Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Nature ; 586(7830): 572-577, 2020 10.
Article in English | MEDLINE | ID: mdl-32726802

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a respiratory disease called coronavirus disease 2019 (COVID-19), the spread of which has led to a pandemic. An effective preventive vaccine against this virus is urgently needed. As an essential step during infection, SARS-CoV-2 uses the receptor-binding domain (RBD) of the spike protein to engage with the receptor angiotensin-converting enzyme 2 (ACE2) on host cells1,2. Here we show that a recombinant vaccine that comprises residues 319-545 of the RBD of the spike protein induces a potent functional antibody response in immunized mice, rabbits and non-human primates (Macaca mulatta) as early as 7 or 14 days after the injection of a single vaccine dose. The sera from the immunized animals blocked the binding of the RBD to ACE2, which is expressed on the cell surface, and neutralized infection with a SARS-CoV-2 pseudovirus and live SARS-CoV-2 in vitro. Notably, vaccination also provided protection in non-human primates to an in vivo challenge with SARS-CoV-2. We found increased levels of RBD-specific antibodies in the sera of patients with COVID-19. We show that several immune pathways and CD4 T lymphocytes are involved in the induction of the vaccine antibody response. Our findings highlight the importance of the RBD domain in the design of SARS-CoV-2 vaccines and provide a rationale for the development of a protective vaccine through the induction of antibodies against the RBD domain.


Subject(s)
Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , COVID-19 , COVID-19 Vaccines , Humans , Macaca mulatta/immunology , Macaca mulatta/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Models, Molecular , Protein Domains , SARS-CoV-2 , Serum/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/immunology , Vaccination
2.
Nature ; 583(7818): 830-833, 2020 07.
Article in English | MEDLINE | ID: mdl-32380511

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019 (COVID-19), which has become a public health emergency of international concern1. Angiotensin-converting enzyme 2 (ACE2) is the cell-entry receptor for severe acute respiratory syndrome coronavirus (SARS-CoV)2. Here we infected transgenic mice that express human ACE2 (hereafter, hACE2 mice) with SARS-CoV-2 and studied the pathogenicity of the virus. We observed weight loss as well as virus replication in the lungs of hACE2 mice infected with SARS-CoV-2. The typical histopathology was interstitial pneumonia with infiltration of considerable numbers of macrophages and lymphocytes into the alveolar interstitium, and the accumulation of macrophages in alveolar cavities. We observed viral antigens in bronchial epithelial cells, macrophages and alveolar epithelia. These phenomena were not found in wild-type mice infected with SARS-CoV-2. Notably, we have confirmed the pathogenicity of SARS-CoV-2 in hACE2 mice. This mouse model of SARS-CoV-2 infection will be valuable for evaluating antiviral therapeutic agents and vaccines, as well as understanding the pathogenesis of COVID-19.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/pathology , Coronavirus Infections/virology , Lung/pathology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Transgenes , Angiotensin-Converting Enzyme 2 , Animals , Antigens, Viral/immunology , Antigens, Viral/metabolism , Betacoronavirus/immunology , Betacoronavirus/metabolism , Bronchi/pathology , Bronchi/virology , COVID-19 , Coronavirus Infections/immunology , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Immunoglobulin G/immunology , Lung/immunology , Lung/virology , Lymphocytes/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Male , Mice , Mice, Transgenic , Pandemics , Pneumonia, Viral/immunology , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , SARS-CoV-2 , Virus Replication , Weight Loss
3.
Proc Natl Acad Sci U S A ; 120(18): e2301775120, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37094153

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is an ongoing global health concern, and effective antiviral reagents are urgently needed. Traditional Chinese medicine theory-driven natural drug research and development (TCMT-NDRD) is a feasible method to address this issue as the traditional Chinese medicine formulae have been shown effective in the treatment of COVID-19. Huashi Baidu decoction (Q-14) is a clinically approved formula for COVID-19 therapy with antiviral and anti-inflammatory effects. Here, an integrative pharmacological strategy was applied to identify the antiviral and anti-inflammatory bioactive compounds from Q-14. Overall, a total of 343 chemical compounds were initially characterized, and 60 prototype compounds in Q-14 were subsequently traced in plasma using ultrahigh-performance liquid chromatography with quadrupole time-of-flight mass spectrometry. Among the 60 compounds, six compounds (magnolol, glycyrrhisoflavone, licoisoflavone A, emodin, echinatin, and quercetin) were identified showing a dose-dependent inhibition effect on the SARS-CoV-2 infection, including two inhibitors (echinatin and quercetin) of the main protease (Mpro), as well as two inhibitors (glycyrrhisoflavone and licoisoflavone A) of the RNA-dependent RNA polymerase (RdRp). Meanwhile, three anti-inflammatory components, including licochalcone B, echinatin, and glycyrrhisoflavone, were identified in a SARS-CoV-2-infected inflammatory cell model. In addition, glycyrrhisoflavone and licoisoflavone A also displayed strong inhibitory activities against cAMP-specific 3',5'-cyclic phosphodiesterase 4 (PDE4). Crystal structures of PDE4 in complex with glycyrrhisoflavone or licoisoflavone A were determined at resolutions of 1.54 Å and 1.65 Å, respectively, and both compounds bind in the active site of PDE4 with similar interactions. These findings will greatly stimulate the study of TCMT-NDRD against COVID-19.


Subject(s)
COVID-19 , Humans , Antiviral Agents/pharmacology , SARS-CoV-2 , Quercetin/pharmacology , Anti-Inflammatory Agents/pharmacology , Molecular Docking Simulation
5.
PLoS Pathog ; 14(11): e1007428, 2018 11.
Article in English | MEDLINE | ID: mdl-30422993

ABSTRACT

Severe influenza A virus infection causes high mortality and morbidity worldwide due to delayed antiviral treatment and inducing overwhelming immune responses, which contribute to immunopathological lung injury. Sirolimus, an inhibitor of mammalian target of rapamycin (mTOR), was effective in improving clinical outcomes in patients with severe H1N1 infection; however, the mechanisms by which it attenuates acute lung injury have not been elucidated. Here, delayed oseltamivir treatment was used to mimic clinical settings on lethal influenza A (H1N1) pdm09 virus (pH1N1) infection mice model. We revealed that delayed oseltamivir plus sirolimus treatment protects mice against lethal pH1N1 infection by attenuating severe lung damage. Mechanistically, the combined treatment reduced viral titer and pH1N1-induced mTOR activation. Subsequently, it suppressed the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated secretion of interleukin (IL)-1ß and IL-18. It was noted that decreased NLRP3 inflammasome activation was associated with inhibited nuclear factor (NF)-κB activation, reduced reactive oxygen species production and increased autophagy. Additionally, the combined treatment reduced the expression of other proinflammatory cytokines and chemokines, and decreased inflammatory cell infiltration in lung tissue and bronchioalveolar lavage fluid. Consistently, it inhibited the mTOR-NF-κB-NLRP3 inflammasome-IL-1ß axis in a lung epithelial cell line. These results demonstrated that combined treatment with sirolimus and oseltamivir attenuates pH1N1-induced severe lung injury, which is correlated with suppressed mTOR-NLRP3-IL-1ß axis and reduced viral titer. Therefore, treatment with sirolimus as an adjuvant along with oseltamivir may be a promising immunomodulatory strategy for managing severe influenza.


Subject(s)
Influenza A Virus, H1N1 Subtype/drug effects , Lung Injury/drug therapy , Lung Injury/virology , Oseltamivir/pharmacology , Sirolimus/pharmacology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Disease Models, Animal , Dogs , Drug Therapy, Combination/methods , Epithelial Cells , Female , Inflammasomes/drug effects , Inflammasomes/immunology , Influenza A Virus, H1N1 Subtype/metabolism , Interleukin-18/immunology , Interleukin-1beta/immunology , Lung/pathology , Lung Injury/metabolism , Lung Injury/pathology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
J Med Virol ; 91(11): 1918-1929, 2019 11.
Article in English | MEDLINE | ID: mdl-31317548

ABSTRACT

Influenza A (H1N1) is a rapidly spreading acute respiratory illness that remains a worldwide burden on public health. To simulate natural infection routes, BALB/C mice were challenged with the H1N1 virus by aerosol and intranasal instillation routes. We compared the weight change and survival of the mice for 14 consecutive days after infection. The infected mice were euthanized at days 3, 5, 7, and 9 to perform necropsies, lung pathological analyses, viral titers measurement, and lung cytokines examination. The aerosol-treated mice showed clinical symptoms on day 4, obvious lung lesions on day 5, rapid weight loss on day 7, peak virus replication in the lungs on days 7 to 9, and bronchial epithelial hyperplasia on day 9. However, after intranasal instillation, the mice exhibited clinical signs on day 2, rapid weight loss and obvious lung lesions on day 3, and peak virus replication in the lungs on days 3 to 5; no bronchial epithelial hyperplasia was detected. High levels of proinflammatory cytokines and chemokines were detected in the lungs of infected mice by both two routes. Disease and lung lesion progressions were slower in the mice that inhaled H1N1-containing aerosols than in those treated by intranasal instillation, and lung lesions were homogeneous in the aerosol group and heterogeneous in the intranasal group. In this study, BALB/C mouse models of H1N1 virus aerosol inhalation were successfully established and compared with mouse models of intranasal inoculation, aerosol mouse models had an infection route and lung pathology characteristics that more closely resembled those observed in humans.


Subject(s)
Disease Models, Animal , Lung/immunology , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Administration, Inhalation , Administration, Intranasal , Aerosols , Animals , Cytokines/immunology , Female , Influenza A Virus, H1N1 Subtype , Lung/pathology , Lung/virology , Mice , Specific Pathogen-Free Organisms , Virus Replication
8.
Proteomics ; 18(19): e1800003, 2018 10.
Article in English | MEDLINE | ID: mdl-30094923

ABSTRACT

Psoriasis is a common chronic autoimmune skin disease involving the activation of T cells. To explore the proteomic signature of peripheral blood mononuclear cells, a quantitative analysis of their global proteome was conducted in samples from Chinese patients with new-onset psoriasis (n = 31) and healthy controls (n = 32) using an integrated quantitative approach with tandem mass tag labeling and LC-MS/MS. Protein annotation, unsupervised hierarchical clustering, functional classification, functional enrichment and cluster, and protein-protein interaction analyses were performed. A total of 5178 proteins were identified, of which 4404 proteins were quantified. The fold-change cutoff was set at 1.2 (patients vs controls); 335 proteins were upregulated, and 107 proteins were downregulated. The bioinformatics analysis indicated that the differentially expressed proteins were involved in processes related to the activation of immune cells including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, cellular energy metabolism, and proliferation. Three upregulated proteins and two phosphorylated proteins in the NF-κB pathway were verified or identified by Western blotting. These results confirm that the NF-κB pathway is critical to psoriasis. In addition, many differentially expressed proteins identified in this study have never before been associated with psoriasis, and further studies on these proteins are necessary.


Subject(s)
Biomarkers/blood , Blood Proteins/analysis , Leukocytes, Mononuclear/metabolism , Proteome/analysis , Psoriasis/blood , Psoriasis/pathology , Adult , Age of Onset , Blood Proteins/metabolism , Case-Control Studies , Chromatography, Liquid , Computational Biology , Female , Humans , Male , Phosphorylation , Proteome/metabolism , Signal Transduction , Tandem Mass Spectrometry
9.
J Infect Dis ; 215(12): 1807-1815, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28472421

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) infection in humans is highly lethal, with a fatality rate of 35%. New prophylactic and therapeutic strategies to combat human infections are urgently needed. We isolated a fully human neutralizing antibody, MCA1, from a human survivor. The antibody recognizes the receptor-binding domain of MERS-CoV S glycoprotein and interferes with the interaction between viral S and the human cellular receptor human dipeptidyl peptidase 4 (DPP4). To our knowledge, this study is the first to report a human neutralizing monoclonal antibody that completely inhibits MERS-CoV replication in common marmosets. Monotherapy with MCA1 represents a potential alternative treatment for human infections with MERS-CoV worthy of evaluation in clinical settings.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , Coronavirus Infections/immunology , Dipeptidyl Peptidase 4/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Virus Replication/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Callithrix , Disease Models, Animal , Humans , Male , Random Allocation
12.
J Infect Dis ; 212(12): 1904-13, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26198719

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe disease in human with an overall case-fatality rate of >35%. Effective antivirals are crucial for improving the clinical outcome of MERS. Although a number of repurposed drugs, convalescent-phase plasma, antiviral peptides, and neutralizing antibodies exhibit anti-MERS-CoV activity in vitro, most are not readily available or have not been evaluated in nonhuman primates. We assessed 3 repurposed drugs with potent in vitro anti-MERS-CoV activity (mycophenolate mofetil [MMF], lopinavir/ritonavir, and interferon-ß1b) in common marmosets with severe disease resembling MERS in humans. The lopinavir/ritonavir-treated and interferon-ß1b-treated animals had better outcome than the untreated animals, with improved clinical (mean clinical scores ↓50.9%-95.0% and ↓weight loss than the untreated animals), radiological (minimal pulmonary infiltrates), and pathological (mild bronchointerstitial pneumonia) findings, and lower mean viral loads in necropsied lung (↓0.59-1.06 log10 copies/glyceraldehyde 3-phosphate dehydrogenase [GAPDH]; P < .050) and extrapulmonary (↓0.11-1.29 log10 copies/GAPDH; P < .050 in kidney) tissues. In contrast, all MMF-treated animals developed severe and/or fatal disease with higher mean viral loads (↑0.15-0.54 log10 copies/GAPDH) than the untreated animals. The mortality rate at 36 hours postinoculation was 67% (untreated and MMF-treated) versus 0-33% (lopinavir/ritonavir-treated and interferon-ß1b-treated). Lopinavir/ritonavir and interferon-ß1b alone or in combination should be evaluated in clinical trials. MMF alone may worsen MERS and should not be used.


Subject(s)
Antiviral Agents/administration & dosage , Coronavirus Infections/drug therapy , Interferon-beta/administration & dosage , Lopinavir/administration & dosage , Ritonavir/administration & dosage , Animals , Callithrix , Coronavirus Infections/pathology , Disease Models, Animal , Drug Repositioning , Male , Middle East Respiratory Syndrome Coronavirus/isolation & purification , Survival Analysis , Treatment Outcome , Viral Load
14.
J Gen Virol ; 96(Pt 1): 46-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25274854

ABSTRACT

The outbreak of human infections caused by the novel avian-origin H7N9 influenza viruses in China since March 2013 underscores the urgent need to find an effective treatment strategy against H7N9 infection in humans. In this study, we assessed the effectiveness of combinations of oseltamivir and two immunomodulators (simvastatin and fenofibrate) against H7N9 infection in a mouse model. Mice treated with oseltamivir plus fenofibrate exhibited the longest mean survival time, the largest reduction of viral titre in lung tissue, the highest levels of CD4(+) and CD8(+) T-lymphocytes, and the greatest decrease in pulmonary inflammation. Thus, the combination of oseltamivir plus fenofibrate improved the outcomes of mice infected with H7N9 virus by simultaneously reducing viral replication and normalizing the aberrant immune response. This drug combination should be considered in randomized controlled trials of treatments for H7N9 patients.


Subject(s)
Fibric Acids/pharmacology , Influenza A Virus, H7N9 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Oseltamivir/pharmacology , Animals , Antiviral Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Drug Therapy, Combination/methods , Female , Lung/virology , Mice , Mice, Inbred BALB C , Survival , Virus Replication/drug effects
15.
Arch Virol ; 160(1): 81-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25408373

ABSTRACT

In previous studies of hand, foot, and mouth disease patients fatally infected with enterovirus 71 (EV71), the distribution of viral protein, but not the genome, was determined. To understand the pathogenesis of EV71, however, it is important to investigate the spread of the viral genome. There have been no pathological studies of in situ EV71 viral RNA in inflammatory cells infiltrating various tissues of fatal cases. We therefore first investigated the distribution and classification of inflammatory cells in various tissues and then performed in situ EV71 RNA hybridization in these tissues to better understand the pathogenesis of EV71 infection. EV71 RNA was found mainly in inflammatory cells infiltrating the central nervous system (CNS), intestines, lungs, and tonsils. Most EV71 RNA-positive inflammatory cells in the CNS were macrophages/microglia and neutrophils infiltrating the perivascular cuffing, microglial nodule, neuronophagia, and meninges. CD68+ macrophages and CD15+ neutrophils were diffusely distributed in tissues with severe pathological changes. This study demonstrates the presence of EV71 RNA in inflammatory cells infiltrating tissues in fatally infected patients. Our findings suggest that fatal EV71 infection with extensive infiltration of macrophages/microglia and neutrophils into the CNS results in severe neurological lesions.


Subject(s)
Enterovirus A, Human/isolation & purification , Hand, Foot and Mouth Disease/virology , Immunity, Cellular/physiology , Inflammation/virology , RNA, Viral/isolation & purification , Genome, Viral , Hand, Foot and Mouth Disease/mortality , Humans , In Situ Hybridization , Tissue Distribution
16.
J Infect Dis ; 209(2): 236-42, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24218506

ABSTRACT

In 2012, a novel coronavirus (CoV) associated with severe respiratory disease, Middle East respiratory syndrome (MERS-CoV; previously known as human coronavirus-Erasmus Medical Center or hCoV-EMC), emerged in the Arabian Peninsula. To date, 114 human cases of MERS-CoV have been reported, with 54 fatalities. Animal models for MERS-CoV infection of humans are needed to elucidate MERS pathogenesis and to develop vaccines and antivirals. In this study, we developed rhesus macaques as a model for MERS-CoV using intratracheal inoculation. The infected monkeys showed clinical signs of disease, virus replication, histological lesions, and neutralizing antibody production, indicating that this monkey model is suitable for studies of MERS-CoV infection.


Subject(s)
Coronavirus Infections/pathology , Coronavirus Infections/virology , Coronavirus/physiology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Histocytochemistry , Macaca mulatta , Virus Replication
17.
J Infect Dis ; 209(4): 551-6, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-23990570

ABSTRACT

The outbreak of human infections caused by novel avian-origin influenza A(H7N9) in China since March 2013 underscores the need to better understand the pathogenicity and transmissibility of these viruses in mammals. In a ferret model, the pathogenicity of influenza A(H7N9) was found to be less than that of an influenza A(H5N1) strain but comparable to that of 2009 pandemic influenza A(H1N1), based on the clinical signs, mortality, virus dissemination, and results of histopathologic analyses. Influenza A(H7N9) could replicate in the upper and lower respiratory tract, the heart, the liver, and the olfactory bulb. It is worth noting that influenza A(H7N9) exhibited a low level of transmission between ferrets via respiratory droplets. There were 4 mutations in the virus isolated from the contact ferret: D678Y in the gene encoding PB2, R157K in the gene encoding hemagglutinin (H3 numbering), I109T in the gene encoding nucleoprotein, and T10I in the gene encoding neuraminidase. These data emphasized that avian-origin influenza A(H7N9) can be transmitted between mammals, highlighting its potential for human-to-human transmissibility.


Subject(s)
Influenza A Virus, H7N9 Subtype/physiology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Administration, Intranasal , Animals , Body Weight , Disease Models, Animal , Environmental Exposure , Ferrets/virology , Influenza A Virus, H7N9 Subtype/pathogenicity , Lung/chemistry , Lung/pathology , Lung/virology , Nasal Cavity/virology , Pharynx/virology
18.
Virol J ; 11: 185, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25367670

ABSTRACT

BACKGROUND: On 19 February 2013, the first patient infected with a novel influenza A H7N9 virus from an avian source showed symptoms of sickness. More than 349 laboratory-confirmed cases and 109 deaths have been reported in mainland China since then. Laboratory-confirmed, human-to-human H7N9 virus transmission has not been documented between individuals having close contact; however, this transmission route could not be excluded for three families. To control the spread of the avian influenza H7N9 virus, we must better understand its pathogenesis, transmissibility, and transmission routes in mammals. Studies have shown that this particular virus is transmitted by aerosols among ferrets. METHODS: To study potential transmission routes in animals with direct or close contact to other animals, we investigated these factors in a murine model. RESULTS: Viable H7N9 avian influenza virus was detected in the upper and lower respiratory tracts, intestine, and brain of model mice. The virus was transmissible between mice in close contact, with a higher concentration of virus found in pharyngeal and ocular secretions, and feces. All these biological materials were contagious for naïve mice. CONCLUSIONS: Our results suggest that the possible transmission routes for the H7N9 influenza virus were through mucosal secretions and feces.


Subject(s)
Disease Models, Animal , Influenza A Virus, H7N9 Subtype/physiology , Influenza, Human/transmission , Animals , Chick Embryo , Female , History, Ancient , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza, Human/virology , Mice , Mice, Inbred BALB C
19.
Immunol Invest ; 43(3): 224-35, 2014.
Article in English | MEDLINE | ID: mdl-24295504

ABSTRACT

Vaccination represents the most economic and effective strategy of preventing influenza pandemics. We previously demonstrated that intranasal immunization of mice with recombinant hemagglutinin and the mast cell activator C48/80 elicited protective immunity against challenge with the 2009 pandemic H1N1 influenza in mice, demonstrating that the novel C48/80 mucosal adjuvant was safe and effective. The present study demonstrated that intranasal immunization with inactivated H1N1 virus and C48/80 elicited protective immunity against lethal challenge with homologous virus, however, when the immunogen was replaced with inactivated H5N1 virus protection was lost. These observations suggested that the adjuvant effects conferred by C48/80 were virus subtype specific and that its use as a broad-spectrum adjuvant for use in immunizations against all influenza viruses needs to be further analyzed.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , Polymers/administration & dosage , Administration, Intranasal , Animals , Female , Humans , Immunization , Influenza, Human/immunology , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated
20.
Zhonghua Bing Li Xue Za Zhi ; 43(2): 109-13, 2014 Feb.
Article in Zh | MEDLINE | ID: mdl-24742571

ABSTRACT

OBJECTIVE: To document ultrastructural changes of brain, spinal cord, skeletal muscle, jejunum and lung of EV71 infection mouse model, and to explore the myotropism and pathogenesis of EV71 in nervous system. METHODS: Ten-day-old suckling mice were infected with EV71 strain via the intraperitoneal route. Mice with paralysis were scarified on day 4 post infection and the brain, spinal cord, skeletal muscle, jejunum and lung were sampled for transmission electron microscopy and light microscopy. RESULTS: Lesions in brain were generally mild with inner chamber swelling in some of mitochondria. Myelin sheaths of medullated fibers were split with vacuolated changes. The Nissl bodies in anterior motor neurons disappeared along with mitochondria swelling, rough endoplasmic reticulum swelling and degranulation. Cytoplasm of anterior motor neurons showed cribriform appearance accompanied by neuronophagia. The bands of skeletal muscle in the infected group disappeared with degeneration and karyopyknosis in myocytes, in addition to mitochondrial swelling. Microvilli of epithelium in jejunum became loosely arranged along with formation of spiral medullary sheath structure and mitochondria swelling. Interstitial pneumonia was observed in lungs with type II pneumocyte proliferation and evacuation of the multilamellar bodies. CONCLUSIONS: EV71 infection causes severe myositis in the mouse model suggesting a strong myotropism of EV71 virus. The presence of lesions of various degrees in central nervous system and changes in anterior motor neurons may be associated with limb paralysis.


Subject(s)
Disease Models, Animal , Enterovirus A, Human , Enterovirus Infections/pathology , Muscle, Skeletal/ultrastructure , Animals , Brain/ultrastructure , Brain/virology , Enterovirus Infections/virology , Jejunum/ultrastructure , Jejunum/virology , Lung/ultrastructure , Lung/virology , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Muscle, Skeletal/virology , Spinal Cord/ultrastructure , Spinal Cord/virology
SELECTION OF CITATIONS
SEARCH DETAIL