Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Methods Protoc ; 7(3)2024 May 15.
Article in English | MEDLINE | ID: mdl-38804336

ABSTRACT

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

2.
Sci Transl Med ; 11(492)2019 05 15.
Article in English | MEDLINE | ID: mdl-31092692

ABSTRACT

Western, Eastern, and Venezuelan equine encephalitis viruses (WEEV, EEEV, and VEEV, respectively) are important mosquito-borne agents that pose public health and bioterrorism threats. Despite considerable advances in understanding alphavirus replication, there are currently no available effective vaccines or antiviral treatments against these highly lethal pathogens. To develop a potential countermeasure for viral encephalitis, we generated a trivalent, or three-component, EEV vaccine composed of virus-like particles (VLPs). Monovalent VLPs elicited neutralizing antibody responses and protected mice and nonhuman primates (NHPs) against homologous challenges, but they were not cross-protective. In contrast, NHPs immunized with trivalent VLPs were completely protected against aerosol challenge by each of these three EEVs. Passive transfer of IgG from immunized NHPs protected mice against aerosolized EEV challenge, demonstrating that the mechanism of protection was humoral. Because they are replication incompetent, these trivalent VLPs represent a potentially safe and effective vaccine that can protect against diverse encephalitis viruses.


Subject(s)
Encephalitis Viruses/immunology , Encephalitis, Arbovirus/immunology , Encephalitis, Arbovirus/prevention & control , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Neutralizing/immunology , Encephalitis, Arbovirus/pathology , Encephalitis, Arbovirus/virology , Immunization , Immunoglobulin G/immunology , Macaca fascicularis , Mice, Inbred BALB C , Vaccines, Virus-Like Particle/ultrastructure
3.
Nat Microbiol ; 2: 17113, 2017 Jul 17.
Article in English | MEDLINE | ID: mdl-28715405

ABSTRACT

Ebola virus (EBOV) persistence in asymptomatic humans and Ebola virus disease (EVD) sequelae have emerged as significant public health concerns since the 2013-2016 EVD outbreak in Western Africa. Until now, studying how EBOV disseminates into and persists in immune-privileged sites was impossible due to the absence of a suitable animal model. Here, we detect persistent EBOV replication coinciding with systematic inflammatory responses in otherwise asymptomatic rhesus monkeys that had survived infection in the absence of or after treatment with candidate medical countermeasures. We document progressive EBOV dissemination into the eyes, brain and testes through vascular structures, similar to observations in humans. We identify CD68+ cells (macrophages/monocytes) as the cryptic EBOV reservoir cells in the vitreous humour and its immediately adjacent tissue, in the tubular lumina of the epididymides, and in foci of histiocytic inflammation in the brain, but not in organs typically affected during acute infection. In conclusion, our data suggest that persistent EBOV infection in rhesus monkeys could serve as a model for persistent EBOV infection in humans, and we demonstrate that promising candidate medical countermeasures may not completely clear EBOV infection. A rhesus monkey model may lay the foundation to study EVD sequelae and to develop therapies to abolish EBOV persistence.


Subject(s)
Asymptomatic Infections , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/virology , Africa, Western , Animals , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Brain/cytology , Brain/virology , Disease Models, Animal , Ebolavirus/isolation & purification , Epididymis/cytology , Epididymis/virology , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Humans , Macaca mulatta , Macrophages/virology , Male , Virus Replication , Vitreous Body/cytology , Vitreous Body/immunology , Vitreous Body/virology
4.
Vaccine ; 34(25): 2762-5, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27129427

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a New World alphavirus. VEEV is highly infectious in aerosolized form and has been identified as a bio-terrorism agent. There is no licensed vaccine for prophylaxis against VEEV. The current IND vaccine is poorly immunogenic and does not protect against an aerosol challenge with virulent VEEV. We have previously shown that VEEV inactivated by 1,5-iodonaphthyl azide (INA) protects against footpad challenge with virulent VEEV. In this study, we inactivated an attenuated strain of VEEV, V3526, with INA and evaluated its protective efficacy against aerosol challenge with wild type VEEV. We demonstrated that among three routes of immunization, intramuscular immunization with INA-inactivate V3526 (INA-iV3526) provided complete protection against aerosol challenge with virulent VEEV. Our data suggests that INA-iV3526 can be explored further for development as an effective vaccine candidate against aerosol challenge of virulent VEEV.


Subject(s)
Encephalomyelitis, Venezuelan Equine/prevention & control , Vaccination/methods , Viral Vaccines/immunology , Aerosols , Animals , Antibodies, Viral/blood , Azides/chemistry , Encephalitis Virus, Venezuelan Equine , Immunoglobulin G/blood , Mice , Neutralization Tests , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Vaccines/administration & dosage
5.
Virol J ; 12: 152, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26420265

ABSTRACT

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


Subject(s)
Alphavirus Infections/pathology , Disease Models, Animal , Encephalitis Virus, Eastern Equine/growth & development , Encephalitis Virus, Eastern Equine/pathogenicity , Administration, Inhalation , Administration, Intranasal , Alphavirus Infections/virology , Animal Structures/pathology , Animal Structures/virology , Animals , Body Fluids/virology , Cytokines/analysis , Female , Injections, Subcutaneous , Mice, Inbred BALB C , Survival Analysis , Viral Load
6.
Virol J ; 12: 154, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26423229

ABSTRACT

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


Subject(s)
Alphavirus Infections/pathology , Alphavirus Infections/virology , Disease Models, Animal , Encephalitis Virus, Eastern Equine/growth & development , Encephalitis Virus, Eastern Equine/physiology , Administration, Inhalation , Administration, Intranasal , Animal Structures/pathology , Animal Structures/virology , Animals , Female , Histocytochemistry , Immunohistochemistry , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Microscopy
7.
PLoS One ; 9(8): e104708, 2014.
Article in English | MEDLINE | ID: mdl-25116127

ABSTRACT

Currently, there are no FDA-licensed vaccines or therapeutics for eastern equine encephalitis virus (EEEV) for human use. We recently developed several methods to inactivate CVEV1219, a chimeric live-attenuated eastern equine encephalitis virus (EEEV). Dosage and schedule studies were conducted to evaluate the immunogenicity and protective efficacy of three potential second-generation inactivated EEEV (iEEEV) vaccine candidates in mice: formalin-inactivated CVEV1219 (fCVEV1219), INA-inactivated CVEV1219 (iCVEV1219) and gamma-irradiated CVEV1219 (gCVEV1219). Both fCVEV1219 and gCVEV1219 provided partial to complete protection against an aerosol challenge when administered by different routes and schedules at various doses, while iCVEV1219 was unable to provide substantial protection against an aerosol challenge by any route, dose, or schedule tested. When evaluating antibody responses, neutralizing antibody, not virus specific IgG or IgA, was the best correlate of protection. The results of these studies suggest that both fCVEV1219 and gCVEV1219 should be evaluated further and considered for advancement as potential second-generation inactivated vaccine candidates for EEEV.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Equine/immunology , Viral Vaccines/immunology , Animals , Azides/chemistry , Encephalitis Virus, Eastern Equine/classification , Encephalomyelitis, Equine/prevention & control , Female , Formaldehyde/chemistry , Gamma Rays , Immunity, Mucosal/immunology , Mice , Mice, Inbred BALB C , Vaccination , Vaccines, Attenuated/immunology
8.
Nature ; 508(7496): 402-5, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24590073

ABSTRACT

Filoviruses are emerging pathogens and causative agents of viral haemorrhagic fever. Case fatality rates of filovirus disease outbreaks are among the highest reported for any human pathogen, exceeding 90% (ref. 1). Licensed therapeutic or vaccine products are not available to treat filovirus diseases. Candidate therapeutics previously shown to be efficacious in non-human primate disease models are based on virus-specific designs and have limited broad-spectrum antiviral potential. Here we show that BCX4430, a novel synthetic adenosine analogue, inhibits infection of distinct filoviruses in human cells. Biochemical, reporter-based and primer-extension assays indicate that BCX4430 inhibits viral RNA polymerase function, acting as a non-obligate RNA chain terminator. Post-exposure intramuscular administration of BCX4430 protects against Ebola virus and Marburg virus disease in rodent models. Most importantly, BCX4430 completely protects cynomolgus macaques from Marburg virus infection when administered as late as 48 hours after infection. In addition, BCX4430 exhibits broad-spectrum antiviral activity against numerous viruses, including bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses. This is the first report, to our knowledge, of non-human primate protection from filovirus disease by a synthetic drug-like small molecule. We provide additional pharmacological characterizations supporting the potential development of BCX4430 as a countermeasure against human filovirus diseases and other viral diseases representing major public health threats.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Filoviridae Infections/prevention & control , Filoviridae Infections/virology , Filoviridae/drug effects , Purine Nucleosides/pharmacology , Adenine/analogs & derivatives , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Disease Models, Animal , Ebolavirus/drug effects , Filoviridae/enzymology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Humans , Injections, Intramuscular , Macaca fascicularis/virology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/virology , Marburgvirus/drug effects , Purine Nucleosides/administration & dosage , Purine Nucleosides/chemistry , Purine Nucleosides/pharmacokinetics , Pyrrolidines , RNA/biosynthesis , Time Factors
9.
J Vet Diagn Invest ; 24(4): 794-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22621943

ABSTRACT

A 2-year-old gelding presented with a history of lethargy and anorexia. Physical examination revealed pleural and abdominal fluid, as well as several masses in the scrotum. The horse became acutely dyspneic despite 7 days of supportive care. Because of the poor prognosis, the owners elected euthanasia. Gross necropsy findings included multiple masses in the scrotum and inguinal canals and along the dorsal peritoneal cavity. The neoplasm infiltrated the kidneys, liver, spleen, mesenteric lymph nodes, mesentery, and abdominal surface of the diaphragm. Histologically, the neoplasm is composed of spindle to round cells arranged in densely cellular areas, vague streams, and rare rosettes. Neoplastic cells were immunoreactive for S-100 protein, glial fibrillary acidic protein, neuron-specific enolase, neurofilament protein, and synaptophysin. Based on gross, histological, and immunohistochemical findings, a diagnosis of peripheral primitive neuroectodermal tumor was made. Primitive neuroectodermal tumors are rarely described in horses that were associated with the eyes.


Subject(s)
Horse Diseases/pathology , Neuroectodermal Tumors, Primitive, Peripheral/veterinary , Peritoneal Neoplasms/veterinary , Animals , Fatal Outcome , Horses , Immunohistochemistry/veterinary , Male , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Peritoneal Neoplasms/pathology
10.
Virol J ; 8: 197, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529366

ABSTRACT

BACKGROUND: Neuroinvasion of Venezuelan equine encephalitis virus (VEEV) and subsequent initiation of inflammation in the brain plays a crucial role in the outcome of VEEV infection in mice. Adhesion molecules expressed on microvascular endothelial cells in the brain have been implicated in the modulation of the blood brain barrier (BBB) and inflammation in brain but their role in VEEV pathogenesis is not very well understood. In this study, we evaluated the expression of extracellular matrix and adhesion molecules genes in the brain of VEEV infected mice. FINDINGS: Several cell to cell adhesion molecules and extracellular matrix protein genes such as ICAM-1, VCAM-1, CD44, Cadherins, integrins, MMPs and Timp1 were differentially regulated post-VEEV infection. ICAM-1 knock-out (IKO) mice infected with VEEV had markedly reduced inflammation in the brain and demonstrated a delay in the onset of clinical symptoms of disease. A differential regulation of inflammatory genes was observed in the IKO mice brain compared to their WT counterparts. CONCLUSIONS: These results improve our present understanding of VEEV induced inflammation in mouse brain.


Subject(s)
Brain/pathology , Brain/virology , Cell Adhesion Molecules/biosynthesis , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/pathology , Inflammation/pathology , Animals , Disease Models, Animal , Encephalomyelitis, Venezuelan Equine/virology , Gene Expression Profiling , Histocytochemistry , Immunohistochemistry , Mice , Mice, Knockout , Microscopy , Rodent Diseases/pathology , Rodent Diseases/virology
11.
J Zoo Wildl Med ; 41(3): 530-4, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20945656

ABSTRACT

A 6-yr-old captive-born female fennec fox (Vulpes zerda) had a history of multiple seizures and was treated with diazepam and phenobarbital therapy. Despite medical treatment, the seizures continued. They were intermittent and progressive, resulting in neurologic deficits and death of the animal within 6 mo of onset of the clinical signs. At necropsy, the animal was in good nutritional condition, and no gross lesions were noted in the brain. Histologically, amphophilic to basophilic, periodic acid-Schiff (PAS) positive, diastase-resistant inclusions were present in the brain, heart, and liver. Ultrastructurally, the inclusions were variably electron dense, fibrillary to occasionally granular, and non-membrane bound. The clinical, histologic, and ultrastructural findings were consistent with Lafora's disease, which in humans is a rare, fatal, autosomal recessive hereditary neurometabolic disorder characterized by progressive myoclonic epilepsy. This is the first report of Lafora's-like disease in a fennec fox.


Subject(s)
Foxes , Lafora Disease/veterinary , Animals , Fatal Outcome , Female , Lafora Disease/pathology
12.
J Zoo Wildl Med ; 38(3): 479-82, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17939360

ABSTRACT

A 12-yr-old, intact male squirrel (Sciurus sp.) presented with a 15 mm-by-20 mm area of alopecia and plaque-like dermal thickening over the left caudolateral thorax. Routine diagnostic tests ruled out more common conditions that result in alopecia, such as dermatophytosis and acariasis. A punch biopsy was obtained under anesthesia and submitted for histopathologic evaluation. The diagnosis of epitheliotropic lymphoma was made, and follow-up surgical excision was performed. Histopathologic features were consistent with epitheliotropic lymphoma, and immunohistochemistry confirmed a T-cell origin. There was no local recurrence, new lesions, or evidence of metastasis 10 mo after surgical excision. To our knowledge, to date, epitheliotropic lymphoma has not been described in a squirrel.


Subject(s)
Mycosis Fungoides/veterinary , Sciuridae , Skin Neoplasms/veterinary , Animals , Male , Mycosis Fungoides/epidemiology , Mycosis Fungoides/pathology , Mycosis Fungoides/surgery , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Treatment Outcome
13.
J Parasitol ; 91(3): 695-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16108571

ABSTRACT

Toxoplasma gondii infection in marine mammals is intriguing and indicative of contamination of the ocean environment with oocysts. T. gondii was identified in a Hawaiian monk seal (Monachus schauinslandi) that had visceral and cerebral lesions. Tachyzoites were found in the lymph nodes, spleen, diaphragm, heart, adrenal glands, and brain. A few tissue cysts were found in sections of the cerebrum. The diagnosis was confirmed serologically, by immunohistochemical staining with T. gondii-specific polyclonal rabbit serum, and by the detection of T. gondii DNA. The genotype was determined to be type III by restriction fragment length polymorphisms of the SAG2 gene. This is the first report of T. gondii infection in a Hawaiian monk seal.


Subject(s)
Seals, Earless/parasitology , Toxoplasma/isolation & purification , Toxoplasmosis, Animal/diagnosis , Adrenal Cortex/parasitology , Adrenal Cortex/pathology , Animals , Brain/parasitology , Brain/pathology , DNA, Protozoan/analysis , Diaphragm/parasitology , Diaphragm/pathology , Genotype , Hawaii , Heart/parasitology , Immunohistochemistry/veterinary , Lymph Nodes/parasitology , Lymph Nodes/pathology , Male , Spleen/parasitology , Spleen/pathology , Toxoplasma/classification , Toxoplasma/genetics , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...