Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
J Virol ; 98(3): e0120623, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38305154

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with greater transmissibility or immune evasion properties has jeopardized the existing vaccine and antibody-based countermeasures. Here, we evaluated the efficacy of boosting pre-immune hamsters with protein nanoparticle vaccines (Novavax, Inc.) containing recombinant Prototype (Wuhan-1) or BA.5 S proteins against a challenge with the Omicron BA.5 variant of SARS-CoV-2. Serum antibody binding and neutralization titers were quantified before challenge, and viral loads were measured 3 days after challenge. Boosting with Prototype or BA.5 vaccine induced similar antibody binding responses against ancestral Wuhan-1 or BA.5 S proteins, and neutralizing activity of Omicron BA.1 and BA.5 variants. One and three months after vaccine boosting, hamsters were challenged with the Omicron BA.5 variant. Prototype and BA.5 vaccine-boosted hamsters had reduced viral infection in the nasal washes, nasal turbinates, and lungs compared to unvaccinated animals. Although no significant differences in virus load were detected between the Prototype and BA.5 vaccine-boosted animals, fewer breakthrough infections were detected in the BA.5-vaccinated hamsters. Thus, immunity induced by Prototype or BA.5 S protein nanoparticle vaccine boosting can protect against the Omicron BA.5 variant in the Syrian hamster model. IMPORTANCE: As SARS-CoV-2 continues to evolve, there may be a need to update the vaccines to match the newly emerging variants. Here, we compared the protective efficacy of the updated BA.5 and the original Wuhan-1 COVID-19 vaccine against a challenge with the BA.5 Omicron variant of SARS-CoV-2 in hamsters. Both vaccines induced similar levels of neutralizing antibodies against multiple variants of SARS-CoV-2. One and three months after the final immunization, hamsters were challenged with BA.5. No differences in protection against the BA.5 variant virus were observed between the two vaccines, although fewer breakthrough infections were detected in the BA.5-vaccinated hamsters. Together, our data show that both protein nanoparticle vaccines are effective against the BA.5 variant of SARS-CoV-2 but given the increased number of breakthrough infections and continued evolution, it is important to update the COVID-19 vaccine for long-term protection.


Subject(s)
COVID-19 Vaccines , Nanovaccines , SARS-CoV-2 , Animals , Cricetinae , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Breakthrough Infections/immunology , Breakthrough Infections/prevention & control , Breakthrough Infections/virology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/immunology , Mesocricetus/immunology , Mesocricetus/virology , Nanovaccines/immunology , SARS-CoV-2/immunology , Immunization, Secondary , Viral Load
2.
J Virol ; 96(15): e0076522, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35862681

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza A virus (IAV) represent two highly transmissible airborne pathogens with pandemic capabilities. Although these viruses belong to separate virus families-SARS-CoV-2 is a member of the family Coronaviridae, while IAV is a member of the family Orthomyxoviridae-both have shown zoonotic potential, with significant animal reservoirs in species in close contact with humans. The two viruses are similar in their capacity to infect human airways, and coinfections resulting in significant morbidity and mortality have been documented. Here, we investigate the interaction between SARS-CoV-2 USA-WA1/2020 and influenza H1N1 A/California/04/2009 virus during coinfection. Competition assays in vitro were performed in susceptible cells that were either interferon type I/III (IFN-I/-III) nonresponsive or IFN-I/-III responsive, in addition to an in vivo golden hamster model. We find that SARS-CoV-2 infection does not interfere with IAV biology in vivo, regardless of timing between the infections. In contrast, we observe a significant loss of SARS-CoV-2 replication following IAV infection. The latter phenotype correlates with increased levels of IFN-I/-III and immune priming that interferes with the kinetics of SARS-CoV-2 replication. Together, these data suggest that cocirculation of SARS-CoV-2 and IAV is unlikely to result in increased severity of disease. IMPORTANCE The human population now has two circulating respiratory RNA viruses with high pandemic potential, namely, SARS-CoV-2 and influenza A virus. As both viruses infect the airways and can result in significant morbidity and mortality, it is imperative that we also understand the consequences of getting coinfected. Here, we demonstrate that the host response to influenza A virus uniquely interferes with SARS-CoV-2 biology although the inverse relationship is not evident. Overall, we find that the host response to both viruses is comparable to that to SARS-CoV-2 infection alone.


Subject(s)
COVID-19 , Coinfection , Cross-Priming , Influenza A Virus, H1N1 Subtype , Influenza, Human , SARS-CoV-2 , Virus Replication , Animals , COVID-19/immunology , COVID-19/mortality , COVID-19/virology , Coinfection/immunology , Coinfection/virology , Cross-Priming/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Interferons/immunology , Mesocricetus/immunology , Mesocricetus/virology , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , Virus Replication/immunology
3.
Sci Rep ; 12(1): 3890, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35273234

ABSTRACT

The new outbreak of coronavirus disease 2019 (COVID-19) has infected and caused the death of millions of people worldwide. Intensive efforts are underway around the world to establish effective treatments. Immunoglobulin from immunized animals or plasma from convalescent patients might constitute a specific treatment to guarantee the neutralization of the virus in the early stages of infection, especially in patients with risk factors and a high probability of progressing to severe disease. Worldwide, a few clinical trials using anti-SARS-CoV-2 immunoglobulins from horses immunized with the entire spike protein or fragments of it in the treatment of patients with COVID-19 are underway. Here, we describe the development of an anti-SARS-CoV-2 equine F(ab')2 immunoglobulin using a newly developed SARS-CoV-2 viral antigen that was purified and inactivated by radiation. Cell-based and preclinical assays showed that the F(ab')2 immunoglobulin successfully neutralizes the virus, is safe in animal models, and reduces the severity of the disease in a hamster model of SARS-CoV-2 infection and disease.


Subject(s)
COVID-19/therapy , Immunoglobulins/therapeutic use , Receptors, Immunologic/therapeutic use , SARS-CoV-2/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Horses/immunology , Humans , Immunoglobulins/immunology , Immunoglobulins/isolation & purification , Male , Mesocricetus/immunology , Plasmapheresis/veterinary , Receptors, Immunologic/immunology
4.
Vopr Virusol ; 66(6): 442-451, 2022 01 08.
Article in Russian | MEDLINE | ID: mdl-35019251

ABSTRACT

INTRODUCTION: Verification of histological changes in respiratory system using Syrian (golden) hamsters (Mesocricetus auratus) as experimental model is an important task for preclinical studies of drugs intended for prevention and treatment of the novel coronavirus infection COVID-19.The aim of this work was to study pathological changes of pulmonary tissue in SARS-CoV-2 (Coronaviridae: Coronavirinae: Betacoronavirus; Sarbecovirus) experimental infection in Syrian hamsters. MATERIAL AND METHODS: Male Syrian hamsters weighting 80-100 g were infected by intranasal administration of culture SARS-CoV-2 at dose 4 × 104 TCID50/ml (TCID is tissue culture infectious dose). Animals were euthanatized on 3, 7 and 14 days after infection, with gravimetric registration. The viral load in lungs was measured using the polymerase chain reaction (PCR). Right lung and trachea tissues were stained with hematoxylin-eosin and according to Mallory. RESULTS AND DISCUSSION: The highest viral replicative activity in lungs was determined 3 days after the infection. After 7 days, on a background of the decrease of the viral load in lungs, a pathologically significant increase of the organ's gravimetric parameters was observed. Within 3 to 14 days post-infection, the lung histologic pattern had been showing the development of inflammation with a succession of infiltrative-proliferative, edematousmacrophagal and fibroblastic changes. It was found that initial changes in respiratory epithelium can proceed without paranecrotic interstitial inflammation, while in the formation of multiple lung parenchyma lesions, damage to the epithelium of bronchioles and acinar ducts can be secondary. The appearance of epithelioid large-cell metaplastic epithelium, forming pseudoacinar structures, was noted as a pathomorphological feature specific to SARS-CoV-2 infection in Syrian hamsters. CONCLUSION: As a result of the study, the specific features of the pathology of the respiratory system in SARSCoV-2 infected Syrian hamsters were described. These findings are of practical importance as reference data that can be used for preclinical studies to assess the effectiveness of vaccines and potential drugs.


Subject(s)
COVID-19 , Lung/pathology , Lung/virology , Mesocricetus , Animals , Coronaviridae , Disease Models, Animal , Inflammation , Male , Mesocricetus/immunology , Mesocricetus/virology , SARS-CoV-2
5.
Viruses ; 13(8)2021 08 19.
Article in English | MEDLINE | ID: mdl-34452513

ABSTRACT

The recent emergence of SARS-CoV-2 in humans from a yet unidentified animal reservoir and the capacity of the virus to naturally infect pets, farmed animals and potentially wild animals has highlighted the need for serological surveillance tools. In this study, the luciferase immunoprecipitation systems (LIPS), employing the spike (S) and nucleocapsid proteins (N) of SARS-CoV-2, was used to examine the suitability of the assay for antibody detection in different animal species. Sera from SARS-CoV-2 naturally-infected mink (n = 77), SARS-CoV-2 experimentally-infected ferrets, fruit bats and hamsters and a rabbit vaccinated with a purified spike protein were examined for antibodies using the SARS-CoV-2 N and/or S proteins. From comparison with the known neutralization status of the serum samples, statistical analyses including calculation of the Spearman rank-order-correlation coefficient and Cohen's kappa agreement were used to interpret the antibody results and diagnostic performance. The LIPS immunoassay robustly detected the presence of viral antibodies in naturally infected SARS-CoV-2 mink, experimentally infected ferrets, fruit bats and hamsters as well as in an immunized rabbit. For the SARS-CoV-2-LIPS-S assay, there was a good level of discrimination between the positive and negative samples for each of the five species tested with 100% agreement with the virus neutralization results. In contrast, the SARS-CoV-2-LIPS-N assay did not consistently differentiate between SARS-CoV-2 positive and negative sera. This study demonstrates the suitability of the SARS-CoV-2-LIPS-S assay for the sero-surveillance of SARS-CoV-2 infection in a range of animal species.


Subject(s)
Antibodies, Viral/blood , COVID-19/veterinary , Mink/immunology , SARS-CoV-2/immunology , Animals , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/immunology , COVID-19 Serological Testing , Chiroptera/immunology , Coronavirus Nucleocapsid Proteins/immunology , Epidemiological Monitoring , Ferrets/immunology , Immunoprecipitation , Mesocricetus/immunology , Phosphoproteins/immunology , Rabbits/immunology , Seroepidemiologic Studies , Spike Glycoprotein, Coronavirus/immunology
6.
Nature ; 597(7874): 103-108, 2021 09.
Article in English | MEDLINE | ID: mdl-34280951

ABSTRACT

The recent emergence of SARS-CoV-2 variants of concern1-10 and the recurrent spillovers of coronaviruses11,12 into the human population highlight the need for broadly neutralizing antibodies that are not affected by the ongoing antigenic drift and that can prevent or treat future zoonotic infections. Here we describe a human monoclonal antibody designated S2X259, which recognizes a highly conserved cryptic epitope of the receptor-binding domain and cross-reacts with spikes from all clades of sarbecovirus. S2X259 broadly neutralizes spike-mediated cell entry of SARS-CoV-2, including variants of concern (B.1.1.7, B.1.351, P.1, and B.1.427/B.1.429), as well as a wide spectrum of human and potentially zoonotic sarbecoviruses through inhibition of angiotensin-converting enzyme 2 (ACE2) binding to the receptor-binding domain. Furthermore, deep-mutational scanning and in vitro escape selection experiments demonstrate that S2X259 possesses an escape profile that is limited to a single substitution, G504D. We show that prophylactic and therapeutic administration of S2X259 protects Syrian hamsters (Mesocricetus auratus) against challenge with the prototypic SARS-CoV-2 and the B.1.351 variant of concern, which suggests that this monoclonal antibody is a promising candidate for the prevention and treatment of emergent variants and zoonotic infections. Our data reveal a key antigenic site that is targeted by broadly neutralizing antibodies and will guide the design of vaccines that are effective against all sarbecoviruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/therapeutic use , COVID-19/prevention & control , SARS-CoV-2/classification , SARS-CoV-2/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , Antibodies, Viral/therapeutic use , Broadly Neutralizing Antibodies/chemistry , COVID-19/immunology , COVID-19/virology , Cross Reactions/immunology , Disease Models, Animal , Female , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Mesocricetus/immunology , Mesocricetus/virology , Mutation , Neutralization Tests , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , Viral Zoonoses/immunology , Viral Zoonoses/prevention & control , Viral Zoonoses/virology
7.
Nature ; 596(7870): 103-108, 2021 08.
Article in English | MEDLINE | ID: mdl-34153975

ABSTRACT

Rapidly emerging SARS-CoV-2 variants jeopardize antibody-based countermeasures. Although cell culture experiments have demonstrated a loss of potency of several anti-spike neutralizing antibodies against variant strains of SARS-CoV-21-3, the in vivo importance of these results remains uncertain. Here we report the in vitro and in vivo activity of a panel of monoclonal antibodies (mAbs), which correspond to many in advanced clinical development by Vir Biotechnology, AbbVie, AstraZeneca, Regeneron and Lilly, against SARS-CoV-2 variant viruses. Although some individual mAbs showed reduced or abrogated neutralizing activity in cell culture against B.1.351, B.1.1.28, B.1.617.1 and B.1.526 viruses with mutations at residue E484 of the spike protein, low prophylactic doses of mAb combinations protected against infection by many variants in K18-hACE2 transgenic mice, 129S2 immunocompetent mice and hamsters, without the emergence of resistance. Exceptions were LY-CoV555 monotherapy and LY-CoV555 and LY-CoV016 combination therapy, both of which lost all protective activity, and the combination of AbbVie 2B04 and 47D11, which showed a partial loss of activity. When administered after infection, higher doses of several mAb cocktails protected in vivo against viruses with a B.1.351 spike gene. Therefore, many-but not all-of the antibody products with Emergency Use Authorization should retain substantial efficacy against the prevailing variant strains of SARS-CoV-2.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/pharmacology , Antibodies, Viral/therapeutic use , COVID-19/virology , Neutralization Tests , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/immunology , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , Chlorocebus aethiops , Female , Humans , Male , Mesocricetus/immunology , Mesocricetus/virology , Mice , Mice, Transgenic , Post-Exposure Prophylaxis , Pre-Exposure Prophylaxis , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells
8.
Dev Comp Immunol ; 119: 104024, 2021 06.
Article in English | MEDLINE | ID: mdl-33503449

ABSTRACT

Hibernation consists of alternating periods of reduced metabolism (torpor) with brief periods of metabolism similar to summer euthermia (arousal). The function of the innate immune system is reduced during hibernation, of which the underlying mechanisms are incompletely understood. Here, we studied neutrophil functionality during hibernation in Syrian hamsters. The inflammatory response to LPS-induced endotoxemia is inhibited in hibernation, partly mediated by reduced IL-6 production in early arousal. Furthermore, neutrophil pathogen binding, phagocytosis and oxidative burst is profoundly reduced in early arousal. Functionality of both summer and early arousal neutrophils was repressed in plasma from early arousal and mixed plasma from early arousal and summer euthermic, but restored by summer euthermic plasma, signifying that a plasma factor in early arousal inhibits TLR-recognition. Identification of the inhibiting factor may offer a target to modulate neutrophil function with relevance to (auto-)inflammatory diseases.


Subject(s)
Hibernation/immunology , Immunity, Innate/immunology , Mesocricetus/immunology , Neutrophils/immunology , Seasons , Acute-Phase Proteins/immunology , Animals , Arousal/genetics , Arousal/physiology , Carrier Proteins/blood , Carrier Proteins/immunology , Cytokines/immunology , Cytokines/metabolism , Gene Expression/immunology , Hibernation/genetics , Hibernation/physiology , Immunity, Innate/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharide Receptors/blood , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Membrane Glycoproteins/blood , Membrane Glycoproteins/immunology , Mesocricetus/genetics , Mesocricetus/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Neutrophils/metabolism , Neutrophils/physiology , Phagocytosis/immunology , Respiratory Burst/immunology , Respiratory Burst/physiology , Time Factors
9.
Front Immunol ; 12: 785349, 2021.
Article in English | MEDLINE | ID: mdl-35095861

ABSTRACT

SARS-CoV-2 infections present a tremendous threat to public health. Safe and efficacious vaccines are the most effective means in preventing the infections. A variety of vaccines have demonstrated excellent efficacy and safety around the globe. Yet, development of alternative forms of vaccines remains beneficial, particularly those with simpler production processes, less stringent storage conditions, and the capability of being used in heterologous prime/boost regimens which have shown improved efficacy against many diseases. Here we reported a novel DNA vaccine comprised of the SARS-CoV-2 spike protein fused with CD40 ligand (CD40L) serving as both a targeting ligand and molecular adjuvant. A single intramuscular injection in Syrian hamsters induced significant neutralizing antibodies 3-weeks after vaccination, with a boost substantially improving immune responses. Moreover, the vaccine also reduced weight loss and suppressed viral replication in the lungs and nasal turbinates of challenged animals. Finally, the incorporation of CD40L into the DNA vaccine was shown to reduce lung pathology more effectively than the DNA vaccine devoid of CD40L. These results collectively indicate that this DNA vaccine candidate could be further explored because of its efficacy and known safety profile.


Subject(s)
CD40 Ligand/immunology , COVID-19/immunology , Mesocricetus/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/virology , Cell Line , Female , HEK293 Cells , Humans , Lung/immunology , Lung/virology , Mesocricetus/virology , Models, Animal , Vaccination/methods , Vaccines, Inactivated/immunology
10.
Nature ; 590(7845): 320-325, 2021 02.
Article in English | MEDLINE | ID: mdl-33260195

ABSTRACT

The expanding pandemic of coronavirus disease 2019 (COVID-19) requires the development of safe, efficacious and fast-acting vaccines. Several vaccine platforms are being leveraged for a rapid emergency response1. Here we describe the development of a candidate vaccine (YF-S0) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses live-attenuated yellow fever 17D (YF17D) vaccine as a vector to express a noncleavable prefusion form of the SARS-CoV-2 spike antigen. We assess vaccine safety, immunogenicity and efficacy in several animal models. YF-S0 has an excellent safety profile and induces high levels of SARS-CoV-2 neutralizing antibodies in hamsters (Mesocricetus auratus), mice (Mus musculus) and cynomolgus macaques (Macaca fascicularis), and-concomitantly-protective immunity against yellow fever virus. Humoral immunity is complemented by a cellular immune response with favourable T helper 1 polarization, as profiled in mice. In a hamster model2 and in macaques, YF-S0 prevents infection with SARS-CoV-2. Moreover, a single dose conferred protection from lung disease in most of the vaccinated hamsters within as little as 10 days. Taken together, the quality of the immune responses triggered and the rapid kinetics by which protective immunity can be attained after a single dose warrant further development of this potent SARS-CoV-2 vaccine candidate.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/immunology , COVID-19/prevention & control , Genetic Vectors/genetics , SARS-CoV-2/immunology , Vaccines, Attenuated/immunology , Yellow Fever Vaccine/genetics , Animals , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/genetics , Cricetinae , Disease Models, Animal , Female , Glycosylation , Macaca fascicularis/genetics , Macaca fascicularis/immunology , Macaca fascicularis/virology , Male , Mesocricetus/genetics , Mesocricetus/immunology , Mesocricetus/virology , Mice , Safety , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics
11.
Methods Mol Biol ; 2134: 243-255, 2020.
Article in English | MEDLINE | ID: mdl-32632875

ABSTRACT

Experimental infections greatly contribute to further deepen our knowledge of infectious diseases. In the case of leptospirosis, hamsters as well as gerbils and guinea pigs have been used as animal models of acute leptospirosis in studying the pathophysiology of the disease. Here we describe a typical Leptospira infection using golden Syrian hamsters. We will also present techniques we use to study the resulting bacterial burden and gene expression patterns in the host in order to decipher the innate immune response to leptospirosis.


Subject(s)
Immunity, Innate/immunology , Leptospirosis/immunology , Mesocricetus/immunology , Mesocricetus/microbiology , Animals , Disease Models, Animal , Female , Gene Expression/immunology , Leptospira/immunology , Male
12.
Methods Mol Biol ; 2134: 257-270, 2020.
Article in English | MEDLINE | ID: mdl-32632876

ABSTRACT

Leptospirosis is a major public health problem, especially in developing countries. Current vaccine studies focus on identifying Leptospira proteins that elicit protective immunity. Here, we describe a method to assess recombinant proteins for their ability to protect hamsters from fatal infection against Leptospira and to provide sterilizing immunity.


Subject(s)
Bacterial Vaccines/immunology , Leptospirosis/immunology , Mesocricetus/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Female , Leptospira/immunology , Leptospirosis/microbiology , Mesocricetus/microbiology , Recombinant Proteins/immunology
13.
Nature ; 583(7818): 834-838, 2020 07.
Article in English | MEDLINE | ID: mdl-32408338

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus with high nucleotide identity to SARS-CoV and to SARS-related coronaviruses that have been detected in horseshoe bats, has spread across the world and had a global effect on healthcare systems and economies1,2. A suitable small animal model is needed to support the development of vaccines and therapies. Here we report the pathogenesis and transmissibility of SARS-CoV-2 in golden (Syrian) hamsters (Mesocricetus auratus). Immunohistochemistry assay demonstrated the presence of viral antigens in nasal mucosa, bronchial epithelial cells and areas of lung consolidation on days 2 and 5 after inoculation with SARS-CoV-2, followed by rapid viral clearance and pneumocyte hyperplasia at 7 days after inoculation. We also found viral antigens in epithelial cells of the duodenum, and detected viral RNA in faeces. Notably, SARS-CoV-2 was transmitted efficiently from inoculated hamsters to naive hamsters by direct contact and via aerosols. Transmission via fomites in soiled cages was not as efficient. Although viral RNA was continuously detected in the nasal washes of inoculated hamsters for 14 days, the communicable period was short and correlated with the detection of infectious virus but not viral RNA. Inoculated and naturally infected hamsters showed apparent weight loss on days 6-7 post-inoculation or post-contact; all hamsters returned to their original weight within 14 days and developed neutralizing antibodies. Our results suggest that features associated with SARS-CoV-2 infection in golden hamsters resemble those found in humans with mild SARS-CoV-2 infections.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/transmission , Coronavirus Infections/virology , Disease Models, Animal , Lung/pathology , Lung/virology , Mesocricetus/virology , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Aerosols , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Antigens, Viral/isolation & purification , Antigens, Viral/metabolism , Betacoronavirus/immunology , Betacoronavirus/isolation & purification , Betacoronavirus/metabolism , Bronchi/pathology , Bronchi/virology , COVID-19 , Coronavirus Infections/immunology , Duodenum/virology , Fomites/virology , Housing, Animal , Kidney/virology , Male , Mesocricetus/immunology , Nasal Mucosa/virology , Pandemics , Pneumonia, Viral/immunology , RNA, Viral/analysis , SARS-CoV-2 , Viral Load , Weight Loss
15.
J Infect Dis ; 212 Suppl 2: S271-6, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25948862

ABSTRACT

Most ebolaviruses can cause severe disease in humans and other primates, with high case fatality rates during human outbreaks. Although these viruses have been studied for almost 4 decades, little is know regarding the mechanisms by which they cause disease and what is important for protection or treatment after infection. Because of the sporadic nature of the outbreaks and difficulties accessing the populations affected by ebolaviruses, little is also known about what constitutes an appropriate immune response to infection in humans that survive infection. Such knowledge would allow a targeted approach to therapies. In contrast to humans, rodents are protected from disease on infection with ebolaviruses, although adapted versions of some of the viruses are lethal in mice, hamsters and guinea pigs. Using the recently described hamster model, along with T-cell depletion strategies, we show that CD4(+) T cells are required for natural immunity to Ebola virus infection and that CD4-dependent antibody responses are required for immunity in this model.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Immunity, Innate/immunology , Mesocricetus/immunology , Mesocricetus/virology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cricetinae , Female , Hemorrhagic Fever, Ebola/virology , Lymphocyte Depletion/methods
16.
Int Arch Allergy Immunol ; 166(1): 30-40, 2015.
Article in English | MEDLINE | ID: mdl-25765367

ABSTRACT

BACKGROUND: An increasing number of asthma cases upon exposure to hamsters and anaphylactic reactions following hamster bites are being reported, but the allergens responsible are still poorly characterized. In the Golden hamster, male-specific submaxillary gland protein (MSP), a lipocalin expressed in a sex- and tissue-specific manner in the submaxillary and lacrimal glands, is secreted in the saliva, tears and urine. The purpose of this study was to determine if MSP is an allergen, to identify IgE-reactive proteins of different hamster species and to analyse potential cross-reactivities. METHODS: Fur extracts were prepared from four hamster species. Hamster-allergic patients were selected based on a history of positive IgE-test to hamster epithelium. The IgE-reactivity of patients' sera was investigated by means of immunoblot and ELISA. IgE-reactive proteins in fur extracts and the submaxillary gland were identified using anti-MSP antibodies, Edman sequencing or mass spectrometry. MSP was purified from Golden hamster and recombinant MSP was expressed in E. coli. RESULTS: Four patients had IgE-antibodies against 20.5-kDa and 24-kDa proteins of Golden hamster fur extract, which were identified as MSP. IgE-reactive MSP-like proteins were detected in European hamster fur extract. Three patient sera showed IgE-reactive bands at 17-21 kDa in Siberian and Roborovski hamster fur extracts. These proteins were identified as two closely related lipocalins. Immunoblot inhibition experiments showed that they are cross-reactive and are different from MSP. CONCLUSION: MSP lipocalin of the Golden hamster was identified as an allergen, and it is different from the cross-reactive lipocalin allergens of Siberian and Roborovski hamsters. Our findings highlight the need for specific tools for the in vitro and in vivo diagnosis of allergy to different hamster species.


Subject(s)
Allergens/immunology , Hair/immunology , Hypersensitivity, Immediate/immunology , Lipocalins/immunology , Submandibular Gland/immunology , Adult , Allergens/chemistry , Allergens/genetics , Animals , Cricetinae , Cricetulus/immunology , Cross Reactions , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression , Hair/chemistry , Humans , Hypersensitivity, Immediate/genetics , Hypersensitivity, Immediate/pathology , Immunoglobulin E/immunology , Lipocalins/chemistry , Lipocalins/genetics , Male , Mesocricetus/immunology , Middle Aged , Phodopus/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sex Factors , Species Specificity , Submandibular Gland/chemistry
17.
Rio de Janeiro; s.n; 2015. ix,81 p. ilus, tab, graf, mapas.
Thesis in Portuguese | LILACS | ID: lil-774226

ABSTRACT

Estudos anteriores no modelo murino demonstraram a possibilidade deindução de proteção através da imunização por via mucosa, tanto com antígenosbrutos (lisado total de promastigotas de Leishmania amazonensis - LaAg) comoatravés de vacina de DNA (DNA plasmideal com o gene que codifica a proteínaLACK - LACK DNA), contra a leishmaniose cutânea (LC) causada por L.amazonensis, e a leishmaniose visceral causada por L. infantum. No entanto,estudos de vacinação contra as espécies do subgênero Viannia, principaisresponsáveis pela LC nas Américas, são dificultados devido à falta de modelosexperimentais de fácil manuseio que reproduzam a doença humana. Recentementefoi demonstrado pelo nosso grupo que o hamster dourado é um modelo adequadopara o estudo da imunopatogênese da leishmaniose cutânea causada por L. (V.)braziliensis e que o antígeno LaAg administrado por via intranasal induziu proteçãocontra a infecção por L. (V.) braziliensis no modelo. Entretanto, as abordagensimunológicas e moleculares para os estudos no modelo hamster são limitadas pelapouca disponibilidade de insumos comerciais disponíveis. Nesse trabalho,padronizamos um ensaio por RT-qPCR para avaliação de marcadores molecularesem pele de hamsters infectados por L. braziliensis, pela expressão gênica de IFN-gama,TGF-beta, TNF, IL-10, IL-4, IL-6, iNOS e arginase, e investigamos o efeito protetor daimunização intranasal com a vacina gênica LACK DNA, administrada em protocolosprime-boost homólogo (50mig/dose) e heterólogo (LACK DNA-50mig / LaAg-10mig) nainfecção por L. braziliensis...


Previous studies in murine models demonstrated the possibility to induceprotection by mucosal immunization with crude antigens (total lysate of Leishmaniaamazonensis promastigotes - LaAg) as well as through DNA vaccine (plasmid DNAwith the gene encoding LACK protein - LACK DNA) against cutaneous leishmaniasis(CL) caused by L. amazonensis and visceral leishmaniasis caused by L. infantum.However, vaccination studies against Viannia subgenus (the main cause for CL inAmericas) are hampered due to the lack of experimental models which are easilyexecuted to mimic this disease as it occurs in humans. Recently, it was demonstratedby our group that the golden hamster is an appropriate model to studyimmunopathological aspects of cutaneous leishmaniasis caused by L. (V.)braziliensis. Furthermore, it was also demonstrated that LaAg antigen administeredintranasally induces protection against L. (V.) braziliensis infection in the model.However, immunological and molecular approaches for studies in hamster modelsare limited by the low availability of commercial products. In this study, westandardized an assay by RT-qPCR for assessment of molecular markers in the skinof hamsters infected by L. braziliensis, through the relative gene expressionquantification of IFN-gama, TGF-beta, TNF, IL-10, IL-4, IL-6, iNOS and arginase, and theprotective effect of intranasal immunization with LACK DNA vaccine (administered inhomologous prime-boost protocol 50 mcg/dose) and heterologous prime-boost(LACK DNA 50 mcg in first dose/LaAg-10mg in second dose), against L. braziliensisinfection. The results demonstrated that immunization with LACK DNA homologousprime boost did not induce protection against L. braziliensis infection in hamstermodel since there was no significant difference in lesion size, in parasite load, andIgG and IgG 2 anti-Leishmania levels on day 110 after infection, compared with thecontrol groups (PBS and DNA)...


Subject(s)
Cricetinae , Leishmaniasis Vaccines , Leishmania braziliensis/immunology , Parasite Load , Mesocricetus/immunology , Reverse Transcriptase Polymerase Chain Reaction
18.
BMC Immunol ; 15: 38, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25424735

ABSTRACT

BACKGROUND: The Syrian golden hamster (Mesocricetus aureus) has been used as a model to study infections caused by a number of human pathogens. Studies of immunopathogenesis in hamster infection models are challenging because of the limited availability of reagents needed to define cellular and molecular determinants. RESULTS: We sequenced a hamster cDNA library and developed a first-generation custom cDNA microarray that included 5131 unique cDNAs enriched for immune response genes. We used this microarray to interrogate the hamster spleen response to Leishmania donovani, an intracellular protozoan that causes visceral leishmaniasis. The hamster model of visceral leishmaniasis is of particular interest because it recapitulates clinical and immunopathological features of human disease, including cachexia, massive splenomegaly, pancytopenia, immunosuppression, and ultimately death. In the microarray a differentially expressed transcript was identified as having at least a 2-fold change in expression between uninfected and infected groups and a False Discovery Rate of <5%. Following a relatively silent early phase of infection (at 7 and 14 days post-infection only 8 and 24 genes, respectively, were differentially expressed), there was dramatic upregulation of inflammatory and immune-related genes in the spleen (708 differentially expressed genes were evident at 28 days post-infection). The differentially expressed transcripts included genes involved in inflammation, immunity, and immune cell trafficking. Of particular interest there was concomitant upregulation of the IFN-γ and interleukin (IL)-4 signaling pathways, with increased expression of a battery of IFN-γ- and IL-4-responsive genes. The latter included genes characteristic of alternatively activated macrophages. CONCLUSIONS: Transcriptional profiling was accomplished in the Syrian golden hamster, for which a fully annotated genome is not available. In the hamster model of visceral leishmaniasis, a robust and functional IFN-γ response did not restrain parasite load and progression of disease. This supports the accumulating evidence that macrophages are ineffectively activated to kill the parasite. The concomitant expression of IL-4/IL-13 and their downstream target genes, some of which were characteristic of alternative macrophage activation, are likely to contribute to this. Further dissection of mechanisms that lead to polarization of macrophages toward a permissive state is needed to fully understand the pathogenesis of visceral leishmaniasis.


Subject(s)
Cytokines/genetics , Gene Expression Profiling , Gene Expression Regulation , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/immunology , Spleen/metabolism , Spleen/parasitology , Animals , Cluster Analysis , Cricetinae , Cytokines/metabolism , DNA, Complementary/genetics , Disease Progression , Expressed Sequence Tags , Gene Ontology , Humans , Interferon-gamma/metabolism , Leishmania donovani/immunology , Mesocricetus/immunology , Mesocricetus/parasitology , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Spleen/pathology , Up-Regulation/genetics
19.
PLoS One ; 9(6): e97911, 2014.
Article in English | MEDLINE | ID: mdl-24926878

ABSTRACT

The present study includes cloning and expression of recombinant Leishmania donovani histone proteins (rLdH2B, rLdH3, rLdH2A and rLdH4), assessment of their immunogenicity in Leishmania infected cured patients/endemic contacts as well as in cured hamsters and finally evaluation of their prophylactic efficacy in hamsters against L. donovani challenge. All recombinant proteins were expressed and purified from the heterologous bacterial host system. Leishmania infected cured patients/endemic contacts as well as cured hamsters exhibited significantly higher proliferative responses to individual recombinant histones and their pooled combination (rLdH2B+rLdH3+rLdH2A+rLdH4) than those of L.donovani infected hosts. The L.donovani soluble antigens (SLD) stimulated PBMCs of cured/exposed and Leishmania patients to produce a mixed Thl/Th2-type cytokine profile, whereas rLdH2B, rLdH3, rLdH2A, rLdH4 and pooled combination (rLdH2-4) stimulated the production of Th1 cytokines IFN-γ, IL-12 and TNF-α but not Th2 cytokines IL-4 or IL-10. The immunogenicity of these histone proteins along with their combination was also checked in cured hamsters where they stimulated higher lymphoproliferation and Nitric oxide production in lymphocytes of cured hamsters than that of infected controls. Moreover, significantly increased IgG2 response, an indicative of cell mediated immunity, was observed in cured hamsters against these individual proteins and their combination as compared to infected hamsters. Further, it was demonstrated that rLdH2B, rLdH3, rLdH2A and rLdH4 and pooled combination were able to provide considerable protection for hamsters against L. donovani challenge. The efficacy was supported by the increased inducible Nitric Oxide Synthase (iNOS) mRNA transcripts and Th1-type cytokines--IFN-γ, IL-12 and TNF-α and down-regulation of IL-4, IL-10 and TGF-ß. Hence, it is inferred that pooled rLdH2-4 elicits Thl-type of immune responses exclusively and confer considerable protection against experimental Visceral Leishmaniasis.


Subject(s)
Antigens, Protozoan/immunology , Histones/metabolism , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/prevention & control , Mesocricetus/parasitology , Adolescent , Adult , Animals , Cricetinae , Cytokines/genetics , Cytokines/metabolism , Female , Histones/genetics , Humans , Immunization/methods , Leishmania donovani/isolation & purification , Leishmania donovani/metabolism , Leishmaniasis, Visceral/veterinary , Male , Mesocricetus/immunology , Middle Aged , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Young Adult
20.
PLoS One ; 9(2): e89412, 2014.
Article in English | MEDLINE | ID: mdl-24586759

ABSTRACT

Leishmaniasis remains one of the world's most devastating neglected tropical diseases. It mainly affects developing countries, where it often co-exists with chronic malnutrition, one of the main risk factors for developing the disease. Few studies have been published, however, on the relationship between leishmaniasis progression and malnutrition. The present paper reports the influence of protein malnutrition on the immune response and visceral disease development in adult hamsters infected with Leishmania infantum fed either standard or low protein diets. The low protein diet induced severe malnutrition in these animals, and upon infection with L. infantum 33% had severe visceral leishmaniasis compared to only 8% of animals fed the standard diet. The infected, malnourished animals showed notable leukocyte depletion, mild specific antibody responses, impairment of lymphoproliferation, presence of parasites in blood (16.67% of the hamsters) and significant increase of the splenic parasite burden. Animals fed standard diet suffered agranulocytosis and monocytopenia, but showed stronger specific immune responses and had lower parasite loads than their malnourished counterparts. The present results show that protein malnutrition promotes visceral leishmaniasis and provide clues regarding the mechanisms underlying the impairment of the immune system.


Subject(s)
Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Malnutrition/immunology , Malnutrition/parasitology , Mesocricetus/immunology , Mesocricetus/parasitology , Proteins/metabolism , Animals , Chronic Disease , Cricetinae , Diet, Protein-Restricted/methods , Leishmania infantum/immunology , Leishmaniasis, Visceral/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/parasitology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/metabolism , Lymphoproliferative Disorders/parasitology , Male , Malnutrition/metabolism , Mesocricetus/metabolism , Neglected Diseases/immunology , Neglected Diseases/metabolism , Neglected Diseases/parasitology , Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL