Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 184(13): 3486-3501.e21, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34077751

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a World Health Organization priority pathogen. CCHFV infections cause a highly lethal hemorrhagic fever for which specific treatments and vaccines are urgently needed. Here, we characterize the human immune response to natural CCHFV infection to identify potent neutralizing monoclonal antibodies (nAbs) targeting the viral glycoprotein. Competition experiments showed that these nAbs bind six distinct antigenic sites in the Gc subunit. These sites were further delineated through mutagenesis and mapped onto a prefusion model of Gc. Pairwise screening identified combinations of non-competing nAbs that afford synergistic neutralization. Further enhancements in neutralization breadth and potency were attained by physically linking variable domains of synergistic nAb pairs through bispecific antibody (bsAb) engineering. Although multiple nAbs protected mice from lethal CCHFV challenge in pre- or post-exposure prophylactic settings, only a single bsAb, DVD-121-801, afforded therapeutic protection. DVD-121-801 is a promising candidate suitable for clinical development as a CCHFV therapeutic.


Subject(s)
Antibodies, Neutralizing/immunology , Hemorrhagic Fever, Crimean/immunology , Survivors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigens, Viral/metabolism , Biophysical Phenomena , Chlorocebus aethiops , Epitope Mapping , Epitopes/metabolism , Female , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/prevention & control , Humans , Immunoglobulin G/metabolism , Male , Mice , Neutralization Tests , Protein Binding , Protein Engineering , Recombinant Proteins/immunology , Vero Cells , Viral Proteins/chemistry
2.
Cell ; 169(5): 878-890.e15, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525755

ABSTRACT

Experimental monoclonal antibody (mAb) therapies have shown promise for treatment of lethal Ebola virus (EBOV) infections, but their species-specific recognition of the viral glycoprotein (GP) has limited their use against other divergent ebolaviruses associated with human disease. Here, we mined the human immune response to natural EBOV infection and identified mAbs with exceptionally potent pan-ebolavirus neutralizing activity and protective efficacy against three virulent ebolaviruses. These mAbs recognize an inter-protomer epitope in the GP fusion loop, a critical and conserved element of the viral membrane fusion machinery, and neutralize viral entry by targeting a proteolytically primed, fusion-competent GP intermediate (GPCL) generated in host cell endosomes. Only a few somatic hypermutations are required for broad antiviral activity, and germline-approximating variants display enhanced GPCL recognition, suggesting that such antibodies could be elicited more efficiently with suitably optimized GP immunogens. Our findings inform the development of both broadly effective immunotherapeutics and vaccines against filoviruses.


Subject(s)
Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/immunology , Survivors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Chlorocebus aethiops , Cross Reactions , Ebolavirus/classification , Ebolavirus/immunology , Female , Ferrets , Hemorrhagic Fever, Ebola/virology , Humans , Kinetics , Mice , Mice, Inbred BALB C , Models, Molecular , Sequence Alignment , Vero Cells
3.
Proc Natl Acad Sci U S A ; 117(7): 3768-3778, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015126

ABSTRACT

Antibody-based therapies are a promising treatment option for managing ebolavirus infections. Several Ebola virus (EBOV)-specific and, more recently, pan-ebolavirus antibody cocktails have been described. Here, we report the development and assessment of a Sudan virus (SUDV)-specific antibody cocktail. We produced a panel of SUDV glycoprotein (GP)-specific human chimeric monoclonal antibodies (mAbs) using both plant and mammalian expression systems and completed head-to-head in vitro and in vivo evaluations. Neutralizing activity, competitive binding groups, and epitope specificity of SUDV mAbs were defined before assessing protective efficacy of individual mAbs using a mouse model of SUDV infection. Of the mAbs tested, GP base-binding mAbs were more potent neutralizers and more protective than glycan cap- or mucin-like domain-binding mAbs. No significant difference was observed between plant and mammalian mAbs in any of our in vitro or in vivo evaluations. Based on in vitro and rodent testing, a combination of two SUDV-specific mAbs, one base binding (16F6) and one glycan cap binding (X10H2), was down-selected for assessment in a macaque model of SUDV infection. This cocktail, RIID F6-H2, provided protection from SUDV infection in rhesus macaques when administered at 50 mg/kg on days 4 and 6 postinfection. RIID F6-H2 is an effective postexposure SUDV therapy and provides a potential treatment option for managing human SUDV infection.


Subject(s)
Antibodies, Viral/administration & dosage , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/drug therapy , Animals , Antibodies, Monoclonal/administration & dosage , Disease Models, Animal , Ebolavirus/genetics , Female , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Immunotherapy , Macaca mulatta , Male , Mice , Viral Proteins/immunology
4.
J Biol Chem ; 293(16): 6201-6211, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29500195

ABSTRACT

Filoviruses (family Filoviridae) include five ebolaviruses and Marburg virus. These pathogens cause a rapidly progressing and severe viral disease with high mortality rates (generally 30-90%). Outbreaks of filovirus disease are sporadic and, until recently, were limited to less than 500 cases. However, the 2013-2016 epidemic in western Africa, caused by Ebola virus (EBOV), illustrated the potential of filovirus outbreaks to escalate to a much larger scale (over 28,000 suspected cases). mAbs against the envelope glycoprotein represent a promising therapeutic platform for managing filovirus infections. However, mAbs that exhibit neutralization or protective properties against multiple filoviruses are rare. Here we examined a panel of engineered bi- and trispecific antibodies, in which variable domains of mAbs that target epitopes from multiple filoviruses were combined, for their capacity to neutralize viral infection across filovirus species. We found that bispecific combinations targeting EBOV and Sudan virus (another ebolavirus), provide potent cross-neutralization and protection in mice. Furthermore, trispecific combinations, targeting EBOV, Sudan virus, and Marburg virus, exhibited strong neutralization potential against all three viruses. These results provide important insights into multispecific antibody engineering against filoviruses and will inform future immunotherapeutic discoveries.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Marburgvirus/immunology , Protein Engineering , Viral Proteins/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/genetics , Epitopes/immunology , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics
5.
J Infect Dis ; 212 Suppl 2: S282-94, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25943199

ABSTRACT

A major obstacle in ebolavirus research is the lack of a small-animal model for Sudan virus (SUDV), as well as other wild-type (WT) ebolaviruses. Here, we expand on research by Bray and by Lever et al suggesting that WT ebolaviruses are pathogenic in mice deficient for the type 1 interferon (IFN) α/ß receptor (IFNα/ßR-/-). We examined the disease course of several WT ebolaviruses: Boneface (SUDV/Bon) and Gulu variants of SUDV, Ebola virus (EBOV), Bundibugyo virus (BDBV), Taï Forest virus, and Reston virus (RESTV). We determined that exposure to WT SUDV or EBOV results in reproducible signs of disease in IFNα/ßR-/- mice, as measured by weight loss and partial lethality. Vaccination with the SUDV or EBOV glycoprotein (GP)-expressing Venezuelan equine encephalitis viral replicon particle vaccine protected these mice from SUDV/Bon and EBOV challenge, respectively. Treatment with SUDV- or EBOV-specific anti-GP antibodies protected mice from challenge when delivered 1-3 days after infection. Serial sampling experiments revealed evidence of disseminated intravascular coagulation in the livers of mice infected with the Boneface variant of SUDV, EBOV, and BDBV. Taken together, these data solidify the IFNα/ßR-/- mouse as an important and useful model for the study of WT EBOV disease.


Subject(s)
Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Receptor, Interferon alpha-beta/deficiency , Virulence/physiology , Animals , Antibodies, Viral/immunology , Cell Line , Chlorocebus aethiops , Disease Models, Animal , Ebola Vaccines/immunology , Ebolavirus/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Replicon/immunology , Vaccination/methods , Vero Cells/virology , Viral Proteins/immunology , Viral Proteins/metabolism , Virulence/immunology
6.
J Virol ; 88(20): 12077-86, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25122801

ABSTRACT

Alphavirus replicons were evaluated as potential vaccine candidates for Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), or eastern equine encephalitis virus (EEEV) when given individually or in combination (V/W/E) to mice or cynomolgus macaques. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in mice to their respective alphavirus. Protection from either subcutaneous or aerosol challenge with VEEV, WEEV, or EEEV was demonstrated out to 12 months after vaccination in mice. Individual replicon vaccines or the combination V/W/E replicon vaccine elicited strong neutralizing antibodies in macaques and demonstrated good protection against aerosol challenge with an epizootic VEEV-IAB virus, Trinidad donkey. Similarly, the EEEV replicon and V/W/E combination vaccine elicited neutralizing antibodies against EEEV and protected against aerosol exposure to a North American variety of EEEV. Both the WEEV replicon and combination V/W/E vaccination, however, elicited poor neutralizing antibodies to WEEV in macaques, and the protection conferred was not as strong. These results demonstrate that a combination V/W/E vaccine is possible for protection against aerosol challenge and that cross-interference between the vaccines is minimal. Importance: Three related viruses belonging to the genus Alphavirus cause severe encephalitis in humans: Venezuelan equine encephalitis virus (VEEV), western equine encephalitis virus (WEEV), and eastern equine encephalitis virus (EEEV). Normally transmitted by mosquitoes, these viruses can cause disease when inhaled, so there is concern that these viruses could be used as biological weapons. Prior reports have suggested that vaccines for these three viruses might interfere with one another. We have developed a combined vaccine for Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis expressing the surface proteins of all three viruses. In this report we demonstrate in both mice and macaques that this combined vaccine is safe, generates a strong immune response, and protects against aerosol challenge with the viruses that cause Venezuelan equine encephalitis, western equine encephalitis, and eastern equine encephalitis.


Subject(s)
Alphavirus/immunology , Antibodies, Neutralizing/immunology , Encephalitis Virus, Eastern Equine/immunology , Replicon , Viral Vaccines/immunology , Alphavirus/classification , Animals , Blotting, Western , Chlorocebus aethiops , Cricetinae , Encephalitis Virus, Eastern Equine/classification , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Macaca fascicularis , Male , Mice , Vero Cells
7.
Virol J ; 12: 152, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26420265

ABSTRACT

BACKGROUND: Eastern equine encephalitis virus (EEEV), an arbovirus, is an important human and veterinary pathogen belonging to one of seven antigenic complexes in the genus Alphavirus, family Togaviridae. EEEV is considered the most deadly of the mosquito-borne alphaviruses due to the high case fatality rate associated with clinical infections, reaching up to 75 % in humans and 90 % in horses. In patients that survive acute infection, neurologic sequelae are often devastating. Although natural infections are acquired by mosquito bite, EEEV is also highly infectious by aerosol. This fact, along with the relative ease of production and stability of this virus, has led it to being identified as a potential agent of bioterrorism. METHODS: To characterize the clinical course and outcome of EEEV strain FL93-939 infection, we compared clinical parameters, cytokine expression, viremia, and viral titers in numerous tissues of mice exposed by various routes. Twelve-week-old female BALB/c mice were infected by the intranasal, aerosol, or subcutaneous route. Mice were monitored for clinical signs of disease and euthanized at specified time points (6 hpi through 8 dpi). Blood and tissues were harvested for cytokine analysis and/or viral titer determination. RESULTS: Although all groups of animals exhibited similar clinical signs after inoculation, the onset and severity differed. The majority of those animals exposed by the aerosol route developed severe clinical signs by 4 dpi. Significant differences were also observed in the viral titers of target tissues, with virus being detected in the brain at 6 hpi in the aerosol study. CONCLUSION: The clinical course and outcome of EEEV infection in mice is dependent on route of exposure. Aerosol exposure to EEEV results in acute onset of clinical signs, rapid neuroinvasion, and 100 % mortality.


Subject(s)
Alphavirus Infections/pathology , Disease Models, Animal , Encephalitis Virus, Eastern Equine/growth & development , Encephalitis Virus, Eastern Equine/pathogenicity , Administration, Inhalation , Administration, Intranasal , Alphavirus Infections/virology , Animal Structures/pathology , Animal Structures/virology , Animals , Body Fluids/virology , Cytokines/analysis , Female , Injections, Subcutaneous , Mice, Inbred BALB C , Survival Analysis , Viral Load
8.
Virol J ; 12: 154, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26423229

ABSTRACT

BACKGROUND: Eastern equine encephalitis virus (EEEV) is an alphavirus with a case fatality rate estimated to be as high as 75 % in humans and 90 % in horses. Surviving patients often have long-lasting and severe neurological sequelae. At present, there is no licensed vaccine or therapeutic for EEEV infection. This study completes the clinical and pathological analysis of mice infected with a North American strain of EEEV by three different routes: aerosol, intranasal, and subcutaneous. Such an understanding is imperative for use of the mouse model in vaccine and antiviral drug development. METHODS: Twelve-week-old female BALB/c mice were infected with EEEV strain FL93-939 by the intranasal, aerosol, or subcutaneous route. Mice were euthanized 6 hpi through 8 dpi and tissues were harvested for histopathological and immunohistochemical analysis. RESULTS: Viral antigen was detected in the olfactory bulb as early as 1-2 dpi in aerosol and intranasal infected mice. However, histologic lesions in the brain were evident about 24 hours earlier (3 dpi vs 4 dpi), and were more pronounced following aerosol infection relative to intranasal infection. Following subcutaneous infection, viral antigen was also detected in the olfactory bulb, though not as routinely or as early. Significant histologic lesions were not observed until 6 dpi. CONCLUSION: These pathologic studies suggest EEEV enters the brain through the olfactory system when mice are exposed via the intranasal and aerosol routes. In contrast, the histopathologic lesions were delayed in the subcutaneous group and it appears the virus may utilize both the vascular and olfactory routes to enter the brain when mice are exposed to EEEV subcutaneously.


Subject(s)
Alphavirus Infections/pathology , Alphavirus Infections/virology , Disease Models, Animal , Encephalitis Virus, Eastern Equine/growth & development , Encephalitis Virus, Eastern Equine/physiology , Administration, Inhalation , Administration, Intranasal , Animal Structures/pathology , Animal Structures/virology , Animals , Female , Histocytochemistry , Immunohistochemistry , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Microscopy
9.
J Virol ; 87(9): 4952-64, 2013 May.
Article in English | MEDLINE | ID: mdl-23408633

ABSTRACT

There are no vaccines or therapeutics currently approved for the prevention or treatment of ebolavirus infection. Previously, a replicon vaccine based on Venezuelan equine encephalitis virus (VEEV) demonstrated protective efficacy against Marburg virus in nonhuman primates. Here, we report the protective efficacy of Sudan virus (SUDV)- and Ebola virus (EBOV)-specific VEEV replicon particle (VRP) vaccines in nonhuman primates. VRP vaccines were developed to express the glycoprotein (GP) of either SUDV or EBOV. A single intramuscular vaccination of cynomolgus macaques with VRP expressing SUDV GP provided complete protection against intramuscular challenge with SUDV. Vaccination against SUDV and subsequent survival of SUDV challenge did not fully protect cynomolgus macaques against intramuscular EBOV back-challenge. However, a single simultaneous intramuscular vaccination with VRP expressing SUDV GP combined with VRP expressing EBOV GP did provide complete protection against intramuscular challenge with either SUDV or EBOV in cynomolgus macaques. Finally, intramuscular vaccination with VRP expressing SUDV GP completely protected cynomolgus macaques when challenged with aerosolized SUDV, although complete protection against aerosol challenge required two vaccinations with this vaccine.


Subject(s)
Ebolavirus/immunology , Encephalitis Virus, Venezuelan Equine/genetics , Hemorrhagic Fever, Ebola/prevention & control , Replicon , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Ebolavirus/genetics , Encephalitis Virus, Venezuelan Equine/physiology , Genetic Vectors/genetics , Genetic Vectors/physiology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/virology , Humans , Macaca fascicularis , Vaccination , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
10.
Methods Protoc ; 7(3)2024 May 15.
Article in English | MEDLINE | ID: mdl-38804336

ABSTRACT

Venezuelan (VEE), eastern (EEE), and western (WEE) equine encephalitis viruses are encephalitic New World alphaviruses that cause periodic epizootic and epidemic outbreaks in horses and humans that may cause severe morbidity and mortality. Currently there are no FDA-licensed vaccines or effective antiviral therapies. Each year, there are a limited number of human cases of encephalitic alphaviruses; thus, licensure of a vaccine or therapeutic would require approval under the FDA animal rule. Approval under the FDA animal rule requires the disease observed in the animal model to recapitulate what is observed in humans. Currently, initial testing of vaccines and therapeutics is performed in the mouse model. Unfortunately, alphavirus disease manifestations in a mouse do not faithfully recapitulate human disease; the VEEV mouse model is lethal whereas in humans VEEV is rarely lethal. In an effort to identify a more appropriate small animal model, we evaluated hamsters in an aerosol exposure model of encephalitic alphavirus infection. The pathology, lethality, and viremia observed in the infected hamsters was inconsistent with what is observed in NHP models and humans. These data suggest that hamsters are not an appropriate model for encephalitic alphaviruses to test vaccines or potential antiviral therapies.

11.
bioRxiv ; 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38659837

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that can cause severe disease in humans with case fatality rates of 10-40%. Although structures of CCHFV glycoproteins GP38 and Gc have provided insights into viral entry and defined epitopes of neutralizing and protective antibodies, the structure of glycoprotein Gn and its interactions with GP38 and Gc have remained elusive. Here, we used structure-guided protein engineering to produce a stabilized GP38-Gn-Gc heterotrimeric glycoprotein complex (GP38-GnH-DS-Gc). A cryo-EM structure of this complex provides the molecular basis for GP38's association on the viral surface, reveals the structure of Gn, and demonstrates that GP38-Gn restrains the Gc fusion loops in the prefusion conformation, facilitated by an N-linked glycan attached to Gn. Immunization with GP38-GnH-DS-Gc conferred 40% protection against lethal IbAr10200 challenge in mice. These data define the architecture of a GP38-Gn-Gc protomer and provide a template for structure-guided vaccine antigen development.

12.
bioRxiv ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38826290

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a priority pathogen transmitted by tick bites, with no vaccines or specific therapeutics approved to date. Severe disease manifestations include hemorrhage, endothelial dysfunction, and multiorgan failure. Infected cells secrete the viral glycoprotein GP38, whose extracellular function is presently unknown. GP38 is considered an important target for vaccine and therapeutic design as GP38-specific antibodies can protect against severe disease in animal models, albeit through a currently unknown mechanism of action. Here, we show that GP38 induces endothelial barrier dysfunction in vitro, and that CCHFV infection, and GP38 alone, can trigger vascular leak in a mouse model. Protective antibodies that recognize specific antigenic sites on GP38, but not a protective neutralizing antibody binding the structural protein Gc, potently inhibit endothelial hyperpermeability in vitro and vascular leak in vivo during CCHFV infection. This work uncovers a function of the secreted viral protein GP38 as a viral toxin in CCHFV pathogenesis and elucidates the mode of action of non-neutralizing GP38-specific antibodies.

13.
Cell Rep ; 43(7): 114502, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39002130

ABSTRACT

Crimean-Congo hemorrhagic fever virus can cause lethal disease in humans yet there are no approved medical countermeasures. Viral glycoprotein GP38, exclusive to Nairoviridae, is a target of protective antibodies and is a key antigen in preclinical vaccine candidates. Here, we isolate 188 GP38-specific antibodies from human survivors of infection. Competition experiments show that these antibodies bind across 5 distinct antigenic sites, encompassing 11 overlapping regions. Additionally, we show structures of GP38 bound with 9 of these antibodies targeting different antigenic sites. Although these GP38-specific antibodies are non-neutralizing, several display protective efficacy equal to or better than murine antibody 13G8 in two highly stringent rodent models of infection. Together, these data expand our understanding regarding this important viral protein and may inform the development of broadly effective CCHFV antibody therapeutics.


Subject(s)
Antibodies, Viral , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Humans , Animals , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Antibodies, Viral/immunology , Mice , Survivors , Antibodies, Neutralizing/immunology , Female , Glycoproteins/immunology , Epitopes/immunology
14.
bioRxiv ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38496658

ABSTRACT

Crimean-Congo hemorrhagic fever virus can cause lethal disease in humans yet there are no approved medical countermeasures. Viral glycoprotein GP38, unique to Nairoviridae, is a target of protective antibodies, but extensive mapping of the human antibody response to GP38 has not been previously performed. Here, we isolated 188 GP38-specific antibodies from human survivors of infection. Competition experiments showed that these antibodies bind across five distinct antigenic sites, encompassing eleven overlapping regions. Additionally, we reveal structures of GP38 bound with nine of these antibodies targeting different antigenic sites. Although GP38-specific antibodies were non-neutralizing, several antibodies were found to have protection equal to or better than murine antibody 13G8 in two highly stringent rodent models of infection. Together, these data expand our understanding regarding this important viral protein and inform the development of broadly effective CCHFV antibody therapeutics.

15.
Nat Commun ; 14(1): 4454, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488123

ABSTRACT

Andes virus (ANDV) and Sin Nombre virus (SNV) are the etiologic agents of severe hantavirus cardiopulmonary syndrome (HCPS) in the Americas for which no FDA-approved countermeasures are available. Protocadherin-1 (PCDH1), a cadherin-superfamily protein recently identified as a critical host factor for ANDV and SNV, represents a new antiviral target; however, its precise role remains to be elucidated. Here, we use computational and experimental approaches to delineate the binding surface of the hantavirus glycoprotein complex on PCDH1's first extracellular cadherin repeat domain. Strikingly, a single amino acid residue in this PCDH1 surface influences the host species-specificity of SNV glycoprotein-PCDH1 interaction and cell entry. Mutation of this and a neighboring residue substantially protects Syrian hamsters from pulmonary disease and death caused by ANDV. We conclude that PCDH1 is a bona fide entry receptor for ANDV and SNV whose direct interaction with hantavirus glycoproteins could be targeted to develop new interventions against HCPS.


Subject(s)
Communicable Diseases , Orthohantavirus , RNA Viruses , Animals , Cricetinae , Point Mutation , Protocadherins , Cadherins , Mesocricetus , Syndrome
16.
PLoS Pathog ; 6(10): e1001146, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20976195

ABSTRACT

We have developed a high-resolution genomic mapping technique that combines transposon-mediated insertional mutagenesis with either capillary electrophoresis or massively parallel sequencing to identify functionally important regions of the Venezuelan equine encephalitis virus (VEEV) genome. We initially used a capillary electrophoresis method to gain insight into the role of the VEEV nonstructural protein 3 (nsP3) in viral replication. We identified several regions in nsP3 that are intolerant to small (15 bp) insertions, and thus are presumably functionally important. We also identified nine separate regions in nsP3 that will tolerate small insertions at low temperatures (30°C), but not at higher temperatures (37°C, and 40°C). Because we found this method to be extremely effective at identifying temperature sensitive (ts) mutations, but limited by capillary electrophoresis capacity, we replaced the capillary electrophoresis with massively parallel sequencing and used the improved method to generate a functional map of the entire VEEV genome. We identified several hundred potential ts mutations throughout the genome and we validated several of the mutations in nsP2, nsP3, E3, E2, E1 and capsid using single-cycle growth curve experiments with virus generated through reverse genetics. We further demonstrated that two of the nsP3 ts mutants were attenuated for virulence in mice but could elicit protective immunity against challenge with wild-type VEEV. The recombinant ts mutants will be valuable tools for further studies of VEEV replication and virulence. Moreover, the method that we developed is applicable for generating such tools for any virus with a robust reverse genetics system.


Subject(s)
Chromosome Mapping/methods , Encephalitis Virus, Venezuelan Equine/genetics , Genome, Viral/genetics , Mutagenesis, Insertional/methods , Sequence Analysis, DNA/methods , Animals , Cells, Cultured , Chlorocebus aethiops , Encephalitis Virus, Venezuelan Equine/physiology , High-Throughput Nucleotide Sequencing/methods , Mice , Mice, Inbred BALB C , Sequence Homology, Nucleic Acid , Vero Cells , Virulence/genetics , Virus Replication/genetics
17.
Sci Transl Med ; 14(636): eabl5399, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35294259

ABSTRACT

The rodent-borne hantavirus Puumala virus (PUUV) and related agents cause hemorrhagic fever with renal syndrome (HFRS) in humans. Other hantaviruses, including Andes virus (ANDV) and Sin Nombre virus, cause a distinct zoonotic disease, hantavirus cardiopulmonary syndrome (HCPS). Although these infections are severe and have substantial case fatality rates, no FDA-approved hantavirus countermeasures are available. Recent work suggests that monoclonal antibodies may have therapeutic utility. We describe here the isolation of human neutralizing antibodies (nAbs) against tetrameric Gn/Gc glycoprotein spikes from PUUV-experienced donors. We define a dominant class of nAbs recognizing the "capping loop" of Gn that masks the hydrophobic fusion loops in Gc. A subset of nAbs in this class, including ADI-42898, bound Gn/Gc complexes but not Gn alone, strongly suggesting that they recognize a quaternary epitope encompassing both Gn and Gc. ADI-42898 blocked the cell entry of seven HCPS- and HFRS-associated hantaviruses, and single doses of this nAb could protect Syrian hamsters and bank voles challenged with the highly virulent HCPS-causing ANDV and HFRS-causing PUUV, respectively. ADI-42898 is a promising candidate for clinical development as a countermeasure for both HCPS and HFRS, and its mode of Gn/Gc recognition informs the development of broadly protective hantavirus vaccines.


Subject(s)
Hantavirus Infections , Hemorrhagic Fever with Renal Syndrome , Orthohantavirus , Puumala virus , Animals , Antibodies, Neutralizing , Antibodies, Viral , Cricetinae , Epitopes , Glycoproteins , Hemorrhagic Fever with Renal Syndrome/prevention & control , Humans
18.
Antiviral Res ; 181: 104854, 2020 09.
Article in English | MEDLINE | ID: mdl-32621945

ABSTRACT

Recent studies highlight that infection with Coxsackievirus B3, Venezuelan equine encephalitis virus (VEEV), Marburg virus, or stimulation using poly I:C (dsRNA), upregulates the signaling adaptor protein MyD88 and impairs the host antiviral type I interferon (IFN) responses. In contrast, MyD88 deficiency (MyD88-/-) increases the type I IFN and survivability of mice implying that MyD88 up regulation limits the type I IFN response. Reasoning that MyD88 inhibition in a virus-like manner may increase type I IFN responses, our studies revealed lipopolysaccharide stimulation of U937 cells or poly I:C stimulation of HEK293-TLR3, THP1 or U87 cells in the presence of a previously reported MyD88 inhibitor (compound 4210) augmented IFN-ß and RANTES production. Consistent with these results, overexpression of MyD88 decreased IFN-ß, whereas MyD88 inhibition rescued IFN-ß production concomitant with increased IRF3 phosphorylation, suggesting IRF-mediated downstream signaling to the IFN-ß response. Further, compound 4210 treatment inhibited MyD88 interaction with IRF3/IRF7 indicating that MyD88 restricts type I IFN signaling through sequestration of IRF3/IRF7. In cell based infection assays, compound 4210 treatment suppressed replication of VEEV, Eastern equine encephalitis virus, Ebola virus (EBOV), Rift Valley Fever virus, Lassa virus, and Dengue virus with IC50 values ranging from 11 to 42 µM. Notably, administration of compound 4210 improved survival, weight change, and clinical disease scores in mice following challenge with VEEV TC-83 and EBOV. Collectively, these results provide evidence that viral infections responsive to MyD88 inhibition lead to activation of IRF3/IRF7 and promoted a type I IFN response, thus, raising the prospect of an approach of host-directed antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Interferon Type I/genetics , Myeloid Differentiation Factor 88/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Viruses/drug effects , Animals , HEK293 Cells , Humans , Inhibitory Concentration 50 , Interferon Regulatory Factor-3/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Phosphorylation , Poly I-C/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/chemical synthesis , THP-1 Cells , Virus Diseases/drug therapy , Virus Replication/drug effects , Viruses/classification
19.
Cell Host Microbe ; 25(1): 39-48.e5, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30629917

ABSTRACT

Passive administration of monoclonal antibodies (mAbs) is a promising therapeutic approach for Ebola virus disease (EVD). However, all mAbs and mAb cocktails that have entered clinical development are specific for a single member of the Ebolavirus genus, Ebola virus (EBOV), and ineffective against outbreak-causing Bundibugyo virus (BDBV) and Sudan virus (SUDV). Here, we advance MBP134, a cocktail of two broadly neutralizing human mAbs, ADI-15878 from an EVD survivor and ADI-23774 from the same survivor but specificity-matured for SUDV GP binding affinity, as a candidate pan-ebolavirus therapeutic. MBP134 potently neutralized all ebolaviruses and demonstrated greater protective efficacy than ADI-15878 alone in EBOV-challenged guinea pigs. A second-generation cocktail, MBP134AF, engineered to effectively harness natural killer (NK) cells afforded additional improvement relative to its precursor in protective efficacy against EBOV and SUDV in guinea pigs. MBP134AF is an optimized mAb cocktail suitable for evaluation as a pan-ebolavirus therapeutic in nonhuman primates.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animal Welfare , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/administration & dosage , Antibodies, Viral/therapeutic use , Antiviral Agents , Disease Models, Animal , Ebolavirus/pathogenicity , Epitopes/immunology , Female , Filoviridae/immunology , Guinea Pigs , Hemorrhagic Fever, Ebola/virology , Humans , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Recombinant Proteins/immunology , Treatment Outcome
20.
Vaccine ; 36(22): 3090-3100, 2018 05 24.
Article in English | MEDLINE | ID: mdl-28216187

ABSTRACT

Infections with filoviruses in humans are highly virulent, causing hemorrhagic fevers which result in up to 90% mortality. In addition to natural infections, the ability to use these viruses as bioterrorist weapons is of significant concern. Currently, there are no licensed vaccines or therapeutics available to combat these infections. The pathogenesis of disease involves the dysregulation of the host's immune system, which results in impairment of the innate and adaptive immune responses, with subsequent development of lymphopenia, thrombocytopenia, hemorrhage, and death. Questions remain with regard to the few survivors of infection, who manage to mount an effective adaptive immune response. These questions concern the humoral and cellular components of this response, and whether such a response can be elicited by an appropriate prophylactic vaccine. The data reported herein describe the production and evaluation of a recombinant subunit Ebola virus vaccine candidate consisting of insect cell expressed Zaire ebolavirus (EBOV) surface glycoprotein (GP) and the matrix proteins VP24 and VP40. The recombinant subunit proteins are shown to be highly immunogenic in mice, yielding both humoral and cellular responses, as well as highly efficacious, providing up to 100% protection against a lethal challenge with live virus. These results demonstrate proof of concept for such a recombinant non-replicating vaccine candidate in the mouse model of EBOV which helps to elucidate immune correlates of protection and warrants further development.


Subject(s)
Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Viral Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Ebolavirus , Female , Hemorrhagic Fever, Ebola/immunology , Immunity, Cellular , Immunity, Humoral , Mice , Mice, Inbred BALB C , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL