55 results on '"Joshua D. Shamblin"'
Search Results
2. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease
- Author
-
Joseph W. Golden, Curtis R. Cline, Xiankun Zeng, Aura R. Garrison, Brian D. Carey, Eric M. Mucker, Lauren E. White, Joshua D. Shamblin, Rebecca L. Brocato, Jun Liu, April M. Babka, Hypaitia B. Rauch, Jeffrey M. Smith, Bradley S. Hollidge, Collin Fitzpatrick, Catherine V. Badger, and Jay W. Hooper
- Subjects
COVID-19 ,Infectious disease ,Medicine - Abstract
The emergence of SARS-CoV-2 has created an international health crisis, and small animal models mirroring SARS-CoV-2 human disease are essential for medical countermeasure (MCM) development. Mice are refractory to SARS-CoV-2 infection owing to low-affinity binding to the murine angiotensin-converting enzyme 2 (ACE2) protein. Here, we evaluated the pathogenesis of SARS-CoV-2 in male and female mice expressing the human ACE2 gene under the control of the keratin 18 promoter (K18). In contrast to nontransgenic mice, intranasal exposure of K18-hACE2 animals to 2 different doses of SARS-CoV-2 resulted in acute disease, including weight loss, lung injury, brain infection, and lethality. Vasculitis was the most prominent finding in the lungs of infected mice. Transcriptomic analysis from lungs of infected animals showed increases in transcripts involved in lung injury and inflammatory cytokines. In the low-dose challenge groups, there was a survival advantage in the female mice, with 60% surviving infection, whereas all male mice succumbed to disease. Male mice that succumbed to disease had higher levels of inflammatory transcripts compared with female mice. To our knowledge, this is the first highly lethal murine infection model for SARS-CoV-2 and should be valuable for the study of SARS-CoV-2 pathogenesis and for the assessment of MCMs.
- Published
- 2020
- Full Text
- View/download PDF
3. Virus-encoded miRNAs in Ebola virus disease
- Author
-
Janice Duy, Anna N. Honko, Louis A. Altamura, Sandra L. Bixler, Suzanne Wollen-Roberts, Nadia Wauquier, Aileen O’Hearn, Eric M. Mucker, Joshua C. Johnson, Joshua D. Shamblin, Justine Zelko, Miriam A. Botto, James Bangura, Moinya Coomber, M. Louise Pitt, Jean-Paul Gonzalez, Randal J. Schoepp, Arthur J. Goff, and Timothy D. Minogue
- Subjects
Medicine ,Science - Abstract
Abstract Ebola virus (EBOV) is a negative-strand RNA virus that replicates in the cytoplasm and causes an often-fatal hemorrhagic fever. EBOV, like other viruses, can reportedly encode its own microRNAs (miRNAs) to subvert host immune defenses. miRNAs are short noncoding RNAs that can regulate gene expression by hybridizing to multiple mRNAs, and viral miRNAs can enhance viral replication and infectivity by regulating host or viral genes. To date, only one EBOV miRNA has been examined in human infection. Here, we assayed mouse, rhesus macaque, cynomolgus macaque, and human samples infected with three EBOV variants for twelve computationally predicted viral miRNAs using RT-qPCR. Ten miRNAs aligned to EBOV variants and were detectable in the four species during disease with several viral miRNAs showing presymptomatic amplification in animal models. miRNA abundances in both the mouse and nonhuman primate models mirrored the human cohort, with miR-1-5p, miR-1-3p, and miR-T3-3p consistently at the highest levels. These striking similarities in the most abundant miRNAs during infection with different EBOV variants and hosts indicate that these miRNAs are potential valuable diagnostic markers and key effectors of EBOV pathogenesis.
- Published
- 2018
- Full Text
- View/download PDF
4. A DNA vaccine delivered by dermal electroporation fully protects cynomolgus macaques against Lassa fever
- Author
-
Kathleen A. Cashman, Eric R. Wilkinson, Carl I. Shaia, Paul R. Facemire, Todd M. Bell, Jeremy J. Bearss, Joshua D. Shamblin, Suzanne E. Wollen, Kate E. Broderick, Niranjan Y. Sardesai, and Connie S. Schmaljohn
- Subjects
lassa fever ,dna vaccine ,electroporation ,Immunologic diseases. Allergy ,RC581-607 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Lassa virus (LASV) is an ambisense RNA virus in the Arenaviridae family and is the etiological agent of Lassa fever, a severe hemorrhagic disease endemic to West and Central Africa. There are no US Food and Drug Administration (FDA)-licensed vaccines available to prevent Lassa fever. in our previous studies, we developed a gene-optimized DNA vaccine that encodes the glycoprotein precursor gene of LASV (Josiah strain) and demonstrated that 3 vaccinations accompanied by dermal electroporation protected guinea pigs from LASV-associated illness and death. Here, we describe an initial efficacy experiment in cynomolgus macaque nonhuman primates (NHPs) in which we followed an identical 3-dose vaccine schedule that was successful in guinea pigs, and a follow-on experiment in which we used an accelerated vaccination strategy consisting of 2 administrations, spaced 4 weeks apart. In both studies, all of the LASV DNA-vaccinated NHPs survived challenge and none of them had measureable, sustained viremia or displayed weight loss or other disease signs post-exposure. Three of 10 mock-vaccinates survived exposure to LASV, but all of them became acutely ill post-exposure and remained chronically ill to the study end point (45 d post-exposure). Two of the 3 survivors experienced sensorineural hearing loss (described elsewhere). These results clearly demonstrate that the LASV DNA vaccine combined with dermal electroporation is a highly effective candidate for eventual use in humans.
- Published
- 2017
- Full Text
- View/download PDF
5. DNA vaccines elicit durable protective immunity against individual or simultaneous infections with Lassa and Ebola viruses in guinea pigs
- Author
-
Kathleen A. Cashman, Eric R. Wilkinson, Suzanne E. Wollen, Joshua D. Shamblin, Justine M. Zelko, Jeremy J. Bearss, Xiankun Zeng, Kate E. Broderick, and Connie S. Schmaljohn
- Subjects
lassa fever ,ebola virus ,lassa virus ,viral hemorrhagic fever ,hemorrhagic fever ,biodefense ,dna vaccines ,guinea pig ,multiagent vaccine ,Immunologic diseases. Allergy ,RC581-607 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
We previously developed optimized DNA vaccines against both Lassa fever and Ebola hemorrhagic fever viruses and demonstrated that they were protective individually in guinea pig and nonhuman primate models. In this study, we vaccinated groups of strain 13 guinea pigs two times, four weeks apart with 50 µg of each DNA vaccine or a mock vaccine at discrete sites by intradermal electroporation. Five weeks following the second vaccinations, guinea pigs were exposed to lethal doses of Lassa virus, Ebola virus, or a combination of both viruses simultaneously. None of the vaccinated guinea pigs, regardless of challenge virus and including the coinfected group, displayed weight loss, fever or other disease signs, and all survived to the study endpoint. All of the mock-vaccinated guinea pigs that were infected with Lassa virus, and all but one of the EBOV-infected mock-vaccinated guinea pigs succumbed. In order to determine if the dual-agent vaccination strategy could protect against both viruses if exposures were temporally separated, we held the surviving vaccinates in BSL-4 for approximately 120 days to perform a cross-challenge experiment in which guinea pigs originally infected with Lassa virus received a lethal dose of Ebola virus and those originally infected with Ebola virus were infected with a lethal dose of Lassa virus. All guinea pigs remained healthy and survived to the study endpoint. This study clearly demonstrates that DNA vaccines against Lassa and Ebola viruses can elicit protective immunity against both individual virus exposures as well as in a mixed-infection environment.
- Published
- 2017
- Full Text
- View/download PDF
6. High Infection Rates for Adult Macaques after Intravaginal or Intrarectal Inoculation with Zika Virus
- Author
-
Andrew D. Haddow, Aysegul Nalca, Franco D. Rossi, Lynn J. Miller, Michael R. Wiley, Unai Perez-Sautu, Samuel C. Washington, Sarah L. Norris, Suzanne E. Wollen-Roberts, Joshua D. Shamblin, Adrienne E. Kimmel, Holly A. Bloomfield, Stephanie M. Valdez, Thomas R. Sprague, Lucia M. Principe, Stephanie A. Bellanca, Stephanie S. Cinkovich, Luis Lugo-Roman, Lisa H. Cazares, William D. Pratt, Gustavo Palacios, Sina Bavari, M. Louise Pitt, and Farooq Nasar
- Subjects
Zika virus ,viruses ,arbovirus ,infection rates ,macaques ,intravaginal inoculation ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Unprotected sexual intercourse between persons residing in or traveling from regions with Zika virus transmission is a risk factor for infection. To model risk for infection after sexual intercourse, we inoculated rhesus and cynomolgus macaques with Zika virus by intravaginal or intrarectal routes. In macaques inoculated intravaginally, we detected viremia and virus RNA in 50% of macaques, followed by seroconversion. In macaques inoculated intrarectally, we detected viremia, virus RNA, or both, in 100% of both species, followed by seroconversion. The magnitude and duration of infectious virus in the blood of macaques suggest humans infected with Zika virus through sexual transmission will likely generate viremias sufficient to infect competent mosquito vectors. Our results indicate that transmission of Zika virus by sexual intercourse might serve as a virus maintenance mechanism in the absence of mosquito-to-human transmission and could increase the probability of establishment and spread of Zika virus in regions where this virus is not present.
- Published
- 2017
- Full Text
- View/download PDF
7. Low potential for mechanical transmission of Ebola virus via house flies (Musca domestica)
- Author
-
Andrew D. Haddow, Farooq Nasar, Christopher W. Schellhase, Roger D. Moon, Susana L. Padilla, Xiankun Zeng, Suzanne E. Wollen-Roberts, Joshua D. Shamblin, Elizabeth C. Grimes, Justine M. Zelko, Kenneth J. Linthicum, Sina Bavari, M. Louise Pitt, and John C. Trefry
- Subjects
Ebola virus ,Filovirus ,Musca domestica ,Fly ,Muscid ,Mechanical ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Ebola virus (EBOV) infection results in high morbidity and mortality and is primarily transmitted in communities by contact with infectious bodily fluids. While clinical and experimental evidence indicates that EBOV is transmitted via mucosal exposure, the ability of non-biting muscid flies to mechanically transmit EBOV following exposure to the face had not been assessed. Results To investigate this transmission route, house flies (Musca domestica Linnaeus) were used to deliver an EBOV/blood mixture to the ocular/nasal/oral facial mucosa of four cynomolgus macaques (Macaca fascicularis Raffles). Following exposure, macaques were monitored for evidence of infection through the conclusion of the study, days 57 and 58. We found no evidence of systemic infection in any of the exposed macaques. Conclusions The results of this study indicate that there is a low potential for the mechanical transmission of EBOV via house flies - the conditions in this study were not sufficient to initiate infection.
- Published
- 2017
- Full Text
- View/download PDF
8. Transcriptomic Analysis Reveals Host miRNAs Correlated with Immune Gene Dysregulation during Fatal Disease Progression in the Ebola Virus Cynomolgus Macaque Disease Model
- Author
-
Christopher P. Stefan, Catherine E. Arnold, Charles J. Shoemaker, Elizabeth E. Zumbrun, Louis A. Altamura, Christina E. Douglas, Cheryl L. Taylor-Howell, Amanda S. Graham, Korey L. Delp, Candace D. Blancett, Keersten M. Ricks, Scott P. Olschner, Joshua D. Shamblin, Suzanne E. Wollen, Justine M. Zelko, Holly A. Bloomfield, Thomas R. Sprague, Heather L. Esham, and Timothy D. Minogue
- Subjects
Ebola virus ,non-human primate ,pathogenesis ,transcriptome ,immunology ,miRNA ,Biology (General) ,QH301-705.5 - Abstract
Ebola virus is a continuing threat to human populations, causing a virulent hemorrhagic fever disease characterized by dysregulation of both the innate and adaptive host immune responses. Severe cases are distinguished by an early, elevated pro-inflammatory response followed by a pronounced lymphopenia with B and T cells unable to mount an effective anti-viral response. The precise mechanisms underlying the dysregulation of the host immune system are poorly understood. In recent years, focus on host-derived miRNAs showed these molecules to play an important role in the host gene regulation arsenal. Here, we describe an investigation of RNA biomarkers in the fatal Ebola virus disease (EVD) cynomolgus macaque model. We monitored both host mRNA and miRNA responses in whole blood longitudinally over the disease course in these non-human primates (NHPs). Analysis of the interactions between these classes of RNAs revealed several miRNA markers significantly correlated with downregulation of genes; specifically, the analysis revealed those involved in dysregulated immune pathways associated with EVD. In particular, we noted strong interactions between the miRNAs hsa-miR-122-5p and hsa-miR-125b-5p with immunological genes regulating both B and T-cell activation. This promising set of biomarkers will be useful in future studies of severe EVD pathogenesis in both NHPs and humans and may serve as potential prognostic targets.
- Published
- 2021
- Full Text
- View/download PDF
9. Ebola Virus Infections in Nonhuman Primates Are Temporally Influenced by Glycoprotein Poly-U Editing Site Populations in the Exposure Material
- Author
-
John C. Trefry, Suzanne E. Wollen, Farooq Nasar, Joshua D. Shamblin, Steven J. Kern, Jeremy J. Bearss, Michelle A. Jefferson, Taylor B. Chance, Jeffery R. Kugelman, Jason T. Ladner, Anna N. Honko, Dean J. Kobs, Morgan Q.S. Wending, Carol L. Sabourin, William D. Pratt, Gustavo F. Palacios, and M. Louise M. Pitt
- Subjects
Ebola virus ,Kikwit ,filovirus ,nonhuman primate ,glycoprotein ,RNA editing ,pathogenesis ,animal model ,vaccine ,therapeutic ,Microbiology ,QR1-502 - Abstract
Recent experimentation with the variants of the Ebola virus that differ in the glycoprotein’s poly-uridine site, which dictates the form of glycoprotein produced through a transcriptional stutter, has resulted in questions regarding the pathogenicity and lethality of the stocks used to develop products currently undergoing human clinical trials to combat the disease. In order to address these concerns and prevent the delay of these critical research programs, we designed an experiment that permitted us to intramuscularly challenge statistically significant numbers of naïve and vaccinated cynomolgus macaques with either a 7U or 8U variant of the Ebola virus, Kikwit isolate. In naïve animals, no difference in survivorship was observed; however, there was a significant delay in the disease course between the two groups. Significant differences were also observed in time-of-fever, serum chemistry, and hematology. In vaccinated animals, there was no statistical difference in survivorship between either challenge groups, with two succumbing in the 7U group compared to 1 in the 8U challenge group. In summary, survivorship was not affected, but the Ebola virus disease course in nonhuman primates is temporally influenced by glycoprotein poly-U editing site populations.
- Published
- 2015
- Full Text
- View/download PDF
10. The utilization of advance telemetry to investigate critical physiological parameters including electroencephalography in cynomolgus macaques following aerosol challenge with eastern equine encephalitis virus
- Author
-
Crystal W. Burke, Simon Authier, Farooq Nasar, Adrienne E. Kimmel, Joshua D. Shamblin, Pamela J. Glass, Michael V. Accardi, Darci R. Smith, Brandi L. Dorsey, Suzanne E. Wollen-Roberts, Sina Bavari, William D. Pratt, Lynn J. Miller, Margaret L. Pitt, John C. Trefry, Thomas R. Sprague, Franco Rossi, and Anthony P. Cardile
- Subjects
0301 basic medicine ,RNA viruses ,Male ,Eastern Equine Encephalitis Virus ,Eastern equine encephalitis virus ,Physiology ,RC955-962 ,Blood Pressure ,Monkeys ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Macaque ,Vascular Medicine ,Body Temperature ,0302 clinical medicine ,Heart Rate ,Arctic medicine. Tropical medicine ,Sinoatrial arrest ,Chlorocebus aethiops ,Medicine and Health Sciences ,Telemetry ,Materials ,Mammals ,Clinical Neurophysiology ,Brain Mapping ,biology ,Eukaryota ,Electroencephalography ,Electrophysiology ,Infectious Diseases ,Bioassays and Physiological Analysis ,Brain Electrophysiology ,Physiological Parameters ,Medical Microbiology ,Viral Pathogens ,Vertebrates ,Physical Sciences ,Viruses ,Respiratory Physiological Phenomena ,Female ,Public aspects of medicine ,RA1-1270 ,Pathogens ,Research Article ,Primates ,Imaging Techniques ,Alphaviruses ,Materials Science ,Cardiology ,Neurophysiology ,Neuroimaging ,Motor Activity ,Research and Analysis Methods ,QT interval ,Microbiology ,Togaviruses ,03 medical and health sciences ,QRS complex ,biology.animal ,Heart rate ,Old World monkeys ,medicine ,Animals ,Humans ,Circadian rhythm ,Microbial Pathogens ,Vero Cells ,Monitoring, Physiologic ,Aerosols ,Biology and life sciences ,business.industry ,Euthanasia ,Alphavirus Infections ,Electrophysiological Techniques ,Public Health, Environmental and Occupational Health ,Organisms ,medicine.disease ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Blood pressure ,Mixtures ,Amniotes ,Encephalitis Virus, Eastern Equine ,Clinical Medicine ,business ,Zoology ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Most alphaviruses are mosquito-borne and can cause severe disease in humans and domesticated animals. In North America, eastern equine encephalitis virus (EEEV) is an important human pathogen with case fatality rates of 30–90%. Currently, there are no therapeutics or vaccines to treat and/or prevent human infection. One critical impediment in countermeasure development is the lack of insight into clinically relevant parameters in a susceptible animal model. This study examined the disease course of EEEV in a cynomolgus macaque model utilizing advanced telemetry technology to continuously and simultaneously measure temperature, respiration, activity, heart rate, blood pressure, electrocardiogram (ECG), and electroencephalography (EEG) following an aerosol challenge at 7.0 log10 PFU. Following challenge, all parameters were rapidly and substantially altered with peak alterations from baseline ranged as follows: temperature (+3.0–4.2°C), respiration rate (+56–128%), activity (-15-76% daytime and +5–22% nighttime), heart rate (+67–190%), systolic (+44–67%) and diastolic blood pressure (+45–80%). Cardiac abnormalities comprised of alterations in QRS and PR duration, QTc Bazett, T wave morphology, amplitude of the QRS complex, and sinoatrial arrest. An unexpected finding of the study was the first documented evidence of a critical cardiac event as an immediate cause of euthanasia in one NHP. All brain waves were rapidly (~12–24 hpi) and profoundly altered with increases of up to 6,800% and severe diffuse slowing of all waves with decreases of ~99%. Lastly, all NHPs exhibited disruption of the circadian rhythm, sleep, and food/fluid intake. Accordingly, all NHPs met the euthanasia criteria by ~106–140 hpi. This is the first of its kind study utilizing state of the art telemetry to investigate multiple clinical parameters relevant to human EEEV infection in a susceptible cynomolgus macaque model. The study provides critical insights into EEEV pathogenesis and the parameters identified will improve animal model development to facilitate rapid evaluation of vaccines and therapeutics., Author summary In North America, EEEV causes the most severe mosquito-borne disease in humans highlighted by fatal encephalitis and permeant debilitating neurological sequelae in survivors. The first confirmed human cases were reported more than 80 years ago and since then multiple sporadic outbreaks have occurred including one of the largest in 2019. Unfortunately, most human infections are diagnosed at the on-set of severe neurological symptoms and consequently a detailed disease course in humans is lacking. This gap in knowledge is a significant obstacle in the development of appropriate animal models to evaluate countermeasures. Here, we performed a cutting-edge study by utilizing a new telemetry technology to understand the course of EEEV infection in a susceptible macaque model by measuring multiple physiological parameters relevant to human disease. Our study demonstrates that the infection rapidly produces considerable alterations in many critical parameters including the electrical activity of the heart and the brain leading to severe disease. The study also highlights the extraordinary potential of new telemetry technology to develop the next generation of animal models to comprehensively investigate pathogenesis as well as evaluate countermeasures to treat and/or prevent EEEV disease.
- Published
- 2021
11. Delayed Disease in Cynomolgus Macaques Exposed to Ebola Virus by an Intranasal Route
- Author
-
Arthur J. Goff, Joshua D. Shamblin, Adrienne E. Kimmel, Catherine L. Wilhelmsen, Justine M. Zelko, Suzanne E. Wollen, and Sara C. Johnston
- Subjects
Male ,viruses ,Immunology ,mucosal ,NHP ,Disease ,medicine.disease_cause ,Macaque ,High morbidity ,biology.animal ,medicine ,Immunology and Allergy ,Animals ,Administration, Intranasal ,Original Research ,Ebola virus ,biology ,business.industry ,Mortality rate ,intranasal ,animal model ,macaque ,Outbreak ,virus diseases ,RC581-607 ,Hemorrhagic Fever, Ebola ,Clinical disease ,Disease Models, Animal ,Macaca fascicularis ,Amylases ,Ebola ,Disease Progression ,RNA, Viral ,Nasal administration ,Female ,Immunologic diseases. Allergy ,business ,EBOV - Abstract
Ebola virus remains a significant public health concern due to high morbidity and mortality rates during recurrent outbreaks in endemic areas. Therefore, the development of countermeasures against Ebola virus remains a high priority, and requires the availability of appropriate animal models for efficacy evaluations. The most commonly used nonhuman primate models for efficacy evaluations against Ebola virus utilize the intramuscular or aerosol route of exposure. Although clinical disease signs are similar to human cases, disease progression in these models is much more rapid, and this can pose significant hurdles for countermeasure evaluations. The objective of the present study was to evaluate the Ebola virus disease course that arises after cynomolgus macaques are exposed to Ebola virus by a mucosal route (the intranasal route). Two different doses (10 pfu and 100 pfu) and delivery methodologies (drop-wise and mucosal atomization device) were evaluated on this study. Differences in clinical disease between dose and delivery groups were not noted. However, a delayed disease course was identified for approximately half of the animals on study, and this delayed disease was dose and administration method independent. Therefore, it appears that mucosal exposure with Ebola virus results in a disease course in cynomolgus macaques that more accurately replicates that which is documented for human cases. In summary, the data presented support the need for further development of this model as a possible alternative to parenteral and small-particle aerosol models for the study of human Ebola virus disease and for countermeasure evaluations.
- Published
- 2021
12. Transcriptomic Analysis Reveals Host miRNAs Correlated with Immune Gene Dysregulation during Fatal Disease Progression in the Ebola Virus Cynomolgus Macaque Disease Model
- Author
-
Scott P Olschner, Christopher P. Stefan, Christina E. Douglas, Candace D. Blancett, Keersten M. Ricks, Catherine E. Arnold, Cheryl L. Taylor-Howell, Timothy D. Minogue, Korey L Delp, Elizabeth E. Zumbrun, Holly A. Bloomfield, Thomas R. Sprague, Suzanne E. Wollen, Justine M. Zelko, Joshua D. Shamblin, Louis A. Altamura, Amanda S. Graham, Charles J. Shoemaker, and Heather L. Esham
- Subjects
0301 basic medicine ,Microbiology (medical) ,mRNA ,Virulence ,Inflammation ,non-human primate ,Disease ,Biology ,medicine.disease_cause ,Microbiology ,Article ,Transcriptome ,Pathogenesis ,immunology ,03 medical and health sciences ,Ebola virus ,0302 clinical medicine ,Immune system ,Virology ,microRNA ,medicine ,cytokine ,lcsh:QH301-705.5 ,miRNA ,pathogenesis ,apoptosis ,030104 developmental biology ,lcsh:Biology (General) ,inflammation ,030220 oncology & carcinogenesis ,Immunology ,medicine.symptom ,transcriptome - Abstract
Ebola virus is a continuing threat to human populations, causing a virulent hemorrhagic fever disease characterized by dysregulation of both the innate and adaptive host immune responses. Severe cases are distinguished by an early, elevated pro-inflammatory response followed by a pronounced lymphopenia with B and T cells unable to mount an effective anti-viral response. The precise mechanisms underlying the dysregulation of the host immune system are poorly understood. In recent years, focus on host-derived miRNAs showed these molecules to play an important role in the host gene regulation arsenal. Here, we describe an investigation of RNA biomarkers in the fatal Ebola virus disease (EVD) cynomolgus macaque model. We monitored both host mRNA and miRNA responses in whole blood longitudinally over the disease course in these non-human primates (NHPs). Analysis of the interactions between these classes of RNAs revealed several miRNA markers significantly correlated with downregulation of genes, specifically, the analysis revealed those involved in dysregulated immune pathways associated with EVD. In particular, we noted strong interactions between the miRNAs hsa-miR-122-5p and hsa-miR-125b-5p with immunological genes regulating both B and T-cell activation. This promising set of biomarkers will be useful in future studies of severe EVD pathogenesis in both NHPs and humans and may serve as potential prognostic targets.
- Published
- 2021
- Full Text
- View/download PDF
13. Development of a coronavirus disease 2019 nonhuman primate model using airborne exposure
- Author
-
Philip A Bowling, Joshua L. Moore, Jimmy O. Fiallos, Brett Beitzel, Leslie Klosterman, Keersten M. Ricks, Christopher Alan Jensen, Alexandra Jay, Jay W. Hooper, Kristen Akers, Denise K. Danner, Jo Lynne Raymond, Terrence Garcia, Susan R. Coyne, Jeffrey R. Kugelman, Aysegul Nalca, Ondraya Frick, Stephen Stevens, David Nyakiti, Joshua A. Johnson, Paul A. Kuehnert, Matthew Durant, Nazira Alli, Willie Sifford, Jeffrey M. Smith, Korey L. Delp, Timothy D. Minogue, Xiankun Zeng, Joshua D. Shamblin, Ruth Kim, Margaret L. Pitt, Wendy Giles, Lauren E. White, Stephanie A. Bellanca, Jeanean Ghering, Fabian Paz, Franco Rossi, Jeffrey W. Koehler, Brian J. Kearney, Kerry Berrier, Tamara L. Clements, Charles J. Shoemaker, Kathleen Gibson, Heather L. Esham, Jennifer Scruggs, David N Dyer, James Barth, and Sara C. Johnston
- Subjects
0301 basic medicine ,Male ,RNA viruses ,Viral Diseases ,Pulmonology ,Coronaviruses ,Physiology ,Monkeys ,Chlorocebus aethiops ,Macaque ,0302 clinical medicine ,Medical Conditions ,Primate ,030212 general & internal medicine ,Enzyme-Linked Immunoassays ,Lung ,Pathology and laboratory medicine ,Virus Testing ,Mammals ,Multidisciplinary ,biology ,Transmission (medicine) ,Eukaryota ,Medical microbiology ,Virus Shedding ,Infectious Diseases ,Viruses ,Vertebrates ,Medicine ,Female ,SARS CoV 2 ,Pathogens ,Research Article ,Primates ,SARS coronavirus ,Science ,Research and Analysis Methods ,Airborne transmission ,Microbiology ,03 medical and health sciences ,Respiratory Disorders ,Immune system ,Diagnostic Medicine ,biology.animal ,Old World monkeys ,Disease Transmission, Infectious ,Animals ,Respiratory Physiology ,Viral shedding ,Immunoassays ,Medicine and health sciences ,Biology and life sciences ,SARS-CoV-2 ,Organisms ,Viral pathogens ,COVID-19 ,Covid 19 ,biology.organism_classification ,Virology ,Macaca mulatta ,Microbial pathogens ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Amniotes ,Respiratory Infections ,Immunologic Techniques ,African Green Monkey ,Zoology - Abstract
Airborne transmission is predicted to be a prevalent route of human exposure with SARS-CoV-2. Aside from African green monkeys, nonhuman primate models that replicate airborne transmission of SARS-CoV-2 have not been investigated. A comparative evaluation of COVID-19 in African green monkeys, rhesus macaques, and cynomolgus macaques following airborne exposure to SARS-CoV-2 was performed to determine critical disease parameters associated with disease progression, and establish correlations between primate and human COVID-19. Respiratory abnormalities and viral shedding were noted for all animals, indicating successful infection. Cynomolgus macaques developed fever, and thrombocytopenia was measured for African green monkeys and rhesus macaques. Type II pneumocyte hyperplasia and alveolar fibrosis were more frequently observed in lung tissue from cynomolgus macaques and African green monkeys. The data indicate that, in addition to African green monkeys, macaques can be successfully infected by airborne SARS-CoV-2, providing viable macaque natural transmission models for medical countermeasure evaluation.
- Published
- 2021
14. The Utilization of Advance Telemetry to Investigate Important Physiological Parameters Including Electroencephalography in Cynomolgus Macaques Following Aerosol Challenge with Eastern Equine Encephalitis Virus
- Author
-
William D. Pratt, Anthony P. Cardile, John C. Trefry, Franco Rossi, Adrienne E. Kimmel, Michael V. Accardi, Lynn Miller, Suzanne E. Wollen-Roberts, Simon Authier, Farooq Nasar, Joshua D. Shamblin, Brandi L. Dorsey, Sina Bavari, Pamela J. Glass, Margaret L. Pitt, Thomas R. Sprague, and Darci R. Smith
- Subjects
business.industry ,Eastern equine encephalitis virus ,Physiology ,medicine.disease ,medicine.disease_cause ,QT interval ,QRS complex ,Blood pressure ,Sinoatrial arrest ,Case fatality rate ,Heart rate ,medicine ,Circadian rhythm ,business - Abstract
Most alphaviruses are mosquito-borne and can cause severe disease in humans and domesticated animals. In North America, eastern equine encephalitis virus (EEEV) is an important human pathogen with case fatality rates of 30-90%. Currently, there are no therapeutics or vaccines to treat and/or prevent human infection. One critical impediment in countermeasure development is the lack of insight into clinically relevant parameters in a susceptible animal model. This study examined the disease course of EEEV in a cynomolgus macaque model utilizing advanced telemetry technology to continuously and simultaneously measure temperature, respiration, activity, heart rate, blood pressure, electrocardiogram (ECG), and electroencephalography (EEG) following an aerosol challenge at 7.0 log10 PFU. Following challenge, all parameters were rapidly and substantially altered with peak alterations from baseline ranged as follows: temperature (+3.0-4.2 °C), respiration rate (+56-128%), activity (+15-76% daytime and +5-22% nighttime), heart rate (+67-190%), systolic (+44-67%) and diastolic blood pressure (+45-80%). Cardiac abnormalities comprised of alterations in QRS and PR duration, QTc Bazett, T wave morphology, amplitude of the QRS complex, and sinoatrial arrest. An unexpected finding of the study was the first documented evidence of a critical cardiac event as an immediate cause of euthanasia in one NHP. All brain waves were rapidly (12-24 hpi) and profoundly altered with increases of up to 6,800% and severe diffuse slowing of all waves with decreases of ∼99%. Lastly, all NHPs exhibited disruption of the circadian rhythm, sleep, and food/fluid intake. Accordingly, all NHPs met the euthanasia criteria by ∼106-140 hpi. This is the first of its kind study utilizing state of the art telemetry to investigate multiple clinical parameters relevant to human EEEV infection in a susceptible cynomolgus macaque model. The study provides critical insights into EEEV pathogenesis and the parameters identified will improve animal model development to facilitate rapid evaluation of vaccines and therapeutics.Author SummaryIn North America, EEEV causes the most severe mosquito-borne disease in humans highlighted by fatal encephalitis and permeant debilitating neurological sequelae in survivors. The first confirmed human cases were reported more than 80 years ago and since then multiple sporadic outbreaks have occurred including one of the largest in 2019. Unfortunately, most human infections are diagnosed at the on-set of severe neurological symptoms and consequently a detailed disease course in humans is lacking. This gap in knowledge is a significant obstacle in the development of appropriate animal models to evaluate countermeasures. Here, we performed a cutting-edge study by utilizing a new telemetry technology to understand the course of EEEV infection in a susceptible macaque model by measuring multiple physiological parameters relevant to human disease. Our study demonstrates that the infection rapidly produces considerable alterations in many critical parameters including the electrical activity of the heart and the brain leading to severe disease. The study also highlights the extraordinary potential of new telemetry technology to develop the next generation of animal models in order to comprehensively investigate pathogenesis as well as evaluate countermeasures to treat and/or prevent EEEV disease.
- Published
- 2020
- Full Text
- View/download PDF
15. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease
- Author
-
Joshua D. Shamblin, Eric M. Mucker, Brian D. Carey, Aura R. Garrison, Hypaitia B. Rauch, Jay W. Hooper, Xiankun Zeng, Jeffrey M. Smith, Bradley S. Hollidge, April M. Babka, Collin Fitzpatrick, Rebecca L. Brocato, Curtis R. Cline, Catherine V. Badger, Jun Liu, Lauren E. White, and Joseph W. Golden
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,Pneumonia, Viral ,Mice, Transgenic ,Disease ,Peptidyl-Dipeptidase A ,Lung injury ,Mouse models ,Severe Acute Respiratory Syndrome ,Virus Replication ,Severity of Illness Index ,Keratin 18 ,Proinflammatory cytokine ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cause of Death ,medicine ,Animals ,Humans ,Lung ,Pandemics ,Molecular pathology ,Infectious disease ,business.industry ,Respiratory disease ,COVID-19 ,General Medicine ,respiratory system ,medicine.disease ,respiratory tract diseases ,Survival Rate ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Angiotensin-converting enzyme 2 ,Disease Progression ,Medicine ,Female ,Angiotensin-Converting Enzyme 2 ,Coronavirus Infections ,business ,Research Article - Abstract
The emergence of SARS-CoV-2 has created an international health crisis, and small animal models mirroring SARS-CoV-2 human disease are essential for medical countermeasure (MCM) development. Mice are refractory to SARS-CoV-2 infection owing to low-affinity binding to the murine angiotensin-converting enzyme 2 (ACE2) protein. Here, we evaluated the pathogenesis of SARS-CoV-2 in male and female mice expressing the human ACE2 gene under the control of the keratin 18 promoter (K18). In contrast to nontransgenic mice, intranasal exposure of K18-hACE2 animals to 2 different doses of SARS-CoV-2 resulted in acute disease, including weight loss, lung injury, brain infection, and lethality. Vasculitis was the most prominent finding in the lungs of infected mice. Transcriptomic analysis from lungs of infected animals showed increases in transcripts involved in lung injury and inflammatory cytokines. In the low-dose challenge groups, there was a survival advantage in the female mice, with 60% surviving infection, whereas all male mice succumbed to disease. Male mice that succumbed to disease had higher levels of inflammatory transcripts compared with female mice. To our knowledge, this is the first highly lethal murine infection model for SARS-CoV-2 and should be valuable for the study of SARS-CoV-2 pathogenesis and for the assessment of MCMs., A highly lethal murine infection model for SARS-CoV-2 using mice transgenic for the human ACE2 protein is described.
- Published
- 2020
16. Stability of SARS-CoV-2 on Produce following a Low-Dose Aerosol Exposure
- Author
-
David E. Harbourt, Taylor R. Watt, Andrew D. Haddow, David N Dyer, Holly A. Bloomfield, and Joshua D. Shamblin
- Subjects
Veterinary medicine ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,030231 tropical medicine ,Food Contamination ,Airborne transmission ,complex mixtures ,03 medical and health sciences ,Betacoronavirus ,0302 clinical medicine ,Jalapeno pepper ,Virology ,Vegetables ,Infectious virus ,Aerosols ,SARS-CoV-2 ,Low dose ,fungi ,Temperature ,food and beverages ,Articles ,Aerosol ,Infectious Diseases ,Fomites ,Fruit ,Environmental science ,Parasitology - Abstract
We modeled the stability of SARS-CoV-2 on apples, tomatoes, and jalapeño peppers at two temperatures following a low-dose aerosol exposure designed to simulate an airborne transmission event involving droplet nuclei. Infectious virus was not recovered postexposure.
- Published
- 2020
17. Development of a Coronavirus Disease 2019 Nonhuman Primate Model Using Airborne Exposure
- Author
-
Leslie Klosterman, Brian J. Kearney, Denise K. Danner, Kerry Berrier, Jimmy O. Fiallos, James Barth, Lauren E. White, Nazira Alli, Joshua A. Johnson, Franco Rossi, Xiankun Zeng, Joshua L. Moore, Jennifer Scruggs, Alexandra Jay, Margaret L. Pitt, Wendy Giles, Terrence Garcia, David N Dyer, Heather L. Esham, Jeanean Ghering, Fabian Paz, Joshua D. Shamblin, Stephen Stevens, Ondraya Frick, Jo Lynne Raymond, Matthew Durant, Aysegul Nalca, Sara C. Johnston, Philip A Bowling, Stephanie A. Bellanca, Ruth Kim, David Nyakiti, Christopher Alan Jensen, Kristen Akers, and Willie Sifford
- Subjects
Coronavirus disease 2019 (COVID-19) ,biology ,Transmission (medicine) ,biology.animal ,African Green Monkey ,Viral shedding ,Virology ,Macaque ,Airborne transmission ,Virus ,Nonhuman primate - Abstract
Airborne transmission is predicted to be a prevalent route of human exposure with SARS-CoV-2. Aside from African green monkeys, nonhuman primate models that replicate airborne transmission of SARS-CoV-2 have not been investigated. A comprehensive and comparative evaluation of COVID-19 in African green monkeys, rhesus macaques, and cynomolgus macaques following airborne exposure to SARS-CoV-2 was performed to define parameters critical to disease progression and the extent to which they correlate with human COVID-19. Respiratory abnormalities and viral shedding were noted for all animals, indicating successful infection. Cynomolgus macaques developed fever, and thrombocytopenia was measured for African green monkeys and rhesus macaques. Type II pneumocyte hyperplasia and alveolar fibrosis were more frequently observed in lung tissue from cynomolgus macaques and African green monkeys. The data indicate that, in addition to African green monkeys, macaques can be successfully infected by airborne SARS-CoV-2, providing viable macaque natural transmission models for medical countermeasure evaluation.One Sentence SummaryNonhuman primates develop COVID-19 following airborne virus exposure.
- Published
- 2020
- Full Text
- View/download PDF
18. Anti-HFRS Human IgG Produced in Transchromosomic Bovines Has Potent Hantavirus Neutralizing Activity and Is Protective in Animal Models
- Author
-
Melanie V. Cohen, Joshua D. Shamblin, Jay W. Hooper, Eddie Sullivan, Rebecca L. Brocato, Padmanabh Chivukula, Lucia M. Principe, Brandon Somerville, Christoph L Bausch, Steven A. Kwilas, Jerel Vega, Priya Karmali, Hua Wu, and Casey C. Perley
- Subjects
Microbiology (medical) ,medicine.drug_class ,lcsh:QR1-502 ,Monoclonal antibody ,Microbiology ,hantavirus ,lcsh:Microbiology ,Neutralization ,Virus ,DNA vaccination ,Hyperimmunization ,03 medical and health sciences ,medicine ,Neutralizing antibody ,Original Research ,030304 developmental biology ,transchromosomic bovine ,marmoset ,0303 health sciences ,passive transfer ,hantaan ,biology ,030306 microbiology ,puumala ,Virology ,hamster ,Polyclonal antibodies ,biology.protein ,HFRS ,Antibody - Abstract
We explored an emerging technology to produce anti-Hantaan virus (HTNV) and anti-Puumala virus (PUUV) neutralizing antibodies for use as pre- or post-exposure prophylactics. The technology involves hyperimmunization of transchomosomic bovines (TcB) engineered to express human polyclonal IgG antibodies with HTNV and PUUV DNA vaccines encoding GnGc glycoproteins. For the anti-HTNV product, TcB was hyperimmunized with HTNV DNA plus adjuvant or HTNV DNA formulated using lipid nanoparticles (LNP). The LNP-formulated vaccine yielded fivefold higher neutralizing antibody titers using 10-fold less DNA. Human IgG purified from the LNP-formulated animal (SAB-159), had anti-HTNV neutralizing antibody titers >100,000. SAB-159 was capable of neutralizing pseudovirions with monoclonal antibody escape mutations in Gn and Gc demonstrating neutralization escape resistance. SAB-159 protected hamsters from HTNV infection when administered pre- or post-exposure, and limited HTNV infection in a marmoset model. An LNP-formulated PUUV DNA vaccine generated purified anti-PUUV IgG, SAB-159P, with a neutralizing antibody titer >600,000. As little as 0.33 mg/kg of SAB-159P protected hamsters against PUUV infection for pre-exposure and 10 mg/kg SAB-159P protected PUUV-infected hamsters post-exposure. These data demonstrate that DNA vaccines combined with the TcB-based manufacturing platform can be used to rapidly produce potent, human, polyclonal, escape-resistant anti-HTNV, and anti-PUUV neutralizing antibodies that are protective in animal models.
- Published
- 2020
- Full Text
- View/download PDF
19. Characterization of Ebola Virus Disease (EVD) in Rhesus Monkeys for Development of EVD Therapeutics
- Author
-
Christiana Blair, Roy Bannister, Sina Bavari, Jacqueline Tarrant, Matthew C. Reed, Ginger Donnelly, Joshua D. Shamblin, Adrienne E. Kimmel, Travis K. Warren, Laura Gomba, Kelly S. Wetzel, Sean A. Van Tongeren, Jesse T. Steffens, Jessica M. Weidner, Tomas Cihlar, Nicole Lackemeyer, Franco Rossi, Jay Wells, Elizabeth E. Zumbrun, Bali Singh, Danielle P. Porter, Holly A. Bloomfield, Eric Lee, Carly Garvey, and Jo Lynne Raymond
- Subjects
Male ,0301 basic medicine ,rhesus ,lcsh:QR1-502 ,Viremia ,Disease ,Systemic inflammation ,medicine.disease_cause ,Injections, Intramuscular ,Macaque ,Article ,lcsh:Microbiology ,evd ,03 medical and health sciences ,0302 clinical medicine ,Virology ,biology.animal ,Coagulopathy ,Animals ,Medicine ,030212 general & internal medicine ,ebola virus ,disease ,Ebola virus ,biology ,business.industry ,animal model ,macaque ,Outbreak ,Hemorrhagic Fever, Ebola ,Ebolavirus ,medicine.disease ,Macaca mulatta ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Disease Progression ,Female ,medicine.symptom ,monkey ,business ,Natural history study - Abstract
Recent Ebola virus (EBOV) outbreaks in West Africa and the Democratic Republic of the Congo have highlighted the urgent need for approval of medical countermeasures for treatment and prevention of EBOV disease (EVD). Until recently, when successes were achieved in characterizing the efficacy of multiple experimental EVD therapeutics in humans, the only feasible way to obtain data regarding potential clinical benefits of candidate therapeutics was by conducting well-controlled animal studies. Nonclinical studies are likely to continue to be important tools for screening and development of new candidates with improved pharmacological properties. Here, we describe a natural history study to characterize the time course and order of progression of the disease manifestations of EVD in rhesus monkeys. In 12 rhesus monkeys exposed by the intramuscular route to 1000 plaque-forming units of EBOV, multiple endpoints were monitored for 28 days following exposure. The disease progressed rapidly with mortality events occurring 7&ndash, 10 days after exposure. Key disease manifestations observed consistently across the infected animals included, but were not limited to, viremia, fever, systemic inflammation, coagulopathy, lymphocytolysis, renal tubular necrosis with mineralization, and hepatocellular degeneration and necrosis.
- Published
- 2020
20. Detailed analysis of the pathologic hallmarks of Nipah virus (Malaysia) disease in the African green monkey infected by the intratracheal route
- Author
-
Curtis Cline, Todd M. Bell, Paul Facemire, Xiankun Zeng, Thomas Briese, W. Ian Lipkin, Joshua D. Shamblin, Heather L. Esham, Ginger C. Donnelly, Joshua C. Johnson, Lisa E. Hensley, Anna N. Honko, and Sara C. Johnston
- Subjects
Henipavirus Infections ,Lung Diseases ,Male ,Disease Models, Animal ,Multidisciplinary ,viruses ,Chlorocebus aethiops ,Malaysia ,Nipah Virus ,Animals ,Encephalitis, Viral - Abstract
Disease associated with Nipah virus infection causes a devastating and often fatal spectrum of syndromes predominated by both respiratory and neurologic conditions. Additionally, neurologic sequelae may manifest months to years later after virus exposure or apparent recovery. In the two decades since this disease emerged, much work has been completed in an attempt to understand the pathogenesis and facilitate development of medical countermeasures. Here we provide detailed organ system-specific pathologic findings following exposure of four African green monkeys to 2.41×105 pfu of the Malaysian strain of Nipah virus. Our results further substantiate the African green monkey as a model of human Nipah virus disease, by demonstrating both the respiratory and neurologic components of disease. Additionally, we demonstrate that a chronic phase of disease exists in this model, that may provide an important opportunity to study the enigmatic late onset and relapse encephalitis as it is described in human disease.
- Published
- 2022
- Full Text
- View/download PDF
21. Development of Clinical-Stage Human Monoclonal Antibodies That Treat Advanced Ebola Virus Disease in Nonhuman Primates
- Author
-
Andrew B. Ward, Christos A. Kyratsous, Ricardo Carrion, Gabriella Worwa, William D. Pratt, Gang Chen, William C. Olson, Terra Potocky, Joel H. Martin, Karl J. Erlandson, John C. Trefry, Ashique Rafique, Peter W. Mason, Hilary M. Staples, Tammy T. Huang, Hannah L. Turner, Robert Babb, Drew Dudgeon, Leah Lipsich, Joshua D. Shamblin, Ying Yan, Yasuteru Sakurai, Thomas M Dreier, Manu Anantpadma, Kevin Yu, Marcela Torres, Sandra L. Bixler, Suzanne E. Wollen, Charles D. Murin, Neil Stahl, Justine M. Zelko, Melissa S Willis, Robert A. Davey, Darya Burakov, Jeanette L. Fairhurst, Ashok Badithe, Margaret L. Pitt, Franco Rossi, Taylor B. Chance, Kimberly Armstrong, and Kristen E. Pascal
- Subjects
0301 basic medicine ,Male ,filovirus ,medicine.drug_class ,Guinea Pigs ,Supplement Articles ,Disease ,medicine.disease_cause ,Monoclonal antibody ,Antibodies, Viral ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Emerging infections ,Medicine ,Animals ,Humans ,Immunology and Allergy ,Glycoproteins ,Ebola virus ,biology ,business.industry ,Antibodies, Monoclonal ,Binding (Molecular Function) ,Hemorrhagic Fever, Ebola ,Virology ,Antibodies, Neutralizing ,Macaca mulatta ,Treatment ,030104 developmental biology ,HEK293 Cells ,Infectious Diseases ,biology.protein ,monoclonal antibodies ,Antibody ,business ,030217 neurology & neurosurgery ,EBOV - Abstract
Background For most classes of drugs, rapid development of therapeutics to treat emerging infections is challenged by the timelines needed to identify compounds with the desired efficacy, safety, and pharmacokinetic profiles. Fully human monoclonal antibodies (mAbs) provide an attractive method to overcome many of these hurdles to rapidly produce therapeutics for emerging diseases. Methods In this study, we deployed a platform to generate, test, and develop fully human antibodies to Zaire ebolavirus. We obtained specific anti-Ebola virus (EBOV) antibodies by immunizing VelocImmune mice that use human immunoglobulin variable regions in their humoral responses. Results Of the antibody clones isolated, 3 were selected as best at neutralizing EBOV and triggering FcγRIIIa. Binding studies and negative-stain electron microscopy revealed that the 3 selected antibodies bind to non-overlapping epitopes, including a potentially new protective epitope not targeted by other antibody-based treatments. When combined, a single dose of a cocktail of the 3 antibodies protected nonhuman primates (NHPs) from EBOV disease even after disease symptoms were apparent. Conclusions This antibody cocktail provides complementary mechanisms of actions, incorporates novel specificities, and demonstrates high-level postexposure protection from lethal EBOV disease in NHPs. It is now undergoing testing in normal healthy volunteers in preparation for potential use in future Ebola epidemics.
- Published
- 2018
- Full Text
- View/download PDF
22. Zika Virus Infection in Syrian Golden Hamsters and Strain 13 Guinea Pigs
- Author
-
Lynn J. Miller, Andrew D. Haddow, Maria Theresa Alera, Christopher W. Schellhase, Richard G. Jarman, Sina Bavari, Suzanne E. Wollen-Roberts, Luis A. Lugo-Roman, Joshua D. Shamblin, M. Louise M. Pitt, Adrienne E. Kimmel, Sarah L. Norris, In-Kyu Yoon, Thomas R. Sprague, Stephanie M. Valdez, and Farooq Nasar
- Subjects
0301 basic medicine ,Injections, Subcutaneous ,Guinea Pigs ,Viremia ,Antibodies, Viral ,Host Specificity ,Neutralization ,Virus ,Zika virus ,03 medical and health sciences ,Virology ,medicine ,Animals ,Disease Resistance ,Mesocricetus ,biology ,Strain (chemistry) ,Zika Virus Infection ,Zika Virus ,Articles ,biology.organism_classification ,medicine.disease ,Antibodies, Neutralizing ,Disease Models, Animal ,Titer ,030104 developmental biology ,Infectious Diseases ,Female ,Parasitology ,Injections, Intraperitoneal ,Golden hamster - Abstract
To evaluate potential immunocompetent small animal models of Zika virus (ZIKV) infection, we inoculated Syrian golden hamsters (subcutaneously or intraperitoneally) and strain 13 guinea pigs (intraperitoneally) with Senegalese ZIKV strain ArD 41525 or Philippines ZIKV strain CPC-0740. We did not detect viremia in hamsters inoculated subcutaneously with either virus strain, although some hamsters developed virus neutralizing antibodies. However, we detected statistically significant higher viremias (P = 0.0285) and a higher median neutralization titer (P = 0.0163) in hamsters inoculated intraperitoneally with strain ArD 41525 compared with strain CPC-0740. Furthermore, some hamsters inoculated with strain ArD 41525 displayed mild signs of disease. By contrast, strain 13 guinea pigs inoculated intraperitoneally with either strain did not have detectable viremias and less than half developed virus neutralizing antibodies. Our results support the use of the Syrian golden hamster intraperitoneal model to explore phenotypic variation between ZIKV strains.
- Published
- 2018
- Full Text
- View/download PDF
23. A DNA vaccine delivered by dermal electroporation fully protects cynomolgus macaques against Lassa fever
- Author
-
Suzanne E. Wollen, Paul R Facemire, Kathleen A. Cashman, Niranjan Y. Sardesai, Carl I Shaia, Kate E. Broderick, Connie S. Schmaljohn, Todd M. Bell, Joshua D. Shamblin, Jeremy J. Bearss, and Eric R. Wilkinson
- Subjects
0301 basic medicine ,DNA vaccine ,electroporation ,Male ,viruses ,Arenaviridae ,Immunology ,medicine.disease_cause ,Administration, Cutaneous ,DNA vaccination ,03 medical and health sciences ,Lassa Fever ,medicine ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Viremia ,Lassa fever ,Immunization Schedule ,Pharmacology ,biology ,Electroporation ,virus diseases ,RNA virus ,Viral Vaccines ,medicine.disease ,biology.organism_classification ,Virology ,Research Papers ,Survival Analysis ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Lassa virus ,Treatment Outcome - Abstract
Lassa virus (LASV) is an ambisense RNA virus in the Arenaviridae family and is the etiological agent of Lassa fever, a severe hemorrhagic disease endemic to West and Central Africa.1,2 There are no US Food and Drug Administration (FDA)-licensed vaccines available to prevent Lassa fever.1,2 in our previous studies, we developed a gene-optimized DNA vaccine that encodes the glycoprotein precursor gene of LASV (Josiah strain) and demonstrated that 3 vaccinations accompanied by dermal electroporation protected guinea pigs from LASV-associated illness and death. Here, we describe an initial efficacy experiment in cynomolgus macaque nonhuman primates (NHPs) in which we followed an identical 3-dose vaccine schedule that was successful in guinea pigs, and a follow-on experiment in which we used an accelerated vaccination strategy consisting of 2 administrations, spaced 4 weeks apart. In both studies, all of the LASV DNA-vaccinated NHPs survived challenge and none of them had measureable, sustained viremia or displayed weight loss or other disease signs post-exposure. Three of 10 mock-vaccinates survived exposure to LASV, but all of them became acutely ill post-exposure and remained chronically ill to the study end point (45 d post-exposure). Two of the 3 survivors experienced sensorineural hearing loss (described elsewhere). These results clearly demonstrate that the LASV DNA vaccine combined with dermal electroporation is a highly effective candidate for eventual use in humans.
- Published
- 2017
24. Modeling mosquito-borne and sexual transmission of Zika virus in an enzootic host, the African green monkey
- Author
-
Sina Bavari, Lynn J. Miller, William D. Pratt, Adrienne E. Kimmel, Gustavo Palacios, Stephanie M. Valdez, Aysegul Nalca, Luis A. Lugo-Roman, Farooq Nasar, Michael R. Wiley, M. Louise M. Pitt, Suzanne E. Wollen-Roberts, Joshua D. Shamblin, Andrew D. Haddow, Unai Pérez-Sautu, Lisa H. Cazares, Lucia M. Principe, and Franco Rossi
- Subjects
0301 basic medicine ,Male ,RNA viruses ,Viral Diseases ,Physiology ,viruses ,RC955-962 ,Aminotransferases ,Monkeys ,Pathology and Laboratory Medicine ,Biochemistry ,Zika virus ,0302 clinical medicine ,Arctic medicine. Tropical medicine ,Immune Physiology ,Chlorocebus aethiops ,Medicine and Health Sciences ,Mammals ,Immune System Proteins ,biology ,Transmission (medicine) ,Zika Virus Infection ,Eukaryota ,Sexually Transmitted Diseases, Viral ,Body Fluids ,Enzymes ,Titer ,Infectious Diseases ,Blood ,Medical Microbiology ,Viral Pathogens ,Viruses ,Vertebrates ,Female ,Public aspects of medicine ,RA1-1270 ,Pathogens ,Anatomy ,Viral load ,Macaque ,Research Article ,Primates ,Viral Release ,Sexual transmission ,030231 tropical medicine ,Immunology ,Sexually Transmitted Diseases ,Vector Borne Diseases ,Viremia ,Microbiology ,Virus ,Antibodies ,03 medical and health sciences ,Transferases ,Virology ,Old World monkeys ,medicine ,Disease Transmission, Infectious ,Animals ,Viral shedding ,Microbial Pathogens ,Biology and life sciences ,Flaviviruses ,Public Health, Environmental and Occupational Health ,Organisms ,Proteins ,Zika Virus ,biology.organism_classification ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Culicidae ,Amniotes ,Enzymology ,Viral Transmission and Infection - Abstract
Mosquito-borne and sexual transmission of Zika virus (ZIKV), a TORCH pathogen, recently initiated a series of large epidemics throughout the Tropics. Animal models are necessary to determine transmission risk and study pathogenesis, as well screen antivirals and vaccine candidates. In this study, we modeled mosquito and sexual transmission of ZIKV in the African green monkey (AGM). Following subcutaneous, intravaginal or intrarectal inoculation of AGMs with ZIKV, we determined the transmission potential and infection dynamics of the virus. AGMs inoculated by all three transmission routes exhibited viremia and viral shedding followed by strong virus neutralizing antibody responses, in the absence of clinical illness. All four of the subcutaneously inoculated AGMs became infected (mean peak viremia: 2.9 log10 PFU/mL, mean duration: 4.3 days) and vRNA was detected in their oral swabs, with infectious virus being detected in a subset of these specimens. Although all four of the intravaginally inoculated AGMs developed virus neutralizing antibody responses, only three had detectable viremia (mean peak viremia: 4.0 log10 PFU/mL, mean duration: 3.0 days). These three AGMs also had vRNA and infectious virus detected in both oral and vaginal swabs. Two of the four intrarectally inoculated AGMs became infected (mean peak viremia: 3.8 log10 PFU/mL, mean duration: 3.5 days). vRNA was detected in oral swabs collected from both of these infected AGMs, and infectious virus was detected in an oral swab from one of these AGMs. Notably, vRNA and infectious virus were detected in vaginal swabs collected from the infected female AGM (peak viral load: 7.5 log10 copies/mL, peak titer: 3.8 log10 PFU/mL, range of detection: 5–21 days post infection). Abnormal clinical chemistry and hematology results were detected and acute lymphadenopathy was observed in some AGMs. Infection dynamics in all three AGM ZIKV models are similar to those reported in the majority of human ZIKV infections. Our results indicate that the AGM can be used as a surrogate to model mosquito or sexual ZIKV transmission and infection. Furthermore, our results suggest that AGMs are likely involved in the enzootic maintenance and amplification cycle of ZIKV., Author summary Zika virus (ZIKV) is primarily maintained in an enzootic cycle involving nonhuman primates and mosquitoes, with epizootics and epidemics occurring when the virus is introduced into naïve populations of nonhuman primates or humans, respectively. While, the primary transmission mechanism of the virus is by the bite on an infected mosquito, ZIKV can also be sexually transmitted. In an effort to develop novel animal models to study ZIKV disease, and to better understand the role of nonhuman primates as amplification and maintenance hosts of ZIKV in nature, we modeled mosquito-borne and sexual transmission of ZIKV in the enzootic host, the African green monkey (AGM). Infection dynamics and neutralizing antibody responses in all three AGM ZIKV models (subcutaneous, intravaginal and intrarectal) in the absence of clinical illness–recapitulated reported generalized human disease course. Furthermore, we detected prolonged shedding with high viral loads and infectious virus in the vaginal swabs collected from an infected female AGM inoculated intrarectally. Notably, these results support limited human clinical evidence that ZIKV transmission can occur during female-to-male vaginal sexual acts, and furthermore indicate the existence of ZIKV super-spreaders. Finally, our results indicate sexual transmission of ZIKV could occur among infected nonhuman primates (e.g. Chlorocebus spp.) in Africa and may serve as a secondary transmission and maintenance mechanism in the absence of mosquito-to-nonhuman primate transmission.
- Published
- 2019
25. Persistent Crimean-Congo hemorrhagic fever virus infection in the testes and within granulomas of non-human primates with latent tuberculosis
- Author
-
Joshua D. Shamblin, Kayla M. Coffin, Darci R. Smith, Matt A. Voorhees, Jeffrey W. Koehler, Xiankun Zeng, Jun Liu, Franco Rossi, Connie S. Schmaljohn, Christopher W. Schellhase, Adrienne E. Kimmel, Charles J. Shoemaker, Justine M. Zelko, Keersten M. Ricks, William D. Pratt, Collin Fitzpatrick, Aura R. Garrison, Randal J. Schoepp, Joseph W. Golden, and Olivier Flusin
- Subjects
Crimean–Congo hemorrhagic fever ,Male ,Physiology ,Fevers ,Monkeys ,Pathology and Laboratory Medicine ,Antibodies, Viral ,Communicable Diseases, Emerging ,Animal Cells ,Immune Physiology ,Testis ,Biology (General) ,Neglected tropical diseases ,Mammals ,0303 health sciences ,Innate Immune System ,Granuloma ,biology ,Latent tuberculosis ,030302 biochemistry & molecular biology ,Eukaryota ,Hematology ,3. Good health ,Actinobacteria ,Vertebrates ,Hemorrhagic Fever Virus, Crimean-Congo ,Granulomas ,Disease Progression ,Crimean-Congo hemorrhagic fever ,Infectious diseases ,Cytokines ,Viral disease ,Cellular Types ,Crimean Congo hemorrhagic fever virus ,Macaque ,Research Article ,Primates ,Tuberculosis ,QH301-705.5 ,Immune Cells ,Immunology ,Viremia ,Viral diseases ,Microbiology ,Virus ,Mycobacterium tuberculosis ,03 medical and health sciences ,Signs and Symptoms ,Diagnostic Medicine ,Latent Tuberculosis ,Virology ,Old World monkeys ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Medicine and health sciences ,Biology and life sciences ,Bacteria ,Host Microbial Interactions ,Tropical diseases ,Organisms ,Cell Biology ,RC581-607 ,Molecular Development ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,Macaca fascicularis ,Immune System ,Amniotes ,Parasitology ,Hemorrhagic Fever, Crimean ,Immunologic diseases. Allergy ,Viral hemorrhagic fevers ,Mycobacterium Tuberculosis ,Developmental Biology - Abstract
Crimean-Congo hemorrhagic fever (CCHF) is the most medically important tick-borne viral disease of humans and tuberculosis is the leading cause of death worldwide by a bacterial pathogen. These two diseases overlap geographically, however, concurrent infection of CCHF virus (CCHFV) with mycobacterial infection has not been assessed nor has the ability of virus to persist and cause long-term sequela in a primate model. In this study, we compared the disease progression of two diverse strains of CCHFV in the recently described cynomolgus macaque model. All animals demonstrated signs of clinical illness, viremia, significant changes in clinical chemistry and hematology values, and serum cytokine profiles consistent with CCHF in humans. The European and Asian CCHFV strains caused very similar disease profiles in monkeys, which demonstrates that medical countermeasures can be evaluated in this animal model against multiple CCHFV strains. We identified evidence of CCHFV persistence in the testes of three male monkeys that survived infection. Furthermore, the histopathology unexpectedly revealed that six additional animals had evidence of a latent mycobacterial infection with granulomatous lesions. Interestingly, CCHFV persisted within the granulomas of two animals. This study is the first to demonstrate the persistence of CCHFV in the testes and within the granulomas of non-human primates with concurrent latent tuberculosis. Our results have important public health implications in overlapping endemic regions for these emerging pathogens., Author summary CCHF is an emerging tick-borne viral disease that is endemic across much of Africa and Asia, and parts of Europe where its range and exposure risk to human populations is expanding. Tuberculosis threatens millions of lives world-wide and is the leading cause of death due to a bacterial pathogen. Concurrent mycobacterial infection with other infectious diseases has been described, but not for CCHFV despite the geographic overlap of these two pathogens. During our study we unexpectedly determined that some of the animals had latent tuberculosis and that CCHFV can persist within the granulomas. Furthermore, our study provides the first direct evidence that CCHFV can replicate and persist in the male genital tract, which has important implications for human sexual transmission. The ability of viral RNA to persist in immune-privileged sites or fluids has been described with increasing frequency for other emerging infectious diseases and can cause a burden on public health. This provides the impetus to utilize the model described here to better understand the mechanisms of CCHFV persistence and its effect on the development of long-term sequelae.
- Published
- 2019
26. Three asymptomatic animal infection models of hemorrhagic fever with renal syndrome caused by hantaviruses
- Author
-
Jay W. Hooper, Alicia M. Moreau, Sharon P. Daye, Joshua D. Shamblin, Casey C. Perley, Kelly S. Wetzel, Donald K. Nichols, Rebecca L. Brocato, and Steven A. Kwilas
- Subjects
0301 basic medicine ,Orthohantavirus ,Pulmonology ,Physiology ,Artificial Gene Amplification and Extension ,Monkeys ,Polymerase Chain Reaction ,Cricetinae ,Immune Physiology ,Chlorocebus aethiops ,Medicine and Health Sciences ,Neutralizing antibody ,Asymptomatic Infections ,Mammals ,Viral Genomics ,Multidisciplinary ,biology ,Infectious dose ,Eukaryota ,virus diseases ,Animal Models ,Genomics ,General Medicine ,Experimental Organism Systems ,Hemorrhagic Fever with Renal Syndrome ,Vertebrates ,Hamsters ,Viral Genome ,Medicine ,Female ,Puumala virus ,Anatomy ,medicine.symptom ,General Agricultural and Biological Sciences ,Research Article ,Primates ,Science ,030106 microbiology ,Viremia ,Microbial Genomics ,Research and Analysis Methods ,Rodents ,Microbiology ,Asymptomatic ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Virology ,Genetics ,medicine ,Animals ,Seroconversion ,Molecular Biology Techniques ,Vero Cells ,Molecular Biology ,Hantaan virus ,New World monkeys ,business.industry ,Organisms ,Ferrets ,Biology and Life Sciences ,Kidneys ,Renal System ,Reverse Transcriptase-Polymerase Chain Reaction ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,030104 developmental biology ,Amniotes ,Respiratory Infections ,Animal Studies ,biology.protein ,Marmosets ,Hantavirus Infection ,business ,Spleen - Abstract
Hantaan virus (HTNV) and Puumala virus (PUUV) are rodent-borne hantaviruses that are the primary causes of hemorrhagic fever with renal syndrome (HFRS) in Europe and Asia. The development of well characterized animal models of HTNV and PUUV infection is critical for the evaluation and the potential licensure of HFRS vaccines and therapeutics. In this study we present three animal models of HTNV infection (hamster, ferret and marmoset), and two animal models of PUUV infection (hamster, ferret). Infection of hamsters with a ~3 times the infectious dose 99% (ID(99)) of HTNV by the intramuscular and ~1 ID(99) of HTNV by the intranasal route leads to a persistent asymptomatic infection, characterized by sporadic viremia and high levels of viral genome in the lung, brain and kidney. In contrast, infection of hamsters with ~2 ID(99) of PUUV by the intramuscular or ~1 ID(99) of PUUV by the intranasal route leads to seroconversion with no detectable viremia, and a transient detection of viral genome. Infection of ferrets with a high dose of either HTNV or PUUV by the intramuscular route leads to seroconversion and gradual weight loss, though kidney function remained unimpaired and serum viremia and viral dissemination to organs was not detected. In marmosets a 1,000 PFU HTNV intramuscular challenge led to robust seroconversion and neutralizing antibody production. Similarly to the ferret model of HTNV infection, no renal impairment, serum viremia or viral dissemination to organs was detected in marmosets. This is the first report of hantavirus infection in ferrets and marmosets.
- Published
- 2019
- Full Text
- View/download PDF
27. Isolation of potent neutralizing antibodies from a survivor of the 2014 Ebola virus outbreak
- Author
-
Joshua D. Shamblin, Wen Li, Kathleen B. J. Pommert, Mark S. Klempner, Ashley E. Piper, Thomas R. Sprague, Pamela J. Glass, Arthur J. Goff, Colby A. Souders, Keith A. Reimann, Andrew B. Ward, Laura M. Walker, Karl Dane Wittrup, Erica Ollmann Saphire, Dennis R. Burton, Heidi L. Smith, Charles D. Murin, Eric Krauland, Tillman U. Gerngross, Suzanne E. Wollen, Lisa A. Cavacini, Devin Sok, Hannah L. Turner, Marnie L. Fusco, and Zachary A. Bornholdt
- Subjects
0301 basic medicine ,medicine.drug_class ,Antigen-Antibody Complex ,Antibodies, Viral ,medicine.disease_cause ,Monoclonal antibody ,Article ,Disease Outbreaks ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Viral Envelope Proteins ,medicine ,Animals ,Humans ,Survivors ,Ebola Vaccines ,Ebolavirus ,Multidisciplinary ,Ebola virus ,Ebola vaccine ,biology ,Immunization, Passive ,Virion ,Antibodies, Monoclonal ,Hemorrhagic Fever, Ebola ,Viral membrane ,Antibodies, Neutralizing ,Virology ,Tissue Donors ,Bundibugyo virus ,030104 developmental biology ,Immunization ,Antibody Formation ,Democratic Republic of the Congo ,biology.protein ,Antibody ,030217 neurology & neurosurgery - Abstract
Profiling the antibody response to Ebola The recent Ebola virus outbreak in West Africa illustrates the need not only for a vaccine but for potential therapies, too. One promising therapy is monoclonal antibodies that target Ebola's membrane-anchored glycoprotein (GP). Bornholdt et al. isolated and characterized 349 antibodies from a survivor of the 2014 outbreak. A large fraction showed some neutralizing activity and several were quite potent. Structural analysis revealed an important site of vulnerability on the membrane stalk region of GP. Antibodies targeting this area were therapeutically effective in Ebola virus–infected mice. Science , this issue p. 1078
- Published
- 2016
- Full Text
- View/download PDF
28. A conserved transcriptional response to intranasal ebolavirus exposure in non-human primates prior to onset of fever
- Author
-
Elyse R. Nagle, John H. Connor, Joshua D. Shamblin, Franco Rossi, Justine M. Zelko, Suzanne E. Wollen, Christina Burrows, Gustavo Palacios, Cheryl L. Taylor-Howell, Catherine E. Arnold, Emily Speranza, Sandra L. Bixler, Louis A. Altamura, Arthur J. Goff, William Aguilar, Timothy D. Minogue, and William D. Pratt
- Subjects
0301 basic medicine ,Ebola virus ,business.industry ,Infectious dose ,030106 microbiology ,Outbreak ,General Medicine ,Disease ,Hemorrhagic Fever, Ebola ,medicine.disease_cause ,Ebolavirus ,Virology ,Article ,Pathogenesis ,03 medical and health sciences ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Case fatality rate ,Gene expression ,medicine ,Animals ,business ,Gene ,Administration, Intranasal - Abstract
Ebola Virus Disease (EVD), caused by the Ebola virus (EBOV), is a severe illness characterized by case fatality rates up to 90%. The sporadic nature of outbreaks in resource-limited areas has hindered the ability to characterize the pathogenesis of EVD at all stages of infection, but particularly during early host responses. Pathogenesis is often studied in non-human primate (NHP) models of disease that replicate major aspects of human EVD. Typically, NHP exposures utilize a large infectious dose, are carried out through intramuscular or aerosol exposure, and have a fairly uniform disease course. In contrast, here we report our analysis of the host response to EBOV following intranasal exposure. Twelve cynomolgus macaques were infected with 100 plaque forming units of EBOV/Makona through intranasal exposure, and presented with varying time to onset of EVD. We utilized RNA-Sequencing and a newly developed NanoString codeset to monitor the host response via changes in RNA transcripts over time. When individual animal gene expression data were phased based on onset of sustained fever, the first clinical sign of severe disease, mathematical models indicated that interferon stimulated genes (ISGs) appeared as early as 4 days before fever onset. This demonstrates that lethal EVD has a uniform and predictable response to infection regardless of time-to-onset. Furthermore, expression of a subset of genes could predict disease development prior to other host-based indications of infection such as fever.
- Published
- 2018
29. Temporal Characterization of Marburg Virus Angola Infection following Aerosol Challenge in Rhesus Macaques
- Author
-
Miriam Botto, John H. Connor, Kenny L. Lin, Joshua C. Johnson, Anna N. Honko, Joshua D. Shamblin, Judy Y. Yen, Lisa E. Hensley, Carly Wlazlowski, Kathleen A. Cashman, Nancy A. Twenhafel, Heather L. Esham, Arthur J. Goff, and Ginger Donnelly
- Subjects
Time Factors ,Immunology ,Spleen ,Viremia ,Microbiology ,Marburg virus ,Virology ,medicine ,Animals ,Marburg Virus Disease ,Longitudinal Studies ,Leukocytosis ,Hypoalbuminemia ,Pathogen ,Aerosols ,biology ,Viral Load ,biology.organism_classification ,medicine.disease ,Immunohistochemistry ,Macaca mulatta ,Rhesus macaque ,medicine.anatomical_structure ,Marburgvirus ,Insect Science ,Host-Pathogen Interactions ,Disease Progression ,Cytokines ,Pathogenesis and Immunity ,medicine.symptom ,Viral load - Abstract
Marburg virus (MARV) infection is a lethal hemorrhagic fever for which no licensed vaccines or therapeutics are available. Development of appropriate medical countermeasures requires a thorough understanding of the interaction between the host and the pathogen and the resulting disease course. In this study, 15 rhesus macaques were sequentially sacrificed following aerosol exposure to the MARV variant Angola, with longitudinal changes in physiology, immunology, and histopathology used to assess disease progression. Immunohistochemical evidence of infection and resulting histopathological changes were identified as early as day 3 postexposure (p.e.). The appearance of fever in infected animals coincided with the detection of serum viremia and plasma viral genomes on day 4 p.e. High (>10 7 PFU/ml) viral loads were detected in all major organs (lung, liver, spleen, kidney, brain, etc.) beginning day 6 p.e. Clinical pathology findings included coagulopathy, leukocytosis, and profound liver destruction as indicated by elevated liver transaminases, azotemia, and hypoalbuminemia. Altered cytokine expression in response to infection included early increases in Th2 cytokines such as interleukin 10 (IL-10) and IL-5 and late-stage increases in Th1 cytokines such as IL-2, IL-15, and granulocyte-macrophage colony-stimulating factor (GM-CSF). This study provides a longitudinal examination of clinical disease of aerosol MARV Angola infection in the rhesus macaque model. IMPORTANCE In this study, we carefully analyzed the timeline of Marburg virus infection in nonhuman primates in order to provide a well-characterized model of disease progression following aerosol exposure.
- Published
- 2015
- Full Text
- View/download PDF
30. Virus-encoded miRNAs in Ebola virus disease
- Author
-
Eric M. Mucker, Miriam Botto, Suzanne E. Wollen-Roberts, Timothy D. Minogue, Moinya Coomber, Jean-Paul Gonzalez, Sandra L. Bixler, M. Louise M. Pitt, Anna N. Honko, Arthur J. Goff, Louis A. Altamura, Aileen O’Hearn, James Bangura, Nadia Wauquier, Randal J. Schoepp, Janice Duy, Justine M. Zelko, Joshua C. Johnson, and Joshua D. Shamblin
- Subjects
0301 basic medicine ,Science ,viruses ,Gene Expression ,medicine.disease_cause ,Virus Replication ,Virus ,Article ,03 medical and health sciences ,Mice ,microRNA ,medicine ,Animals ,Humans ,RNA, Messenger ,Infectivity ,Multidisciplinary ,Ebola virus ,biology ,Gene Expression Profiling ,RNA ,RNA virus ,Hemorrhagic Fever, Ebola ,biology.organism_classification ,Ebolavirus ,Virology ,Macaca mulatta ,Rhesus macaque ,Macaca fascicularis ,MicroRNAs ,030104 developmental biology ,Viral replication ,Medicine - Abstract
Ebola virus (EBOV) is a negative-strand RNA virus that replicates in the cytoplasm and causes an often-fatal hemorrhagic fever. EBOV, like other viruses, can reportedly encode its own microRNAs (miRNAs) to subvert host immune defenses. miRNAs are short noncoding RNAs that can regulate gene expression by hybridizing to multiple mRNAs, and viral miRNAs can enhance viral replication and infectivity by regulating host or viral genes. To date, only one EBOV miRNA has been examined in human infection. Here, we assayed mouse, rhesus macaque, cynomolgus macaque, and human samples infected with three EBOV variants for twelve computationally predicted viral miRNAs using RT-qPCR. Ten miRNAs aligned to EBOV variants and were detectable in the four species during disease with several viral miRNAs showing presymptomatic amplification in animal models. miRNA abundances in both the mouse and nonhuman primate models mirrored the human cohort, with miR-1-5p, miR-1-3p, and miR-T3-3p consistently at the highest levels. These striking similarities in the most abundant miRNAs during infection with different EBOV variants and hosts indicate that these miRNAs are potential valuable diagnostic markers and key effectors of EBOV pathogenesis.
- Published
- 2017
31. Temporal Progression of Lesions in Guinea Pigs Infected With Lassa Virus
- Author
-
M. E. Mattix, Eric R. Wilkinson, Xiankun Zeng, Shelley P. Honnold, Joshua D. Shamblin, Candace D. Blancett, Jeremy J. Bearss, Todd M. Bell, Ginger Donnelly, Kathleen A. Cashman, Keith A. Koistinen, and Carl I. Shaia
- Subjects
0301 basic medicine ,Male ,030106 microbiology ,Guinea Pigs ,Spleen ,Thymus Gland ,medicine.disease_cause ,Kidney ,West africa ,Viral hemorrhagic fever ,Pathogenesis ,03 medical and health sciences ,Lassa Fever ,Adrenal Glands ,medicine ,Animals ,Viremia ,Lassa fever ,Lassa virus ,Lung ,Skin ,General Veterinary ,business.industry ,Myocardium ,medicine.disease ,Virology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunization ,Liver ,Immunology ,Disease Progression ,Female ,Lymph Nodes ,business ,Pneumonia (non-human) - Abstract
Lassa virus (LASV) infection causes an acute, multisystemic viral hemorrhagic fever that annually infects an estimated 100 000 to 300 000 persons in West Africa. This pathogenesis study evaluated the temporal progression of disease in guinea pigs following aerosol and subcutaneous inoculation of the Josiah strain of LASV as well as the usefulness of Strain 13 guinea pigs as an animal model for Lassa fever. After experimental infection, guinea pigs ( Cavia porcellus; n = 67) were serially sampled to evaluate the temporal progression of infection, gross and histologic lesions, and serum chemistry and hematologic changes. Guinea pigs developed viremia on day 5 to 6 postexposure (PE), with clinical signs appearing by day 7 to 8 PE. Complete blood counts revealed lymphopenia and thrombocytopenia. Gross pathologic findings included skin lesions and congested lungs. Histologic lesions consisted of cortical lymphoid depletion by day 6 to 7 PE with lymphohistiocytic interstitial pneumonia at 7 to 8 days PE. Scattered hepatocellular degeneration and cell death were also noted in the liver and, to a lesser extent, in other tissues including the haired skin, lung, heart, adrenal gland, lymph nodes, thymus, and spleen. The first cell types to demonstrate staining for viral antigen were fibroblastic reticular cells and macrophages/dendritic cells in the lymph nodes on day 5 to 6 PE. This study demonstrates similarities between Lassa viral disease in human infections and experimental guinea pig infection. These shared pathologic characteristics support the utility of guinea pigs as an additional animal model for vaccine and therapeutic development under the Food and Drug Administration’s Animal Rule.
- Published
- 2017
32. Experimental Aerosolized Guinea Pig–Adapted Zaire Ebolavirus (Variant
- Author
-
T. E. Bunton, Suzanne E. Wollen, Nancy A. Twenhafel, D. R. Sizemore, M. M. Ogg, Carl I. Shaia, Sara C. Johnston, Joshua D. Shamblin, and L. M. Pitt
- Subjects
Male ,Zaire ebolavirus ,Guinea Pigs ,Spleen ,Biology ,medicine.disease_cause ,Guinea pig ,Macrophages, Alveolar ,medicine ,Animals ,Humans ,Interstitial pneumonia ,Lung ,Aerosolization ,Aerosols ,Ebolavirus ,General Veterinary ,Pneumonia ,Hemorrhagic Fever, Ebola ,respiratory system ,medicine.disease ,Virology ,Intracytoplasmic inclusion ,Disease Models, Animal ,medicine.anatomical_structure ,Liver ,Female ,Pneumonia (non-human) - Abstract
Eight guinea pigs were aerosolized with guinea pig–adapted Zaire ebolavirus (variant: Mayinga) and developed lethal interstitial pneumonia that was distinct from lesions described in guinea pigs challenged subcutaneously, nonhuman primates challenged by the aerosol route, and natural infection in humans. Guinea pigs succumbed with significant pathologic changes primarily restricted to the lungs. Intracytoplasmic inclusion bodies were observed in many alveolar macrophages. Perivasculitis was noted within the lungs. These changes are unlike those of documented subcutaneously challenged guinea pigs and aerosolized filoviral infections in nonhuman primates and human cases. Similar to findings in subcutaneously challenged guinea pigs, there were only mild lesions in the liver and spleen. To our knowledge, this is the first report of aerosol challenge of guinea pigs with guinea pig–adapted Zaire ebolavirus (variant: Mayinga). Before choosing this model for use in aerosolized ebolavirus studies, scientists and pathologists should be aware that aerosolized guinea pig–adapted Zaire ebolavirus (variant: Mayinga) causes lethal pneumonia in guinea pigs.
- Published
- 2014
- Full Text
- View/download PDF
33. The pathogenesis of genetically diverse strains of Crimean-Congo hemorrhagic fever virus in the cynomolgus macaque model
- Author
-
Justine M. Zelko, Charles J. Shoemaker, William D. Pratt, Keersten M. Ricks, Aura R. Garrison, Franco Rossi, Darci R. Smith, Collin J. Fitzpatrick, Joshua D. Shamblin, Connie S. Schmaljohn, Randal J. Schoepp, Jeffrey W. Koehler, O. Flusin, Matthew A. Voorhees, Adrienne E. Kimmel, and Joseph W. Golden
- Subjects
Microbiology (medical) ,Pathogenesis ,Infectious Diseases ,General Medicine ,Biology ,Cynomolgus macaque ,Virology ,Crimean Congo hemorrhagic fever virus - Published
- 2019
- Full Text
- View/download PDF
34. Low potential for mechanical transmission of Ebola virus via house flies (Musca domestica)
- Author
-
Justine M. Zelko, M. Louise M. Pitt, John C. Trefry, Farooq Nasar, Joshua D. Shamblin, Suzanne E. Wollen-Roberts, Sina Bavari, Xiankun Zeng, Kenneth J. Linthicum, Susana L. Padilla, Christopher W. Schellhase, Andrew D. Haddow, Elizabeth C. Grimes, and Roger D. Moon
- Subjects
0301 basic medicine ,Mechanical transmission ,030231 tropical medicine ,Short Report ,Biology ,Nose ,medicine.disease_cause ,Eye ,Fly ,lcsh:Infectious and parasitic diseases ,03 medical and health sciences ,High morbidity ,Feces ,Ebola virus ,0302 clinical medicine ,Houseflies ,medicine ,Animals ,Transmission ,lcsh:RC109-216 ,Muscid ,Mucous Membrane ,Transmission (medicine) ,fungi ,Fomite ,Mouth Mucosa ,Outbreak ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Mechanical ,Virology ,Filovirus ,Insect Vectors ,Macaca fascicularis ,030104 developmental biology ,Infectious Diseases ,Parasitology ,Face ,Musca domestica ,Musca - Abstract
Background Ebola virus (EBOV) infection results in high morbidity and mortality and is primarily transmitted in communities by contact with infectious bodily fluids. While clinical and experimental evidence indicates that EBOV is transmitted via mucosal exposure, the ability of non-biting muscid flies to mechanically transmit EBOV following exposure to the face had not been assessed. Results To investigate this transmission route, house flies (Musca domestica Linnaeus) were used to deliver an EBOV/blood mixture to the ocular/nasal/oral facial mucosa of four cynomolgus macaques (Macaca fascicularis Raffles). Following exposure, macaques were monitored for evidence of infection through the conclusion of the study, days 57 and 58. We found no evidence of systemic infection in any of the exposed macaques. Conclusions The results of this study indicate that there is a low potential for the mechanical transmission of EBOV via house flies - the conditions in this study were not sufficient to initiate infection.
- Published
- 2016
35. Development of a Novel Nonhuman Primate Model for Rift Valley Fever
- Author
-
Ginger Donnelly, Brian H. Bird, Darci R. Smith, Sara C. Johnston, Joshua D. Shamblin, Sarah E. McCarthy, Miriam Botto, Bridget Lewis, Ashley E. Keeney, Stuart T. Nichol, César G. Albariño, and Lisa E. Hensley
- Subjects
Rift Valley Fever ,Immunology ,Retinitis ,Viremia ,Microbiology ,Virus ,Virology ,biology.animal ,medicine ,Animals ,Humans ,Rift Valley fever ,Hepatitis ,Virulence ,biology ,Marmoset ,Callithrix ,Rift Valley fever virus ,medicine.disease ,biology.organism_classification ,Macaca mulatta ,Disease Models, Animal ,Insect Science ,Pathogenesis and Immunity ,Encephalitis - Abstract
Rift Valley fever (RVF) virus (RVFV) can cause severe human disease characterized by either acute-onset hepatitis, delayed-onset encephalitis, retinitis and blindness, or a hemorrhagic syndrome. The existing nonhuman primate (NHP) model for RVF utilizes an intravenous (i.v.) exposure route in rhesus macaques ( Macaca mulatta ). Severe disease in these animals is infrequent, and large cohorts are needed to observe significant morbidity and mortality. To overcome these drawbacks, we evaluated the infectivity and pathogenicity of RVFV in the common marmoset ( Callithrix jacchus ) by i.v., subcutaneous (s.c.), and intranasal exposure routes to more closely mimic natural exposure. Marmosets were more susceptible to RVFV than rhesus macaques and experienced higher rates of morbidity, mortality, and viremia and marked aberrations in hematological and chemistry values. An overwhelming infection of hepatocytes was a major consequence of infection of marmosets by the i.v. and s.c. exposure routes. Additionally, these animals displayed signs of hemorrhagic manifestations and neurological impairment. Based on our results, the common marmoset model more closely resembles severe human RVF disease and is therefore an ideal model for the evaluation of potential vaccines and therapeutics.
- Published
- 2012
- Full Text
- View/download PDF
36. The pathogenesis of Rift Valley fever virus in the mouse model
- Author
-
Joshua C. Johnson, Anna N. Honko, Maureen Kennedy, Darci R. Smith, Christopher Reed, Lisa E. Hensley, Keith E. Steele, Joshua D. Shamblin, and Jennifer Chapman
- Subjects
Rift Valley Fever ,Viremia ,Pathogenesis ,Biology ,Antibodies, Viral ,Virus ,Mouse model ,Mice ,Virology ,medicine ,Animals ,Humans ,Encephalitis, Viral ,Rift Valley fever ,Tropism ,Hepatitis ,medicine.disease ,Rift Valley fever virus ,Disease Models, Animal ,Liver ,Apoptosis ,Immunology ,Encephalitis - Abstract
Detailed studies describing the pathogenesis of Rift Valley fever (RVF) virus (RVFV) in the mouse model are lacking. A fully characterized small animal model of RVF is needed to evaluate potential vaccines and therapeutics. In this study, we characterized the pathogenesis of RVFV throughout the disease course in mice. Infection produced high-titer viremia and demonstrated RVFV tropism for a variety of tissue and individual cell types. Overwhelming infection of hepatocytes, accompanied by apoptosis, was a major consequence of infection. The majority of mice died or were euthanatized between days 3 and 6 postinfection with severe hepatitis. The remaining mice effectively cleared virus from the liver and blood, but exhibited neuroinvasion and developed panencephalitis. In addition, we characterized a number of other virological, clinicopathological, and histopathological features of RVFV infection in mice. The mouse model therefore mimics both the acute-onset hepatitis and delayed-onset encephalitis that are dominant features of severe human RVF.
- Published
- 2010
- Full Text
- View/download PDF
37. Attenuation and efficacy of live-attenuated Rift Valley fever virus vaccine candidates in non-human primates
- Author
-
Ginger Donnelly, Ashley E. Piper, Stuart T. Nichol, Miriam Botto, Lisa E. Hensley, Sara C. Johnston, Connie S. Schmaljohn, Darci R. Smith, Joshua D. Shamblin, César G. Albariño, and Brian H. Bird
- Subjects
RNA viruses ,0301 basic medicine ,Rift Valley Fever ,Physiology ,Drug Evaluation, Preclinical ,Human pathogen ,Disease ,Viral Nonstructural Proteins ,Monkeys ,Antibodies, Viral ,Biochemistry ,0302 clinical medicine ,Immune Physiology ,Bunyaviruses ,Medicine and Health Sciences ,Public and Occupational Health ,Enzyme-Linked Immunoassays ,Rift Valley fever ,Pathology and laboratory medicine ,Sequence Deletion ,Mammals ,Vaccines ,Immune System Proteins ,biology ,lcsh:Public aspects of medicine ,Viral Vaccine ,Vaccination ,Eukaryota ,Callithrix ,Animal Models ,Medical microbiology ,Vaccination and Immunization ,Infectious Diseases ,Experimental Organism Systems ,Veterinary Diseases ,Vertebrates ,Viruses ,Pathogens ,Research Article ,Primates ,lcsh:Arctic medicine. Tropical medicine ,Infectious Disease Control ,lcsh:RC955-962 ,Immunology ,030231 tropical medicine ,Viremia ,Vaccines, Attenuated ,Research and Analysis Methods ,Microbiology ,Antibodies ,03 medical and health sciences ,Vaccine Development ,medicine ,Animals ,Humans ,Antigens ,Immunoassays ,New World monkeys ,Biology and life sciences ,Organisms ,Viral pathogens ,Public Health, Environmental and Occupational Health ,Proteins ,Outbreak ,Viral Vaccines ,lcsh:RA1-1270 ,Rift Valley fever virus ,biology.organism_classification ,medicine.disease ,Virology ,Microbial pathogens ,Disease Models, Animal ,030104 developmental biology ,Amniotes ,Immunologic Techniques ,Veterinary Science ,Preventive Medicine ,Marmosets - Abstract
Rift Valley fever virus (RVFV) is an important mosquito-borne veterinary and human pathogen that has caused large outbreaks of severe disease throughout Africa and the Arabian Peninsula. Currently, no licensed vaccine or therapeutics exists to treat this potentially deadly disease. The explosive nature of RVFV outbreaks and the severe consequences of its accidental or intentional introduction into RVFV-free areas provide the impetus for the development of novel vaccine candidates for use in both livestock and humans. Rationally designed vaccine candidates using reverse genetics have been used to develop deletion mutants of two known RVFV virulence factors, the NSs and NSm genes. These recombinant viruses were demonstrated to be protective and immunogenic in rats, mice, and sheep, without producing clinical illness in these animals. Here, we expand upon those findings and evaluate the single deletion mutant (ΔNSs rRVFV) and double deletion mutant (ΔNSs-ΔNSm rRVFV) vaccine candidates in the common marmoset (Callithrix jacchus), a non-human primate (NHP) model resembling severe human RVF disease. We demonstrate that both the ΔNSs and ΔNSs-ΔNSm rRVFV vaccine candidates were found to be safe and immunogenic in the current study. The vaccinated animals received a single dose of vaccine that led to the development of a robust antibody response. No vaccine-induced adverse reactions, signs of clinical illness or infectious virus were detected in the vaccinated marmosets. All vaccinated animals that were subsequently challenged with RVFV were protected against viremia and liver disease. In summary, our results provide the basis for further development of the ΔNSs and ΔNSs-ΔNSm rRVFV as safe and effective human RVFV vaccines for this significant public health threat., Author summary Rift Valley fever (RVF) is an important neglected tropical disease that has caused severe epidemics and epizootics throughout Africa and the Arabian Peninsula. Severe outbreaks have involved tens of thousands of both human and livestock cases for which no effective, commercially available human vaccines are available. Vaccine candidates have been developed based on the complete deletion of two known RVF virus virulence factors, the NSs and NSm genes. These vaccines were previously demonstrated to be protective in rats, mice, and sheep. In this study, we expand upon those results and evaluate the vaccine candidates in a non-human primate model for RVF. The animals received a single dose of vaccine that led to the development of a robust immune response. No vaccine-induced adverse reactions, signs of clinical illness or infectious virus were detected in the vaccinated animals. All vaccinated animals that were subsequently challenged with RVF virus were protected against viremia and liver disease. These results demonstrate that the vaccines are safe and effective in non-human primates, which provides the impetus for further development of these candidates for use in humans.
- Published
- 2018
- Full Text
- View/download PDF
38. Lymphocyte Death in a Mouse Model of Ebola Virus Infection
- Author
-
Joan B. Geisbert, Joshua D. Shamblin, Lisa E. Hensley, Denise R. Braun, Aura R. Garrison, Jason Paragas, Steven B. Bradfute, and Thomas W. Geisbert
- Subjects
CD4-Positive T-Lymphocytes ,Primates ,Programmed cell death ,T-Lymphocytes ,Lymphocyte ,T cell ,CD8-Positive T-Lymphocytes ,Lymphocyte Depletion ,Mice ,medicine ,Animals ,Immunology and Allergy ,Lymphocytes ,Mononegavirales ,B cell ,Cell Death ,biology ,T lymphocyte ,Hemorrhagic Fever, Ebola ,biology.organism_classification ,Virology ,Killer Cells, Natural ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Apoptosis ,Immunology ,CD8 - Abstract
Background. A striking feature of Zaire Ebola virus (ZEBOV) infection in nonhuman primates is the rapid depletion of T and NK lymphocytes by apoptosis. In a mouse model of ZEBOV infection, lymphocyte death is a prominent finding; however, the mechanism of death and the lymphocyte subsets that are targeted remain unknown. Methods. We extended the characterization of lymphocyte death in a mouse model of ZEBOV infection by evaluating lymphocytes during the course of disease, using flow cytometry, electron microscopy, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL). Results. B cell, CD4 + and CD8 + T cell, and NK cell counts all dropped dramatically in the blood of infected BALB/c mice, and lymphocyte death was observed in the spleen by means of TUNEL staining and in the blood by means of electron microscopy. Morphologically, lymphocyte death occurred by both classic apoptosis and apoptosis-like programmed cell death. Conclusions. The early and severe loss of peripheral blood NK and CD8 + lymphocytes in ZEBOV-infected mice is similar to that seen in macaques. The morphological basis of lymphocyte death in ZEBOV-infected mice appears to be both classic apoptosis and apoptosis-like programmed cell death, although lymphocyte apoptosis in ZEBOV-infected nonhuman primates seems to occur primarily via classic apoptosis. The mouse model of ZEBOV infection may be useful for the screening of therapeutics directed against limiting lymphocyte death.
- Published
- 2007
- Full Text
- View/download PDF
39. Ebola Virus Infections in Nonhuman Primates Are Temporally Influenced by Glycoprotein Poly-U Editing Site Populations in the Exposure Material
- Author
-
M. Louise M. Pitt, Joshua D. Shamblin, John C. Trefry, Steven J. Kern, Gustavo Palacios, Jeffery R. Kugelman, Carol L. K. Sabourin, Taylor B. Chance, Farooq Nasar, Anna N. Honko, William D. Pratt, Jeremy J. Bearss, Dean J. Kobs, Jason T. Ladner, Suzanne E. Wollen, Michelle A. Jefferson, and Morgan Q.S. Wending
- Subjects
Poly U ,filovirus ,glycoprotein ,RNA editing ,medicine.medical_specialty ,Virulence Factors ,viruses ,lcsh:QR1-502 ,Statistical difference ,nonhuman primate ,Disease ,Biology ,medicine.disease_cause ,Injections, Intramuscular ,Ebola virus ,Kikwit ,pathogenesis ,animal model ,vaccine ,therapeutic ,Article ,lcsh:Microbiology ,Pathogenesis ,Viral Envelope Proteins ,Virology ,Survivorship curve ,Internal medicine ,medicine ,Animals ,chemistry.chemical_classification ,Hematology ,Hemorrhagic Fever, Ebola ,Survival Analysis ,Disease Models, Animal ,Macaca fascicularis ,Infectious Diseases ,chemistry ,Immunology ,Glycoprotein - Abstract
Recent experimentation with the variants of the Ebola virus that differ in the glycoprotein’s poly-uridine site, which dictates the form of glycoprotein produced through a transcriptional stutter, has resulted in questions regarding the pathogenicity and lethality of the stocks used to develop products currently undergoing human clinical trials to combat the disease. In order to address these concerns and prevent the delay of these critical research programs, we designed an experiment that permitted us to intramuscularly challenge statistically significant numbers of naïve and vaccinated cynomolgus macaques with either a 7U or 8U variant of the Ebola virus, Kikwit isolate. In naïve animals, no difference in survivorship was observed, however, there was a significant delay in the disease course between the two groups. Significant differences were also observed in time-of-fever, serum chemistry, and hematology. In vaccinated animals, there was no statistical difference in survivorship between either challenge groups, with two succumbing in the 7U group compared to 1 in the 8U challenge group. In summary, survivorship was not affected, but the Ebola virus disease course in nonhuman primates is temporally influenced by glycoprotein poly-U editing site populations.
- Published
- 2015
- Full Text
- View/download PDF
40. Dose Response of MARV/Angola Infection in Cynomolgus Macaques following IM or Aerosol Exposure
- Author
-
Miriam Botto, Suzanne E. Wollen, Jo Lynne Raymond, Kenny L. Lin, Arthur J. Goff, Carly Wlazlowski, Joshua D. Shamblin, Sara C. Johnston, Nancy A. Twenhafel, and Eric R. Wilkinson
- Subjects
medicine.medical_specialty ,lcsh:Medicine ,Kaplan-Meier Estimate ,Biology ,Cynomolgus macaque ,complex mixtures ,Injections, Intramuscular ,Marburg virus disease ,Case fatality rate ,medicine ,Animals ,Marburg Virus Disease ,lcsh:Science ,Aerosols ,Multidisciplinary ,Clinical pathology ,lcsh:R ,Significant difference ,Temperature ,Marburgvirus ,biology.organism_classification ,Virology ,Target dose ,Macaca fascicularis ,RNA, Viral ,lcsh:Q ,Histopathology ,Research Article - Abstract
Marburg virus infection in humans causes a hemorrhagic disease with a high case fatality rate. Countermeasure development requires the use of well-characterized animal models that mimic human disease. To further characterize the cynomolgus macaque model of MARV/Angola, two independent dose response studies were performed using the intramuscular or aerosol routes of exposure. All animals succumbed at the lowest target dose; therefore, a dose effect could not be determined. For intramuscular-exposed animals, 100 PFU was the first target dose that was not significantly different than higher target doses in terms of time to disposition, clinical pathology, and histopathology. Although a significant difference was not observed between aerosol-exposed animals in the 10 PFU and 100 PFU target dose groups, 100 PFU was determined to be the lowest target dose that could be consistently obtained and accurately titrated in aerosol studies.
- Published
- 2015
41. Pathogenesis of Bolivian Hemorrhagic Fever in Guinea Pigs
- Author
-
Kathleen A. Cashman, Todd M. Bell, Ginger Donnelly, C. I. Shaia, Shelley P. Honnold, Eric R. Wilkinson, J. W. Raymond, T. E. Bunton, and Joshua D. Shamblin
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Guinea Pigs ,Tracheobronchial lymph nodes ,Cavia ,Pallor ,Epithelium ,Hemorrhagic Fever, American ,Viral hemorrhagic fever ,03 medical and health sciences ,Adrenal Glands ,medicine ,Animals ,Humans ,Lung ,Pancreas ,Arenaviruses, New World ,Aerosols ,General Veterinary ,biology ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,Liver ,Bolivian hemorrhagic fever ,Immunology ,Female ,Lymph ,Lymph Nodes ,medicine.symptom ,Encephalitis - Abstract
Machupo virus, the cause of Bolivian hemorrhagic fever, is a highly lethal viral hemorrhagic fever with no Food and Drug Administration–approved vaccines or therapeutics. This study evaluated the guinea pig as a model using the Machupo virus–Chicava strain administered via aerosol challenge. Guinea pigs ( Cavia porcellus) were serially sampled to evaluate the temporal progression of infection, gross and histologic lesions, and sequential changes in serum chemistry and hematology. The incubation period was 5 to 12 days, and complete blood counts revealed leukopenia with lymphopenia and thrombocytopenia. Gross pathologic findings included congestion and hemorrhage of the gastrointestinal mucosa and serosa, noncollapsing lungs with fluid exudation, enlarged lymph nodes, and progressive pallor and friability of the liver. Histologic lesions consisted of foci of degeneration and cell death in the haired skin, liver, pancreas, adrenal glands, lymph nodes, tongue, esophagus, salivary glands, renal pelvis, small intestine, and large intestine. Lymphohistiocytic interstitial pneumonia was also present. Inflammation within the central nervous system, interpreted as nonsuppurative encephalitis, was histologically apparent approximately 16 days postexposure and was generally progressive. Macrophages in the tracheobronchial lymph node, on day 5 postexposure, were the first cells to demonstrate visible viral antigen. Viral antigen was detected throughout the lymphoid system by day 9 postexposure, followed by prominent spread within epithelial tissues and then brain. This study provides insight into the course of Machupo virus infection and supports the utility of guinea pigs as an additional animal model for vaccine and therapeutic development.
- Published
- 2015
42. Detailed analysis of the African green monkey model of Nipah virus disease
- Author
-
Todd M. Bell, Ginger Donnelly, W. Ian Lipkin, Heather L. Esham, Anna N. Honko, Joshua D. Shamblin, Lisa E. Hensley, Joshua C. Johnson, Thomas Briese, Sara C. Johnston, and William D. Pratt
- Subjects
medicine.medical_specialty ,Epidemiology ,lcsh:Medicine ,Nipah virus ,Disease ,Biology ,Communicable Diseases ,Cercopithecus aethiops ,Virology ,Chlorocebus aethiops ,medicine ,Animals ,lcsh:Science ,Plaque-forming unit ,Henipavirus Infections ,Public health ,Multidisciplinary ,Clinical pathology ,lcsh:R ,Malaysia ,medicine.disease ,Disease Models, Animal ,Pneumonia ,Diseases--Animal models ,Immunology ,Disease Progression ,Encephalitis ,Histopathology ,lcsh:Q ,African Green Monkey ,Research Article ,Systemic vasculitis - Abstract
Henipaviruses are implicated in severe and frequently fatal pneumonia and encephalitis in humans. There are no approved vaccines or treatments available for human use, and testing of candidates requires the use of well-characterized animal models that mimic human disease. We performed a comprehensive and statistically-powered evaluation of the African green monkey model to define parameters critical to disease progression and the extent to which they correlate with human disease. African green monkeys were inoculated by the intratracheal route with 2.5 × 10(4) plaque forming units of the Malaysia strain of Nipah virus. Physiological data captured using telemetry implants and assessed in conjunction with clinical pathology were consistent with shock, and histopathology confirmed widespread tissue involvement associated with systemic vasculitis in animals that succumbed to acute disease. In addition, relapse encephalitis was identified in 100% of animals that survived beyond the acute disease phase. Our data suggest that disease progression in the African green monkey is comparable to the variable outcome of Nipah virus infection in humans.
- Published
- 2015
43. Efficacy of tecovirimat (ST-246) in nonhuman primates infected with variola virus (Smallpox)
- Author
-
Robert C. Holman, Inger K. Damon, Nadia Gallardo, Cody Clemmons, Darin S. Carroll, Arthur J. Goff, Paul Hudson, Douglas W. Grosenbach, Joshua D. Shamblin, Jason M. Mehal, Victoria A. Olson, Eric M. Mucker, and Dennis E. Hruby
- Subjects
Male ,viruses ,Poxviridae Infections ,Isoindoles ,Antiviral Agents ,Random Allocation ,Medicine ,Smallpox ,Animals ,Pharmacology (medical) ,Orthopoxvirus ,Viral shedding ,Smallpox vaccine ,Pharmacology ,biology ,business.industry ,Tecovirimat ,Variola virus ,biology.organism_classification ,medicine.disease ,Virology ,Vaccination ,Clinical trial ,Infectious Diseases ,Treatment Outcome ,Immunology ,Benzamides ,Macaca ,business - Abstract
Naturally occurring smallpox has been eradicated but remains a considerable threat as a biowarfare/bioterrorist weapon (F. Fleck, Bull. World Health Organ. 81: 917–918, 2003). While effective, the smallpox vaccine is currently not recommended for routine use in the general public due to safety concerns ( http://www.bt.cdc.gov/agent/smallpox/vaccination ). Safe and effective countermeasures, particularly those effective after exposure to smallpox, are needed. Currently, SIGA Technologies is developing the small-molecule oral drug, tecovirimat (previously known as ST-246), as a postexposure therapeutic treatment of orthopoxvirus disease, including smallpox. Tecovirimat has been shown to be efficacious in preventing lethal orthopoxviral disease in numerous animal models (G. Yang, D. C. Pevear, M. H. Davies, M. S. Collett, T. Bailey, et al., J. Virol. 79: 13139–13149, 2005; D. C. Quenelle, R. M. Buller, S. Parker, K. A. Keith, D. E. Hruby, et al., Antimicrob. Agents Chemother., 51: 689–695, 2007; E. Sbrana, R. Jordan, D. E. Hruby, R. I. Mateo, S. Y. Xiao, et al., Am. J. Trop. Med. Hyg. 76: 768–773, 2007). Furthermore, in clinical trials thus far, the drug appears to be safe, with a good pharmacokinetic profile. In this study, the efficacy of tecovirimat was evaluated in both a prelesional and postlesional setting in nonhuman primates challenged intravenously with 1 × 10 8 PFU of Variola virus (VARV; the causative agent of smallpox), a model for smallpox disease in humans. Following challenge, 50% of placebo-treated controls succumbed to infection, while all tecovirimat-treated animals survived regardless of whether treatment was started at 2 or 4 days postinfection. In addition, tecovirimat treatment resulted in dramatic reductions in dermal lesion counts, oropharyngeal virus shedding, and viral DNA circulating in the blood. Although clinical disease was evident in tecovirimat-treated animals, it was generally very mild and appeared to resolve earlier than in placebo-treated controls that survived infection. Tecovirimat appears to be an effective smallpox therapeutic in nonhuman primates, suggesting that it is reasonably likely to provide therapeutic benefit in smallpox-infected humans.
- Published
- 2013
44. Aerosol exposure to Rift Valley fever virus causes earlier and more severe neuropathology in the murine model, which has important implications for therapeutic development
- Author
-
Aysegul Nalca, Kenny L. Lin, Gene G. Olinger, Ginger Donnelly, Brian M. Friedrich, Joshua D. Shamblin, Ashley E. Keeney, Catherine L. Wilhelmsen, Christopher Reed, Darci R. Smith, and Lisa E. Hensley
- Subjects
medicine.medical_specialty ,lcsh:Arctic medicine. Tropical medicine ,Rift Valley Fever ,lcsh:RC955-962 ,Retinitis ,Pathogenesis ,Biology ,Antiviral Agents ,Microbiology ,Virus ,Mice ,chemistry.chemical_compound ,Virology ,Emerging Viral Diseases ,Internal medicine ,Ribavirin ,medicine ,Animals ,Hepatitis ,Mice, Inbred BALB C ,Hematology ,Transmission (medicine) ,lcsh:Public aspects of medicine ,Public Health, Environmental and Occupational Health ,Peripheral Nervous System Diseases ,Viral Vaccines ,lcsh:RA1-1270 ,Rift Valley fever virus ,Antivirals ,medicine.disease ,Host-Pathogen Interaction ,Disease Models, Animal ,Animal Models of Infection ,Emerging Infectious Diseases ,Infectious Diseases ,Blood chemistry ,chemistry ,Immunology ,Female ,Encephalitis ,Research Article - Abstract
Rift Valley fever virus (RVFV) is an important mosquito-borne veterinary and human pathogen that can cause severe disease including acute-onset hepatitis, delayed-onset encephalitis, retinitis and blindness, or a hemorrhagic syndrome. Currently, no licensed vaccine or therapeutics exist to treat this potentially deadly disease. Detailed studies describing the pathogenesis of RVFV following aerosol exposure have not been completed and candidate therapeutics have not been evaluated following an aerosol exposure. These studies are important because while mosquito transmission is the primary means for human infection, it can also be transmitted by aerosol or through mucosal contact. Therefore, we directly compared the pathogenesis of RVFV following aerosol exposure to a subcutaneous (SC) exposure in the murine model by analyzing survival, clinical observations, blood chemistry, hematology, immunohistochemistry, and virus titration of tissues. Additionally, we evaluated the effectiveness of the nucleoside analog ribavirin administered prophylactically to treat mice exposed by aerosol and SC. The route of exposure did not significantly affect the survival, chemistry or hematology results of the mice. Acute hepatitis occurred despite the route of exposure. However, the development of neuropathology occurred much earlier and was more severe in mice exposed by aerosol compared to SC exposed mice. Mice treated with ribavirin and exposed SC were partially protected, whereas treated mice exposed by aerosol were not protected. Early and aggressive viral invasion of brain tissues following aerosol exposure likely played an important role in ribavirin's failure to prevent mortality among these animals. Our results highlight the need for more candidate antivirals to treat RVFV infection, especially in the case of a potential aerosol exposure. Additionally, our study provides an account of the key pathogenetic differences in RVF disease following two potential exposure routes and provides important insights into the development and evaluation of potential vaccines and therapeutics to treat RVFV infection., Author Summary Rift Valley fever (RVF) is an important neglected tropical disease that has caused severe epidemics and epizootics throughout Africa and the Arabian Peninsula. Severe outbreaks have involved tens of thousands of both human and livestock cases for which no effective, commercially available human vaccines or antiviral drugs are available. Mosquito transmission is the primary means for human infection although it can also be transmitted by aerosol or through mucosal contact. In this study, we directly compared the pathogenesis of RVF virus (RVFV) following aerosol exposure to a peripheral exposure in the mouse model. Additionally, we evaluated the effectiveness of ribavirin, a known antiviral compound, administered prior to virus exposure. The virus affected the liver despite the route of exposure. However, viral invasion of brain tissues occurred much earlier and was more severe in mice exposed by aerosol compared to peripherally exposed mice. Mice exposed peripherally and treated with ribavirin were partially protected whereas treated mice exposed by aerosol were not protected. Our results have important implications for understanding the pathogenesis of RVFV and for the development and evaluation of potential vaccines and therapeutics.
- Published
- 2013
45. Natural History of Aerosol Exposure with Marburg Virus in Rhesus Macaques
- Author
-
Joshua C. Johnson, Carly Wlazlowski, Ginger Donnelly, Nancy A. Twenhafel, Arthur J. Goff, Evan C. Ewers, Joshua D. Shamblin, Miriam Botto, Heather L. Esham, Lisa E. Hensley, and William D. Pratt
- Subjects
filovirus ,Male ,0301 basic medicine ,aerosol ,nonhuman primate ,Kidney Function Tests ,Article ,Marburg virus ,03 medical and health sciences ,Liver Function Tests ,Marburg virus disease ,Virology ,medicine ,Animals ,Marburg Virus Disease ,Viremia ,Blood coagulation test ,telemetry ,animal model ,medicine.diagnostic_test ,biology ,business.industry ,Acute kidney injury ,medicine.disease ,Marburgvirus ,biology.organism_classification ,Macaca mulatta ,Blood Cell Count ,030104 developmental biology ,Infectious Diseases ,Immunology ,Cytokines ,Female ,Blood Coagulation Tests ,Azotemia ,Liver function tests ,business ,Blood sampling - Abstract
Marburg virus causes severe and often lethal viral disease in humans, and there are currently no Food and Drug Administration (FDA) approved medical countermeasures. The sporadic occurrence of Marburg outbreaks does not allow for evaluation of countermeasures in humans, so therapeutic and vaccine candidates can only be approved through the FDA animal rule-a mechanism requiring well-characterized animal models in which efficacy would be evaluated. Here, we describe a natural history study where rhesus macaques were surgically implanted with telemetry devices and central venous catheters prior to aerosol exposure with Marburg-Angola virus, enabling continuous physiologic monitoring and blood sampling without anesthesia. After a three to four day incubation period, all animals developed fever, viremia, and lymphopenia before developing tachycardia, tachypnea, elevated liver enzymes, decreased liver function, azotemia, elevated D-dimer levels and elevated pro-inflammatory cytokines suggesting a systemic inflammatory response with organ failure. The final, terminal period began with the onset of sustained hypotension, dehydration progressed with signs of major organ hypoperfusion (hyperlactatemia, acute kidney injury, hypothermia), and ended with euthanasia or death. The most significant pathologic findings were marked infection of the respiratory lymphoid tissue with destruction of the tracheobronchial and mediastinal lymph nodes, and severe diffuse infection in the liver, and splenitis.
- Published
- 2016
- Full Text
- View/download PDF
46. Ultrastructural study of Rift Valley fever virus in the mouse model
- Author
-
Keith E. Steele, Anna N. Honko, Joshua D. Shamblin, Lisa E. Hensley, Darci R. Smith, and Christopher Reed
- Subjects
Central Nervous System ,Rift Valley Fever ,Lymphoid Tissue ,Apoptosis ,Pathogenesis ,Biology ,Virus Replication ,Viral hemorrhagic fever ,Mouse model ,Hepatitis ,Mice ,Microscopy, Electron, Transmission ,Virology ,medicine ,Electron microscopy ,Animals ,Humans ,Encephalitis, Viral ,Rift Valley fever ,Mice, Inbred BALB C ,Stem Cells ,medicine.disease ,Rift Valley fever virus ,Immunohistochemistry ,Disease Models, Animal ,Lymphatic system ,Viral replication ,Liver ,Ultrastructure ,Immunology ,Hepatocytes ,Female ,Stem cell ,Encephalitis - Abstract
Detailed ultrastructural studies of Rift Valley fever virus (RVFV) in the mouse model are needed to develop and characterize a small animal model of RVF for the evaluation of potential vaccines and therapeutics. In this study, the ultrastructural features of RVFV infection in the mouse model were analyzed. The main changes in the liver included the presence of viral particles in hepatocytes and hepatic stem cells accompanied by hepatocyte apoptosis. However, viral particles were observed rarely in the liver; in contrast, particles were extremely abundant in the CNS. Despite extensive lymphocytolysis, direct evidence of viral replication was not observed in the lymphoid tissue. These results correlate with the acute-onset hepatitis and delayed-onset encephalitis that are dominant features of severe human RVF, but suggest that host immune-mediated mechanisms contribute significantly to pathology. The results of this study expand our knowledge of RVFV–host interactions and further characterize the mouse model of RVF.
- Published
- 2012
47. A novel respiratory model of infection with monkeypox virus in cynomolgus macaques
- Author
-
Arthur J. Goff, Kenny L. Lin, Jason Paragas, Norman Kreiselmeier, Lisa E. Hensley, Carly Wlazlowski, Eric M. Mucker, Chad Foster, Jennifer Chapman, James V. Lawler, and Joshua D. Shamblin
- Subjects
viruses ,Immunology ,Disease ,Biology ,Microbiology ,Monkeypox ,Virology ,medicine ,Smallpox ,Animals ,Respiratory system ,Monkeypox virus ,Respiratory Tract Infections ,Respiratory tract infections ,Respiratory infection ,virus diseases ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,Macaca fascicularis ,Insect Science ,Pathogenesis and Immunity ,Viral disease - Abstract
Variola, the causative agent of smallpox, and the related monkeypox virus are both select agents that, if purposefully released, would cause public panic and social disruption. For this reason research continues in the areas of animal model and therapeutic development. Orthopoxviruses show a widely varying degree of host specificity, making development of accurate animal models difficult. In this paper, we demonstrate a novel respiratory infection technique that resulted in “classic” orthopox disease in nonhuman primates and takes the field of research one step closer to a better animal model.
- Published
- 2011
48. Susceptibility of Marmosets (Callithrix jacchus) to Monkeypox Virus: A Low Dose Prospective Model for Monkeypox and Smallpox Disease
- Author
-
Camenzind G. Robinson, Joshua D. Shamblin, John W. Huggins, Jennifer Chapman, Louis Huzella, Lisa E. Hensley, and Eric M. Mucker
- Subjects
Male ,viruses ,lcsh:Medicine ,Viremia ,Models, Biological ,Monkeypox ,Chlorocebus aethiops ,Hemorrhagic smallpox ,medicine ,Animals ,Smallpox ,Prospective Studies ,Monkeypox virus ,lcsh:Science ,Smallpox virus ,Vero Cells ,Multidisciplinary ,biology ,lcsh:R ,virus diseases ,Callithrix ,Variola virus ,Viral Load ,biology.organism_classification ,medicine.disease ,Virology ,Immunology ,lcsh:Q ,Disease Susceptibility ,Research Article - Abstract
Although current nonhuman primate models of monkeypox and smallpox diseases provide some insight into disease pathogenesis, they require a high titer inoculum, use an unnatural route of infection, and/or do not accurately represent the entire disease course. This is a concern when developing smallpox and/or monkeypox countermeasures or trying to understand host pathogen relationships. In our studies, we altered half of the test system by using a New World nonhuman primate host, the common marmoset. Based on dose finding studies, we found that marmosets are susceptible to monkeypox virus infection, produce a high viremia, and have pathological features consistent with smallpox and monkeypox in humans. The low dose (48 plaque forming units) required to elicit a uniformly lethal disease and the extended incubation (preclinical signs) are unique features among nonhuman primate models utilizing monkeypox virus. The uniform lethality, hemorrhagic rash, high viremia, decrease in platelets, pathology, and abbreviated acute phase are reflective of early-type hemorrhagic smallpox.
- Published
- 2015
- Full Text
- View/download PDF
49. Immunogenicity of a highly attenuated MVA smallpox vaccine and protection against monkeypox
- Author
-
Leigh Anne Eller, J. Charles Whitbeck, Eric M. Mucker, Joshua D. Shamblin, David A. Kulesh, Jeffrey L. Americo, Gary H. Cohen, Susan H. Zwiers, John W. Huggins, Bernard Moss, Peter B. Jahrling, Linda S. Wyatt, David M. Miller, Chris Hartmann, Roselyn J. Eisenberg, Patricia L. Earl, David L. Jackson, and Mark J. Martinez
- Subjects
viruses ,Vaccinia virus ,Chick Embryo ,CD8-Positive T-Lymphocytes ,Vaccines, Attenuated ,complex mixtures ,Cell Line ,Dryvax ,Monkeypox ,chemistry.chemical_compound ,Interferon-gamma ,Medicine ,Animals ,Humans ,Poxviridae ,Orthopoxvirus ,Monkeypox virus ,Smallpox vaccine ,Multidisciplinary ,biology ,business.industry ,ACAM2000 ,Fibroblasts ,Viral Load ,medicine.disease ,biology.organism_classification ,Virology ,Macaca fascicularis ,chemistry ,Immunology ,DNA, Viral ,Models, Animal ,Vaccinia ,business ,Smallpox Vaccine - Abstract
The potential use of smallpox as a biological weapon has led to the production and stockpiling of smallpox vaccine and the immunization of some healthcare workers. Another public health goal is the licensing of a safer vaccine that could benefit the millions of people advised not to take the current one because they or their contacts have increased susceptibility to severe vaccine side effects. As vaccines can no longer be tested for their ability to prevent smallpox, licensing will necessarily include comparative immunogenicity and protection studies in non-human primates. Here we compare the highly attenuated modified vaccinia virus Ankara (MVA) with the licensed Dryvax vaccine in a monkey model. After two doses of MVA or one dose of MVA followed by Dryvax, antibody binding and neutralizing titres and T-cell responses were equivalent or higher than those induced by Dryvax alone. After challenge with monkeypox virus, unimmunized animals developed more than 500 pustular skin lesions and became gravely ill or died, whereas vaccinated animals were healthy and asymptomatic, except for a small number of transient skin lesions in animals immunized only with MVA.
- Published
- 2003
50. The utilization of advance telemetry to investigate critical physiological parameters including electroencephalography in cynomolgus macaques following aerosol challenge with eastern equine encephalitis virus.
- Author
-
John C Trefry, Franco D Rossi, Michael V Accardi, Brandi L Dorsey, Thomas R Sprague, Suzanne E Wollen-Roberts, Joshua D Shamblin, Adrienne E Kimmel, Pamela J Glass, Lynn J Miller, Crystal W Burke, Anthony P Cardile, Darci R Smith, Sina Bavari, Simon Authier, William D Pratt, Margaret L Pitt, and Farooq Nasar
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Most alphaviruses are mosquito-borne and can cause severe disease in humans and domesticated animals. In North America, eastern equine encephalitis virus (EEEV) is an important human pathogen with case fatality rates of 30-90%. Currently, there are no therapeutics or vaccines to treat and/or prevent human infection. One critical impediment in countermeasure development is the lack of insight into clinically relevant parameters in a susceptible animal model. This study examined the disease course of EEEV in a cynomolgus macaque model utilizing advanced telemetry technology to continuously and simultaneously measure temperature, respiration, activity, heart rate, blood pressure, electrocardiogram (ECG), and electroencephalography (EEG) following an aerosol challenge at 7.0 log10 PFU. Following challenge, all parameters were rapidly and substantially altered with peak alterations from baseline ranged as follows: temperature (+3.0-4.2°C), respiration rate (+56-128%), activity (-15-76% daytime and +5-22% nighttime), heart rate (+67-190%), systolic (+44-67%) and diastolic blood pressure (+45-80%). Cardiac abnormalities comprised of alterations in QRS and PR duration, QTc Bazett, T wave morphology, amplitude of the QRS complex, and sinoatrial arrest. An unexpected finding of the study was the first documented evidence of a critical cardiac event as an immediate cause of euthanasia in one NHP. All brain waves were rapidly (~12-24 hpi) and profoundly altered with increases of up to 6,800% and severe diffuse slowing of all waves with decreases of ~99%. Lastly, all NHPs exhibited disruption of the circadian rhythm, sleep, and food/fluid intake. Accordingly, all NHPs met the euthanasia criteria by ~106-140 hpi. This is the first of its kind study utilizing state of the art telemetry to investigate multiple clinical parameters relevant to human EEEV infection in a susceptible cynomolgus macaque model. The study provides critical insights into EEEV pathogenesis and the parameters identified will improve animal model development to facilitate rapid evaluation of vaccines and therapeutics.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.