PdCV isolates possess unique genetic characteristics and share a close relationship with Hong Kong and South Korean PdCV strains and coronaviruses (CoVs) of Asian leopard cats and Chinese ferret-badgers. The PdCV-positive intestinal content (Ohio CVM1) and the cell culture-adapted PdCV Michigan (MI) strain were orally inoculated into gnotobiotic and/or conventional piglets. Within 1 to 3 days postinfection, profuse watery diarrhea, vomiting, and dehydration were observed. Clinical signs were associated with epithelial necrosis in the gastric pits and small intestine, the latter resulting in severe villous atrophy. Mild interstitial pneumonia was identified in the lungs of PdCV-infected piglets. High levels of viral RNA (8 to 11 log RNA copies/g) were detected in intestinal tissues/luminal contents and feces of infected piglets, whereas moderate RNA levels (2 to 5 log RNA copies/g) were detected in blood, lung, liver, and kidney, indicating multisystemic dissemination of the virus. Polyclonal immune serum against PdCV but not immune serum against porcine epidemic diarrhea virus (PEDV) reacted with PdCV-infected small-intestinal epithelial cells, indicating that PdCV is antigenically distinct from PEDV. Collectively, we demonstrate for the first time that PdCV caused severe gastrointestinal diseases in swine. IMPORTANCE: Porcine coronaviruses (CoVs) are major viral infectious diseases of swine. Examples of porcine CoVs include porcine transmissible gastroenteritis coronavirus (TGEV), porcine epidemic diarrhea virus (PEDV), and porcine respiratory coronavirus (PRCV). In February 2014, another porcine CoV, porcine deltacoronavirus (PdCV), emerged in Ohio and Indiana and subsequently spread rapidly across the United States and Canada, causing significant economic losses. Here, we report the detailed genetic characterization, phylogeny, and virulence of emergent PdCV strains in the United States. We found that PdCV caused severe diarrhea, vomiting, and dehydration in gnotobiotic and conventional piglets, signs that were clinically indistinguishable from those caused by PEDV and TGEV. In addition to extensive intestinal lesions, PdCV caused significant lesions in the stomach and mild pulmonary lesions that have not been reported for TGEV and PEDV. The finding that PdCV is a significant enteric disease of swine highlights the need to develop effective measures to control this disease.


Subject(s)
Coronavirus Infections/veterinary , Coronavirus/classification , Coronavirus/pathogenicity , Diarrhea/veterinary , Swine Diseases/virology , Animals , Cluster Analysis , Coronavirus/genetics , Coronavirus/isolation & purification , Coronavirus Infections/pathology , Coronavirus Infections/virology , Diarrhea/complications , Diarrhea/pathology , Diarrhea/virology , Feces/virology , Intestine, Small/pathology , Intestine, Small/virology , Molecular Sequence Data , Phylogeny , Pneumonia, Viral/pathology , Pneumonia, Viral/veterinary , Pneumonia, Viral/virology , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Homology , Stomach/pathology , Stomach/virology , Swine , United States , Virulence
14.
Can Vet J ; 55(9): 857-64, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25183893

ABSTRACT

Bordetella bronchiseptica (Bb) whole cell bacterins have been replaced with acelluar vaccines. We evaluated the response to the acellular Bb vaccines in Bb-seropositive commingled laboratory beagles and client-owned dogs with various lifestyles and vaccination histories. A single parenteral dose of the acellular Bb vaccine resulted in consistent anamnestic IgG, and to a lesser, but notable extent, IgA, Bb-reactive antibody responses in the seropositive beagles. Associated with the increase in antibodies measured by enzyme-linked immunosorbent assay (ELISA) was an increase in the complement (C)-dependent IgG antibody mediated bactericidal effect on Bb in vitro. Antibody responses in client-owned dogs were more variable and were dependent upon the vaccination history and serological evidence of previous Bb exposure. Antibodies from vaccinated dogs recognized several Bb proteins, notably P68 (pertactin) and P220 (fimbrial hemagglutinin), the response to which has been shown to be disease-sparing in Bp infections. These antibody responses were similar to those in experimentally infected dogs and in dogs that had received a widely used whole cell bacterin.


Réponses des anticorps àBordetella bronchisepticachez des chiens vaccinés et infectés. Des bactérines de Bordetella bronchiseptica (Bb) ont été remplacées par des vaccins acellulaires. Nous avons évalué la réponse aux vaccins Bb acellulaires chez un groupe de Beagles de laboratoire séropositifs pour le Bb et des chiens de clients ayant divers styles de vie et antécédents de vaccination. Une seule dose parentérale du vaccin Bb acellulaire s'est traduite par une réponse IgG anamnestique uniforme, et à degré inférieur mais significatif, des réponses IgA et des anticorps réactifs à Bb chez les Beagles séropositifs. Une hausse des anticorps mesurés par ELISA a été accompagnée d'une augmentation de l'effet bactéricide atténué des anticorps dépendants IgG du complément (C) sur Bb in vitro. Les réponses des anticorps chez les chiens appartenant à des clients étaient plus variables et dépendaient des antécédents de vaccination et des preuves sérologiques d'une exposition antérieure à Bb. Les anticorps de chiens vaccinés reconnaissaient plusieurs protéines Bb, notamment P68 (pertactine) et P220 (hémagglutinine fimbriale), dont la réponse a été démontrée comme une protection contre la maladie lors d'une infection par Bb. Ces réponses des anticorps étaient semblables à celles des chiens infectés par expérimentation et à celles des chiens qui avaient reçu des bactérines à bacilles entiers généralement utilisés.(Traduit par Isabelle Vallières).


Subject(s)
Antibodies, Bacterial/blood , Bordetella Infections/veterinary , Bordetella bronchiseptica/immunology , Dog Diseases/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Bordetella Infections/immunology , Bordetella Infections/prevention & control , Dogs , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Immunoglobulin G/blood , Injections, Subcutaneous/veterinary , Male , Vaccination/veterinary
15.
PLoS Pathog ; 10(6): e1004209, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967579

ABSTRACT

Salmonella enterica serovar Typhimurium (Salmonella) is one of the most significant food-borne pathogens affecting both humans and agriculture. We have determined that Salmonella encodes an uptake and utilization pathway specific for a novel nutrient, fructose-asparagine (F-Asn), which is essential for Salmonella fitness in the inflamed intestine (modeled using germ-free, streptomycin-treated, ex-germ-free with human microbiota, and IL10-/- mice). The locus encoding F-Asn utilization, fra, provides an advantage only if Salmonella can initiate inflammation and use tetrathionate as a terminal electron acceptor for anaerobic respiration (the fra phenotype is lost in Salmonella SPI1- SPI2- or ttrA mutants, respectively). The severe fitness defect of a Salmonella fra mutant suggests that F-Asn is the primary nutrient utilized by Salmonella in the inflamed intestine and that this system provides a valuable target for novel therapies.


Subject(s)
Asparagine/metabolism , Fructose/metabolism , Intestinal Mucosa/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Anaerobiosis , Animals , Bacterial Proteins/genetics , Biological Transport/genetics , Cation Transport Proteins/genetics , Disease Models, Animal , Energy Metabolism/genetics , Humans , Inflammation/immunology , Inflammation/microbiology , Interleukin-10/genetics , Intestines/immunology , Intestines/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections, Animal/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development
16.
J Virol ; 88(5): 2913-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371058

ABSTRACT

UNLABELLED: One role of mRNA cap guanine-N-7 (G-N-7) methylation is to facilitate the efficient translation of mRNA. The role of mRNA cap ribose 2'-O methylation is enigmatic, although recent work has implicated this as a signature to avoid detection of RNA by the innate immune system (S. Daffis, K. J. Szretter, J. Schriewer, J. Q. Li, S. Youn, J. Errett, T. Y. Lin, S. Schneller, R. Zust, H. P. Dong, V. Thiel, G. C. Sen, V. Fensterl, W. B. Klimstra, T. C. Pierson, R. M. Buller, M. Gale, P. Y. Shi, M. S. Diamond, Nature 468:452-456, 2010, doi:10.1038/nature09489). Working with vesicular stomatitis virus (VSV), we previously showed that a panel of recombinant VSVs carrying mutations at a predicted methyltransferase catalytic site (rVSV-K1651A, -D1762A, and -E1833Q) or S-adenosylmethionine (SAM) binding site (rVSV-G1670A, -G1672A, and -G4A) were defective in cap methylation and were also attenuated for growth in cell culture. Here, we analyzed the virulence of these recombinants in mice. We found that rVSV-K1651A, -D1762A, and -E1833Q, which are defective in both G-N-7 and 2'-O methylation, were highly attenuated in mice. All three viruses elicited a high level of neutralizing antibody and provided full protection against challenge with the virulent VSV. In contrast, mice inoculated with rVSV-G1670A and -G1672A, which are defective only in G-N-7 methylation, were attenuated in vivo yet retained a low level of virulence. rVSV-G4A, which is completely defective in both G-N-7 and 2'-O methylation, also exhibited low virulence in mice despite the fact that productive viral replication was not detected in lung and brain. Taken together, our results suggest that abrogation of viral mRNA cap methylation can serve as an approach to attenuate VSV, and perhaps other nonsegmented negative-strand RNA viruses, for potential application as vaccines and viral vectors. IMPORTANCE: Nonsegmented negative-sense (NNS) RNA viruses include a wide range of significant human, animal, and plant pathogens. For many of these viruses, there are no vaccines or antiviral drugs available. mRNA cap methylation is essential for mRNA stability and efficient translation. Our current understanding of mRNA modifications of NNS RNA viruses comes largely from studies of vesicular stomatitis virus (VSV). In this study, we showed that recombinant VSVs (rVSVs) defective in mRNA cap methylation were attenuated in vitro and in vivo. In addition, these methyltransferase (MTase)-defective rVSVs triggered high levels of antibody responses and provided complete protection against VSV infection. Thus, this study will not only contribute to our understanding of the role of mRNA cap MTase in viral pathogenesis but also facilitate the development of new live attenuated vaccines for VSV, and perhaps other NNS RNA viruses, by inhibiting viral mRNA cap methylation.


Subject(s)
RNA Caps/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/metabolism , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Brain/pathology , Brain/virology , Cell Line , Defective Viruses/genetics , Defective Viruses/metabolism , Female , Lethal Dose 50 , Lung/pathology , Lung/virology , Methylation , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Phenotype , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/pathogenicity , Viral Load , Virulence , Virus Replication
17.
Virus Res ; 164(1-2): 4-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21945213

ABSTRACT

Porcine circoviruses (PCVs) belong to the genus Circovirus, family Circoviridae, and are the smallest non-enveloped, single stranded, negative sense, circular DNA viruses that replicate autonomously in mammalian cells. Two types of PCV have been characterised, PCV1 and PCV2 and these two viruses show 83% sequence identity at open reading frame (ORF) 1 and 67% identity at ORF2. PCV1 is a non-pathogenic virus of pigs. In contrast, PCV2 has emerged as a major pathogen of swine around the world. The discovery of PCV1 and how the subsequent studies on this virus eventually led to the recognition and characterisation of PCV2, and the disease scenarios associated with PCV2, serve as a model of how multidisciplinary collaboration among field veterinarians, diagnosticians and researchers can lead to the rapid characterisation and control of a globally important emerging disease.


Subject(s)
Circoviridae Infections/veterinary , Circovirus/isolation & purification , Circovirus/pathogenicity , Swine Diseases/epidemiology , Swine Diseases/history , Animals , Circoviridae Infections/epidemiology , Circoviridae Infections/history , Circovirus/classification , Circovirus/genetics , History, 20th Century , History, 21st Century , Swine , Swine Diseases/virology
18.
Virus Res ; 164(1-2): 90-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22100341

ABSTRACT

Porcine circovirus type 2 (PCV2) was retrospectively identified by serology in swine populations as an asymptomatic infection at least 25 years prior to the first reported case of PCV2-associated postweaning multisystemic wasting syndrome (PMWS). To investigate the sudden emergence of PMWS, viral sequences were amplified from frozen archived (1970-1971) porcine tissues and the complete genome of archival PCV2 was determined. The ORF1 gene product (viral DNA replicase) was homologous to contemporary PCV2 ORF1. In ORF2 (viral nucleocapsid gene) archival PCV2, a consistent linear nine-base sequence difference at base positions 1331 through 1339 was observed. The deduced amino acid sequence from these base changes alters the nucleocapsid conformation within the second immunogenic epitope from a hydrophobic (contemporary PCV2) to a hydrophilic (archival PCV2) configuration. To test the hypothesis that archival PCV2 was avirulent, cloned engineered archival and contemporary PCV2 genomes were constructed wherein the ORF1 gene was identical in each clone and the ORF2 gene (nucleocapsid protein) was sequence-identical in both clones except for the nine-base difference (bases 1331-1339), corresponding to archival and contemporary PCV2 viruses respectively. Clones were transfected into porcine kidney (PK) 15 cells and, after sequence confirmation, further passed in PK15 and 3D4/2 porcine alveolar macrophage cell cultures. Virulence trials in gnotobiotic piglets were conducted with cloned PCV2s. The data show that archival PCV2 is avirulent when compared to contemporary PCV2 and supports the hypothesis that the emergence of virulent contemporary PCV2 was a result of mutational events within this critical epitope after 1971.


Subject(s)
Circovirus/genetics , Circovirus/pathogenicity , Nucleocapsid/genetics , Porcine Postweaning Multisystemic Wasting Syndrome/virology , Virulence Factors/genetics , Virus Replication , Amino Acid Motifs , Animals , Circovirus/growth & development , DNA, Viral/chemistry , DNA, Viral/genetics , Porcine Postweaning Multisystemic Wasting Syndrome/pathology , Sequence Analysis, DNA , Swine , Virulence
19.
Can J Vet Res ; 76(4): 301-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23543956

ABSTRACT

This study investigated if parenteral administration of a prototype adjuvanted vaccine against porcine circovirus type 2 (PCV2) could override maternally derived antibodies and induce acquired immunity in young piglets. Piglets with high levels of maternal PCV2 antibodies at 1 wk of age were randomly grouped into vaccinates and controls on the basis of body weight and inoculated with the vaccine or a control preparation twice, with an interval of 3 wk. Both groups were challenged 3 wk after the booster vaccination and euthanized 3 wk after challenge. The pigs were evaluated for clinical disease, histologic lesions in sections of gastric and left inguinal lymph nodes stained with hematoxylin and eosin, and the amount of PCV2 antigen in the lymph nodes by immunohistochemical study. The PCV2 antibody titers were monitored by competitive enzyme-linked immunosorbent assay throughout the experiment. The vaccinates showed significantly less decline (P < 0.05) in PCV2 antibody titers after the booster vaccination. Clinical disease did not develop in any of the piglets. The vaccinates and controls did not differ in either histologic lesions or amount of PCV2 antigen in the lymph nodes. This study demonstrated some evidence of priming of young piglets in the presence of maternal antibodies. Further studies are recommended to determine the optimum concentration of PCV2 antigen and a suitable adjuvant for the vaccine to achieve the full potential of the strategy of inducing acquired immunity in young piglets that have maternally derived antibodies.


Cette étude visait à déterminer si l'administration parentérale d'un prototype de vaccin avec adjuvant dirigé contre le circovirus porcin de type 2 (PCV2) pouvait outrepasser les anticorps maternels et induire une immunité acquise chez les jeunes porcelets. Les porcelets avec des niveaux élevés d'anticorps maternels anti-PCV2 à 1 sem d'âge étaient regroupés de manière aléatoire en vaccinés et témoins basés sur le poids corporel et inoculés avec le vaccin ou une préparation témoin deux fois à un intervalle de trois semaines. Les deux groupes ont été soumis à une infection défi 3 sem après la vaccination de rappel et euthanasiés 3 sem après l'infection. Les porcs ont été évalués pour la présence de maladie clinique, de lésions histologiques dans des sections de noeuds lymphatiques gastriques et inguinal gauche colorés avec de l'hématoxyline et éosine, et la quantité d'antigène PCV2 dans les noeuds lymphatiques par étude immunohistochimique. Les titres d'anticorps anti-PCV2 ont été suivis par épreuve immuno-enzymatique compétitive tout au long de l'expérience. Les animaux vaccinés ont présenté une diminution significativement moindre (P < 0,05) des titres d'anticorps anti-PCV2 après le rappel de vaccin. La maladie clinique ne s'est développée chez aucun des porcelets. Les animaux vaccinés et les témoins n'ont pas différé quant aux lésions histologiques et à la quantité d'antigènes de PCV2 dans les noeuds lymphatiques. Cette étude a démontré quelques évidences d'amorçage de l'immunité chez les jeunes porcelets en présence d'anticorps maternels. Des études supplémentaires sont recommandées afin de déterminer la concentration optimale d'antigène de PCV2 et un adjuvant adéquat pour le vaccin dans le but d'atteindre le plein potentiel de la stratégie d'induire une immunité acquise chez les jeunes porcelets possédant des anticorps maternels.(Traduit par Docteur Serge Messier).


Subject(s)
Circoviridae Infections/veterinary , Circovirus/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Viral Vaccines/administration & dosage , Adaptive Immunity/immunology , Animals , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Immunity, Maternally-Acquired/immunology , Immunohistochemistry/veterinary , Lymph Nodes/virology , Swine , Swine Diseases/immunology , Vaccination/veterinary , Viral Vaccines/immunology
20.
Can J Vet Res ; 74(3): 237-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20885851

ABSTRACT

The objective of this study was to improve the visual localization of urease activity of Helicobacter pylori-like organisms (HPLO) on swine gastric mucosa by in vitro optimization of the urea concentration and pH indicator of a urease test reagent. Five 21-day-old conventional pigs were infected orally with HPLO (3 pigs) or Brucella broth alone (2 pigs). At 17 d after infection the pigs were euthanized and their stomachs excised and tested for HPLO by a modified urease test formulation sprayed onto the gastric mucosa, as well as confirmatory culture and isolation of HPLO from urease-positive sites. This study showed improved detection of HPLO in porcine gastric mucosa with the use of a modified urease test formulation containing 5% urea and the pH indicator bromocresol purple compared with the use of a conventional formulation of 2% urea and phenol red. This test can readily be applied to achieve a presumptive diagnosis of HPLO in cases of gastritis or gastric esophageal ulceration in pigs.


Subject(s)
Gastric Mucosa/microbiology , Helicobacter Infections/veterinary , Helicobacter/isolation & purification , Swine Diseases/microbiology , Urease/metabolism , Animals , Helicobacter/classification , Helicobacter Infections/microbiology , Species Specificity , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...