Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38176410

ABSTRACT

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Subject(s)
Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Receptors, LDL , Animals , Mice , Alphavirus/physiology , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Eastern Equine/ultrastructure , Encephalomyelitis, Equine/metabolism , Horses , Protein Binding , Receptors, LDL/ultrastructure
2.
bioRxiv ; 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38014196

ABSTRACT

The very low-density lipoprotein receptor (VLDLR) is comprised of eight LDLR type A (LA) domains and supports entry of distantly related Eastern equine encephalitis (EEEV) and Semliki Forest (SFV) alphaviruses. Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage different LA domains simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection, highlighting complexity in domain usage. Whereas all EEEV strains show conservation of two VLDLR binding sites, the EEEV PE-6 strain and other EEE complex members feature a single amino acid substitution that mediates binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.

3.
Cell ; 186(22): 4818-4833.e25, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37804831

ABSTRACT

MXRA8 is a receptor for chikungunya (CHIKV) and other arthritogenic alphaviruses with mammalian hosts. However, mammalian MXRA8 does not bind to alphaviruses that infect humans and have avian reservoirs. Here, we show that avian, but not mammalian, MXRA8 can act as a receptor for Sindbis, western equine encephalitis (WEEV), and related alphaviruses with avian reservoirs. Structural analysis of duck MXRA8 complexed with WEEV reveals an inverted binding mode compared with mammalian MXRA8 bound to CHIKV. Whereas both domains of mammalian MXRA8 bind CHIKV E1 and E2, only domain 1 of avian MXRA8 engages WEEV E1, and no appreciable contacts are made with WEEV E2. Using these results, we generated a chimeric avian-mammalian MXRA8 decoy-receptor that neutralizes infection of multiple alphaviruses from distinct antigenic groups in vitro and in vivo. Thus, different alphaviruses can bind MXRA8 encoded by different vertebrate classes with distinct engagement modes, which enables development of broad-spectrum inhibitors.


Subject(s)
Alphavirus , Animals , Humans , Chikungunya Fever , Chikungunya virus/chemistry , Mammals , Receptors, Virus/metabolism
4.
iScience ; 26(10): 107764, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37736038

ABSTRACT

Pandemic SARS-CoV-2 has undergone rapid evolution resulting in the emergence of many variants with mutations in the spike protein, some of which appear to evade antibody neutralization, transmit more efficiently, and/or exhibit altered virulence. This raises significant concerns regarding the efficacy of anti-S monoclonal antibody-based therapeutics which have failed against variant SARS-CoV-2 viruses. To address this concern, SAB-185, a human anti-SARS-CoV-2 polyclonal antibody was generated in the DiversitAb platform. SAB-185 exhibited equivalent, robust in vitro neutralization for Munich, Alpha, Beta, Gamma, and Δ144-146 variants and, although diminished, retained PRNT50 and PRNT80 neutralization endpoints for Delta and Omicron variants. Human ACE2 transgenic Syrian hamsters, which exhibit lethal SARS-CoV-2 disease, were protected from mortality after challenge with the Munich, Alpha, Beta, Delta, and Δ144-146 variants and clinical signs after non-lethal Omicron BA.1 infection. This suggests that SAB-185 may be an effective immunotherapy even in the presence of ongoing viral mutation.

5.
PLoS Pathog ; 18(6): e1009946, 2022 06.
Article in English | MEDLINE | ID: mdl-35696423

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a positively-stranded RNA arbovirus of the genus Alphavirus that causes encephalitis in humans. Cynomolgus macaques are a relevant model of the human disease caused by VEEV and are useful in exploring pathogenic mechanisms and the host response to VEEV infection. Macaques were exposed to small-particle aerosols containing virus derived from an infectious clone of VEEV strain INH-9813, a subtype IC strain isolated from a human infection. VEEV-exposed macaques developed a biphasic fever after infection similar to that seen in humans. Maximum temperature deviation correlated with the inhaled dose, but fever duration did not. Neurological signs, suggestive of virus penetration into the central nervous system (CNS), were predominantly seen in the second febrile period. Electroencephalography data indicated a statistically significant decrease in all power bands and circadian index during the second febrile period that returned to normal after fever resolved. Intracranial pressure increased late in the second febrile period. On day 6 post-infection macaques had high levels of MCP-1 and IP-10 chemokines in the CNS, as well as a marked increase of T lymphocytes and activated microglia. More than four weeks after infection, VEEV genomic RNA was found in the brain, cerebrospinal fluid and cervical lymph nodes. Pro-inflammatory cytokines & chemokines, infiltrating leukocytes and pathological changes were seen in the CNS tissues of macaques euthanized at these times. These data are consistent with persistence of virus replication and/or genomic RNA and potentially, inflammatory sequelae in the central nervous system after resolution of acute VEEV disease.


Subject(s)
Encephalitis Virus, Venezuelan Equine , Encephalomyelitis, Venezuelan Equine , Animals , Central Nervous System , Encephalitis Virus, Venezuelan Equine/genetics , Horses/genetics , Inflammation , Macaca fascicularis , RNA, Viral/genetics
6.
Hum Vaccin Immunother ; 18(2): 1940652, 2022 04 29.
Article in English | MEDLINE | ID: mdl-34228597

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with amino-acid substitutions and deletions in spike protein (S) can reduce the effectiveness of monoclonal antibodies (mAbs) and may compromise immunity induced by vaccines. We report a polyclonal, fully human, anti-SARS-CoV-2 immunoglobulin produced in transchromosomic bovines (Tc-hIgG-SARS-CoV-2) hyperimmunized with two doses of plasmid DNA encoding the SARS-CoV-2 Wuhan strain S gene, followed by repeated immunization with S protein purified from insect cells. The resulting Tc-hIgG-SARS-CoV-2, termed SAB-185, efficiently neutralizes SARS-CoV-2, and vesicular stomatitis virus (VSV) SARS-CoV-2 chimeras in vitro. Neutralization potency was retained for S variants including S477N, E484K, and N501Y, substitutions present in recent variants of concern. In contrast to the ease of selection of escape variants with mAbs and convalescent human plasma, we were unable to isolate VSV-SARS-CoV-2 mutants resistant to Tc-hIgG-SARS-CoV-2 neutralization. This fully human immunoglobulin that potently inhibits SARS-CoV-2 infection may provide an effective therapeutic to combat COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Cattle , Humans , Immunoglobulin G , Neutralization Tests/methods , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus
7.
Viruses ; 15(1)2022 12 20.
Article in English | MEDLINE | ID: mdl-36680046

ABSTRACT

The Department of Defense recently began an effort to improve and standardize virus challenge materials and efficacy determination strategies for testing therapeutics and vaccines. This includes stabilization of virus genome sequences in cDNA form where appropriate, use of human-derived virus isolates, and noninvasive strategies for determination of challenge virus replication. Eventually, it is desired that these approaches will satisfy the FDA "Animal Rule" for licensure, which substitutes animal efficacy data when human data are unlikely to be available. To this end, we created and examined the virulence phenotype of cDNA clones of prototypic human infection-derived strains of the alphaviruses, Venezuelan (VEEV INH9813), eastern (EEEV V105) and western (WEEV Fleming) equine encephalitis viruses, and created fluorescent and luminescent reporter expression vectors for evaluation of replication characteristics in vitro and in vivo. Sequences of minimally passaged isolates of each virus were used to synthesize full-length cDNA clones along with a T7 transcription promoter-based bacterial propagation vector. Viruses generated from the cDNA clones were compared with other "wild type" strains derived from cDNA clones and GenBank sequences to identify and eliminate putative tissue culture artifacts accumulated in the cell passaged biological stocks. This was followed by examination of aerosol and subcutaneous infection and disease in mouse models. A mutation that increased heparan sulfate binding was identified in the VEEV INH9813 biological isolate sequence and eliminated from the cDNA clone. Viruses derived from the new human isolate cDNA clones showed similar mouse virulence to existing clone-derived viruses after aerosol or subcutaneous inoculation.


Subject(s)
Encephalitis Virus, Venezuelan Equine , Encephalitis Virus, Western Equine , United States , Humans , Animals , Horses , Mice , DNA, Complementary/genetics , Phenotype , Clone Cells
8.
bioRxiv ; 2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34341790

ABSTRACT

Pandemic SARS CoV-2 has been undergoing rapid evolution during spread throughout the world resulting in the emergence of many Spike protein variants, some of which appear to either evade antibody neutralization, transmit more efficiently, or potentially exhibit increased virulence. This raises significant concerns regarding the long-term efficacy of protection elicited after primary infection and/or from vaccines derived from single virus Spike (S) genotypes, as well as the efficacy of anti-S monoclonal antibody based therapeutics. Here, we used fully human polyclonal human IgG (SAB-185), derived from hyperimmunization of transchromosomic bovines with DNA plasmids encoding the SARS-CoV-2 Wa-1 strain S protein or purified ectodomain of S protein, to examine the neutralizing capacity of SAB-185 in vitro and the protective efficacy of passive SAB-185 antibody (Ab) transfer in vivo . The Ab preparation was tested for neutralization against five variant SARS-CoV-2 strains: Munich (Spike D614G), UK (B.1.1.7), Brazil (P.1) and SA (B.1.3.5) variants, and a variant isolated from a chronically infected immunocompromised patient (Spike Δ144-146). For the in vivo studies, we used a new human ACE2 (hACE2) transgenic Syrian hamster model that exhibits lethality after SARS-Cov-2 challenge and the Munich, UK, SA and Δ144-146 variants. SAB-185 neutralized each of the SARS-CoV-2 strains equivalently on Vero E6 cells, however, a control convalescent human serum sample was less effective at neutralizing the SA variant. In the hamster model, prophylactic SAB-185 treatment protected the hamsters from fatal disease and minimized clinical signs of infection. These results suggest that SAB-185 may be an effective treatment for patients infected with SARS CoV-2 variants.

9.
Cell ; 184(17): 4414-4429.e19, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34416146

ABSTRACT

Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.


Subject(s)
Alphavirus/immunology , Antibodies, Viral/immunology , Conserved Sequence/immunology , Epitopes/immunology , Viral Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chlorocebus aethiops , Epitope Mapping , Epitopes/chemistry , Humans , Male , Mice, Inbred C57BL , Models, Biological , Monocytes/metabolism , Vero Cells , Viral Proteins/chemistry , Virus Release
10.
PLoS Pathog ; 17(2): e1009308, 2021 02.
Article in English | MEDLINE | ID: mdl-33534855

ABSTRACT

Aerosol exposure to eastern equine encephalitis virus (EEEV) can trigger a lethal viral encephalitis in cynomolgus macaques which resembles severe human disease. Biomarkers indicative of central nervous system (CNS) infection by the virus and lethal outcome of disease would be useful in evaluating potential medical countermeasures, especially for therapeutic compounds. To meet requirements of the Animal Rule, a better understanding of the pathophysiology of EEEV-mediated disease in cynomolgus macaques is needed. In this study, macaques given a lethal dose of clone-derived EEEV strain V105 developed a fever between 2-3 days post infection (dpi) and succumbed to the disease by 6 dpi. At the peak of the febrile phase, there was a significant increase in the delta electroencephalography (EEG) power band associated with deep sleep as well as a sharp rise in intracranial pressure (ICP). Viremia peaked early after infection and was largely absent by the onset of fever. Granulocytosis and elevated plasma levels of IP-10 were found early after infection. At necropsy, there was a one hundred- to one thousand-fold increase in expression of traumatic brain injury genes (LIF, MMP-9) as well as inflammatory cytokines and chemokines (IFN-γ, IP-10, MCP-1, IL-8, IL-6) in the brain tissues. Phenotypic analysis of leukocytes entering the brain identified cells as primarily lymphoid (T, B, NK cells) with lower levels of infiltrating macrophages and activated microglia. Massive amounts of infectious virus were found in the brains of lethally-infected macaques. While no infectious virus was found in surviving macaques, quantitative PCR did find evidence of viral genomes in the brains of several survivors. These data are consistent with an overwhelming viral infection in the CNS coupled with a tremendous inflammatory response to the infection that may contribute to the disease outcome. Physiological monitoring of EEG and ICP represent novel methods for assessing efficacy of vaccines or therapeutics in the cynomolgus macaque model of EEEV encephalitis.


Subject(s)
Aerosols/adverse effects , Biomarkers/analysis , Brain/immunology , Brain/pathology , Encephalitis Virus, Eastern Equine/pathogenicity , Encephalitis, Viral/immunology , Fever/immunology , Animals , Brain/virology , Cytokines/metabolism , Disease Models, Animal , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Female , Fever/pathology , Fever/virology , Macaca fascicularis , Male
11.
Cell ; 183(7): 1884-1900.e23, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33301709

ABSTRACT

Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.


Subject(s)
Aerosols/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/prevention & control , Adult , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Cryoelectron Microscopy , Disease Models, Animal , Encephalitis Virus, Eastern Equine/ultrastructure , Encephalomyelitis, Equine/virology , Epitopes/chemistry , Female , Glycoproteins/immunology , Humans , Mice , Models, Molecular , Mutagenesis/genetics , Neutralization Tests , Protein Binding , Protein Domains , Recombinant Proteins/immunology , Sindbis Virus/immunology , Virion/immunology , Virion/ultrastructure , Virus Internalization
12.
Nature ; 588(7837): 308-314, 2020 12.
Article in English | MEDLINE | ID: mdl-33208938

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a neurotropic alphavirus transmitted by mosquitoes that causes encephalitis and death in humans1. VEEV is a biodefence concern because of its potential for aerosol spread and the current lack of sufficient countermeasures. The host factors that are required for VEEV entry and infection remain poorly characterized. Here, using a genome-wide CRISPR-Cas9-based screen, we identify low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3)-a highly conserved yet poorly characterized member of the scavenger receptor superfamily-as a receptor for VEEV. Gene editing of mouse Ldlrad3 or human LDLRAD3 results in markedly reduced viral infection of neuronal cells, which is restored upon complementation with LDLRAD3. LDLRAD3 binds directly to VEEV particles and enhances virus attachment and internalization into host cells. Genetic studies indicate that domain 1 of LDLRAD3 (LDLRAD3(D1)) is necessary and sufficient to support infection by VEEV, and both anti-LDLRAD3 antibodies and an LDLRAD3(D1)-Fc fusion protein block VEEV infection in cell culture. The pathogenesis of VEEV infection is abrogated in mice with deletions in Ldlrad3, and administration of LDLRAD3(D1)-Fc abolishes disease caused by several subtypes of VEEV, including highly virulent strains. The development of a decoy-receptor fusion protein suggests a strategy for the prevention of severe VEEV infection and associated disease in humans.


Subject(s)
Encephalitis Virus, Venezuelan Equine/metabolism , Receptors, LDL/metabolism , Receptors, Virus/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/virology , Female , Genetic Complementation Test , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Virus/genetics , Virus Attachment , Virus Internalization
13.
PLoS Pathog ; 16(9): e1008903, 2020 09.
Article in English | MEDLINE | ID: mdl-32946524

ABSTRACT

Vaccines are urgently needed to combat the global coronavirus disease 2019 (COVID-19) pandemic, and testing of candidate vaccines in an appropriate non-human primate (NHP) model is a critical step in the process. Infection of African green monkeys (AGM) with a low passage human isolate of SARS-CoV-2 by aerosol or mucosal exposure resulted in mild clinical infection with a transient decrease in lung tidal volume. Imaging with human clinical-grade 18F-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) co-registered with computed tomography (CT) revealed pulmonary lesions at 4 days post-infection (dpi) that resolved over time. Infectious virus was shed from both respiratory and gastrointestinal (GI) tracts in all animals in a biphasic manner, first between 2-7 dpi followed by a recrudescence at 14-21 dpi. Viral RNA (vRNA) was found throughout both respiratory and gastrointestinal systems at necropsy with higher levels of vRNA found within the GI tract tissues. All animals seroconverted simultaneously for IgM and IgG, which has also been documented in human COVID-19 cases. Young AGM represent an species to study mild/subclinical COVID-19 disease and with possible insights into live virus shedding. Future vaccine evaluation can be performed in AGM with correlates of efficacy being lung lesions by PET/CT, virus shedding, and tissue viral load.


Subject(s)
Betacoronavirus , Coronavirus Infections/diagnostic imaging , Gastrointestinal Tract/virology , Pneumonia, Viral/diagnostic imaging , Virus Shedding/physiology , Animals , COVID-19 , Chlorocebus aethiops , Coronavirus Infections/virology , Lung/pathology , Lung/virology , Pandemics , Pneumonia, Viral/virology , Positron Emission Tomography Computed Tomography/methods , SARS-CoV-2
14.
J Gen Virol ; 101(11): 1156-1169, 2020 11.
Article in English | MEDLINE | ID: mdl-32821033

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), emerged at the end of 2019 and by mid-June 2020 the virus had spread to at least 215 countries, caused more than 8 000 000 confirmed infections and over 450 000 deaths, and overwhelmed healthcare systems worldwide. Like severe acute respiratory syndrome coronavirus (SARS-CoV), which emerged in 2002 and caused a similar disease, SARS-CoV-2 is a betacoronavirus. Both viruses use human angiotensin-converting enzyme 2 (hACE2) as a receptor to enter cells. However, the SARS-CoV-2 spike (S) glycoprotein has a novel insertion that generates a putative furin cleavage signal and this has been postulated to expand the host range. Two low-passage (P) strains of SARS-CoV-2 (Wash1 : P4 and Munich : P1) were cultured twice in Vero E6 cells and characterized virologically. Sanger and MinION sequencing demonstrated significant deletions in the furin cleavage signal of Wash1 : P6 and minor variants in the Munich : P3 strain. Cleavage of the S glycoprotein in SARS-CoV-2-infected Vero E6 cell lysates was inefficient even when an intact furin cleavage signal was present. Indirect immunofluorescence demonstrated that the S glycoprotein reached the cell surface. Since the S protein is a major antigenic target for the development of neutralizing antibodies, we investigated the development of neutralizing antibody titres in serial serum samples obtained from COVID-19 human patients. These were comparable regardless of the presence of an intact or deleted furin cleavage signal. These studies illustrate the need to characterize virus stocks meticulously prior to performing either in vitro or in vivo pathogenesis studies.


Subject(s)
COVID-19/metabolism , COVID-19/virology , Furin/metabolism , Host-Pathogen Interactions , SARS-CoV-2/physiology , Virus Replication , Adaptation, Physiological , Animals , Antibodies, Neutralizing/immunology , COVID-19/epidemiology , COVID-19/immunology , Chlorocebus aethiops , Furin/immunology , Genetic Variation , Hospitalization , Host-Pathogen Interactions/immunology , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Neutralization Tests , Proteolysis , RNA, Viral , Sequence Analysis, RNA , Vero Cells , Viral Load
15.
bioRxiv ; 2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32607507

ABSTRACT

SARS-CoV-2, the causative agent of COVID-19, emerged at the end of 2019 and by mid-June 2020, the virus has spread to at least 215 countries, caused more than 8,000,000 confirmed infections and over 450,000 deaths, and overwhelmed healthcare systems worldwide. Like SARS-CoV, which emerged in 2002 and caused a similar disease, SARS-CoV-2 is a betacoronavirus. Both viruses use human angiotensin-converting enzyme 2 (hACE2) as a receptor to enter cells. However, the SARS-CoV-2 spike (S) glycoprotein has a novel insertion that generates a putative furin cleavage signal and this has been postulated to expand the host range. Two low passage (P) strains of SARS-CoV-2 (Wash1: P4 and Munich: P1) were cultured twice in Vero-E6 cells and characterized virologically. Sanger and MinION sequencing demonstrated significant deletions in the furin cleavage signal of Wash1: P6 and minor variants in the Munich: P3 strain. Cleavage of the S glycoprotein in SARS-CoV-2-infected Vero-E6 cell lysates was inefficient even when an intact furin cleavage signal was present. Indirect immunofluorescence demonstrated the S glycoprotein reached the cell surface. Since the S protein is a major antigenic target for the development of neutralizing antibodies we investigated the development of neutralizing antibody titers in serial serum samples obtained from COVID-19 human patients. These were comparable regardless of the presence of an intact or deleted furin cleavage signal. These studies illustrate the need to characterize virus stocks meticulously prior to performing either in vitro or in vivo pathogenesis studies.

16.
Mil Med ; 181(8): 756-66, 2016 08.
Article in English | MEDLINE | ID: mdl-27483511

ABSTRACT

Dengue, one of the most widespread infectious diseases, has affected U.S. military readiness throughout history. We explored the dengue diagnosis capability gap by circulating a questionnaire among military end users to determine in what capacity diagnostic test results are needed and how these results would be used at various roles of care in the Military Health System. Results were used to generate target product profiles for potential diagnostic tests. We determined that at far-forward locations, diagnostic tests need to be rugged and easy to use and are primarily needed to inform medical evacuation decisions. In mobile or fixed hospitals, diagnostics can be less portable but must be accurate enough to inform patient care decisions reliably. We then evaluated the suitability of using rapid diagnostic tests and enzyme-linked immunosorbent assays based on published performance characteristics, and we used a model to determine positive and negative predictive values in certain simulated deployments. In far-forward settings, a rapid diagnostic test comprising both antigen- and antibody-based detection can fulfill the capability gap with reasonable accuracy, whereas at higher roles of care immunoglobulin M-enzyme-linked immunosorbent assay was determined to be the most suitable option.


Subject(s)
Dengue/diagnosis , Diagnostic Tests, Routine/standards , Sensitivity and Specificity , Antigen-Antibody Reactions , Dengue Virus , Diagnostic Tests, Routine/methods , Enzyme-Linked Immunosorbent Assay/methods , Humans , Military Medicine/methods , Military Medicine/standards , Public Health/methods , Surveys and Questionnaires , Time Factors , Workforce
17.
Diagn Microbiol Infect Dis ; 83(1): 30-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26032430

ABSTRACT

During dengue outbreaks, acute diagnosis at the patient's point of need followed by appropriate supportive therapy reduces morbidity and mortality. To facilitate needed diagnosis, we developed and optimized a reverse transcription loop-mediated isothermal amplification (RT-LAMP) assay that detects all 4 serotypes of dengue virus (DENV). We used a quencher to reduce nonspecific amplification. The assay does not require expensive thermocyclers, utilizing a simple water bath to maintain the reaction at 63 °C. Results can be visualized using UV fluorescence, handheld readers, or lateral flow immunochromatographic tests. We report a sensitivity of 86.3% (95% confidence interval [CI], 72.7-94.8%) and specificity of 93.0% (95% CI, 83.0-98.1%) using a panel of clinical specimens characterized by DENV quantitative reverse transcription-polymerase chain reaction. This pan-serotype DENV RT-LAMP can be adapted to field-expedient formats where it can provide actionable diagnosis near the patient's point of need.


Subject(s)
Dengue Virus/classification , Dengue Virus/isolation & purification , Dengue/diagnosis , Dengue/virology , Nucleic Acid Amplification Techniques/methods , Serogroup , Dengue Virus/genetics , Humans , RNA, Viral/metabolism , Reverse Transcription , Sensitivity and Specificity , Temperature
18.
Am J Trop Med Hyg ; 93(1): 46-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25940193

ABSTRACT

Loss of sample integrity during specimen transport can lead to false-negative diagnostic results. In an effort to improve upon the status quo, we used dengue as a model RNA virus to evaluate the stabilization of RNA and antibodies in three commercially available sample stabilization products: Whatman FTA Micro Cards (GE Healthcare Life Sciences, Pittsburgh, PA), DNAstable Blood tubes (Biomatrica, San Diego, CA), and ViveST tubes (ViveBio, Alpharetta, GA). Both contrived and clinical dengue-positive specimens were stored on these products at ambient temperature or 37°C for up to 1 month. Antibody and viral RNA levels were measured by enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) assays, respectively, and compared with frozen unloaded controls. We observed reduced RNA and antibody levels between stabilized contrived samples and frozen controls at our earliest time point, and this was particularly pronounced for the FTA cards. However, despite some time and temperature dependent loss, a 94.6-97.3% agreement was observed between stabilized clinical specimens and their frozen controls for all products. Additional considerations such as cost, sample volume, matrix, and ease of use should inform any decision to incorporate sample stabilization products into a diagnostic testing workflow. We conclude that DNAstable Blood and ViveST tubes are useful alternatives to traditional filter paper for ambient temperature shipment of clinical specimens for downstream molecular and serological testing.


Subject(s)
Antibodies, Viral/analysis , Cryopreservation , Dengue Virus/genetics , Dengue/diagnosis , RNA, Viral/analysis , Specimen Handling/methods , Temperature , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulins/analysis , RNA Stability , Reverse Transcriptase Polymerase Chain Reaction , Specimen Handling/instrumentation , Time Factors
19.
Vaccine ; 29(40): 7009-19, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21787825

ABSTRACT

BACKGROUND: Shigella flexneri 2a lipopolysaccharide 50 is a nasally delivered subunit vaccine consisting of a macromolecular complex composed of LPS, IpaB, IpaC and IpaD. The current study examined vaccine safety and immunogenicity across a dose range and the clinical performance of a new intranasal delivery device. METHODS: Volunteers (N=36) were randomized to receive vaccine via the Dolphin™ (Valois of America, Congers, New York) intranasal spray device at one of three doses (240, 480, and 690 µg) on days 0, 14, and 28. Another group (N=8) received the 240 µg dose via pipette. Vaccine safety was actively monitored and antigen-specific humoral and mucosal immune responses were determined. RESULTS: There were no serious adverse events and the majority of adverse events (98%) were mild. Antibody secreting cells (ASC), plasma, and mucosal immune responses to Shigella antigens were detected at all three dose levels with the 690 µg dose inducing the highest magnitude and frequency of responses. Vaccination with comparable doses of Invaplex 50 via the Dolphin™ resulted in higher plasma and ASC immune responses as compared to pipette delivery. CONCLUSION: In this trial the S. flexneri 2a Invaplex 50 vaccine was safe, well-tolerated and induced robust levels of antigen-specific intestinal IgA and ASC responses. The spray device performed well and offered an advantage over pipette intranasal delivery.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Immunity, Mucosal/immunology , Lipopolysaccharides/immunology , Shigella Vaccines/administration & dosage , Shigella Vaccines/immunology , Shigella flexneri/immunology , Administration, Intranasal , Adolescent , Adult , Animals , Antibodies, Bacterial/blood , Antibody-Producing Cells/immunology , Double-Blind Method , Drug Administration Routes , Female , Guinea Pigs , Humans , Immunity, Humoral/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Male , Mice , Middle Aged , Nasal Sprays , Shigella Vaccines/adverse effects , Vaccination/methods , Young Adult
20.
Clin Infect Dis ; 49(10): 1512-9, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19842970

ABSTRACT

BACKGROUND: A robust human challenge model for Campylobacter jejuni is an important tool for the evaluation of candidate vaccines. The previously established model conveys a potential risk of Guillain-Barré syndrome attributable to lipooligosaccharide ganglioside mimicry. This work establishes a new C. jejuni human challenge model that uses a strain (CG8421) without ganglioside mimicry and that applies Campylobacter-specific cellular immunity screening to achieve high attack rates at lower inoculum doses. METHODS: Healthy Campylobacter-naive adults participated in an open-label challenge trial. Participants were dosed with C. jejuni CG8421 and followed as inpatients. Pattern of illness, bacterial shedding, and immunologic responses were determined. RESULTS: Following screening, 23 subjects received 1 X 10(6) or 1 X 10(5) colony-forming units of C. jejuni, with attack rates (percentage of patients who became ill) of 100% (1 X 10(6) colony-forming units) or 93% (1 X 10(5) colony-forming units). Every subject shed CG8421; the median time to diarrhea onset was 72.3 h (interquartile range, 53.9-99.9 h). Symptoms included abdominal cramps (74%), nausea (65%), and fever (39%). No major safety concerns occurred, including bacteremia, hypotension, or postinfectious sequelae. Unexpectedly, recrudescent infection occurred in 2 subjects (1 subject without Campylobacter-specific adaptive immune responses and 1 with azithromycin resistance acquired in vivo); both infections cleared after receipt of additional antibiotics. Cumulative Campylobacter-specific immune responses were as follows: serologic response occurred in 87% (immunoglobulin [Ig] A) and 48% (IgG) of subjects, in vitro interferon-gamma production occurred in 91% of subjects, and 96% of subjects had IgA antibody-secreting cells and fecal IgA detected. CONCLUSIONS: The C. jejuni CG8421 challenge model provides a safe and effective tool, without the risk of Guillain-Barré syndrome. The model demonstrates high attack rates after lower doses of challenge inoculum, provides further understanding of immunologic responses, and permits future investigation of candidate Campylobacter vaccines.


Subject(s)
Bacterial Vaccines/immunology , Campylobacter Infections/microbiology , Campylobacter Infections/pathology , Campylobacter jejuni/immunology , Campylobacter jejuni/pathogenicity , Drug Evaluation/methods , Adolescent , Adult , Antibodies, Bacterial/blood , Campylobacter Infections/immunology , Campylobacter Infections/prevention & control , Diarrhea/immunology , Diarrhea/microbiology , Diarrhea/pathology , Feces/chemistry , Feces/microbiology , Female , Human Experimentation , Humans , Immunoglobulin A/analysis , Immunoglobulin G/blood , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...