Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
PLoS Pathog ; 20(6): e1012343, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935789

ABSTRACT

Rift Valley fever virus (RVFV) is an encephalitic bunyavirus that can infect neurons in the brain. There are no approved therapeutics that can protect from RVFV encephalitis. Innate immunity, the first line of defense against infection, canonically antagonizes viruses through interferon signaling. We found that interferons did not efficiently protect primary cortical neurons from RVFV, unlike other cell types. To identify alternative neuronal antiviral pathways, we screened innate immune ligands and discovered that the TLR2 ligand Pam3CSK4 inhibited RVFV infection, and other bunyaviruses. Mechanistically, we found that Pam3CSK4 blocks viral fusion, independent of TLR2. In a mouse model of RVFV encephalitis, Pam3CSK4 treatment protected animals from infection and mortality. Overall, Pam3CSK4 is a bunyavirus fusion inhibitor active in primary neurons and the brain, representing a new approach toward the development of treatments for encephalitic bunyavirus infections.


Subject(s)
Lipopeptides , Neurons , Rift Valley Fever , Rift Valley fever virus , Animals , Rift Valley fever virus/drug effects , Mice , Lipopeptides/pharmacology , Rift Valley Fever/virology , Rift Valley Fever/prevention & control , Neurons/metabolism , Neurons/virology , Mice, Inbred C57BL , Humans , Immunity, Innate/drug effects , Encephalitis, Viral/virology , Encephalitis, Viral/immunology , Encephalitis, Viral/prevention & control , Encephalitis, Viral/drug therapy , Antiviral Agents/pharmacology
2.
J Virol ; 95(23): e0150621, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495703

ABSTRACT

Rift Valley fever virus (RVFV) is an arbovirus found throughout Africa. It causes disease that is typically mild and self-limiting; however, some infected individuals experience severe manifestations, including hepatitis, encephalitis, or even death. Reports of RVFV encephalitis are notable among immunosuppressed individuals, suggesting a role for adaptive immunity in preventing this severe complication. This phenomenon has been modeled in C57BL/6 mice depleted of CD4 T cells prior to infection with DelNSs RVFV (RVFV containing a deletion of nonstructural protein NSs), resulting in late-onset encephalitis accompanied by high levels of viral RNA in the brain in 30% of animals. In this study, we sought to define the specific type(s) of CD4 T cells that mediate protection from RVFV encephalitis. The viral epitopes targeted by CD4 and CD8 T cells were defined in C57BL/6 mice, and tetramers for both CD4 and CD8 T cells were generated. RVFV-specific CD8 T cells were expanded and of a cytotoxic and proliferating phenotype in the liver following infection. RVFV-specific CD4 T cells were identified in the liver and spleen following infection and phenotyped as largely Th1 or Tfh subtypes. Knockout mice lacking various aspects of pathways important in Th1 and Tfh development and function were used to demonstrate that T-bet, CD40, CD40L, and major histocompatibility complex class II (MHC-II) mediated protection from RVFV encephalitis, while gamma interferon (IFN-γ) and interleukin-12 (IL-12) were dispensable. Virus-specific antibody responses correlated with protection from encephalitis in all mouse strains, suggesting that Tfh/B cell interactions modulate clinical outcome in this model. IMPORTANCE The prevention of RVFV encephalitis requires intact adaptive immunity. In this study, we developed reagents to detect RVFV-specific T cells and provide evidence for Tfh cells and CD40/CD40L interactions as critical mediators of this protection.


Subject(s)
CD40 Antigens , CD40 Ligand , Encephalitis, Viral/prevention & control , Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Rift Valley fever virus/physiology , T-Lymphocytes/immunology , Africa , Animals , Antibody Formation , B-Lymphocytes/immunology , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Epitopes , Female , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
PLoS Pathog ; 14(1): e1006822, 2018 01.
Article in English | MEDLINE | ID: mdl-29352287

ABSTRACT

Emergency hematopoiesis facilitates the rapid expansion of inflammatory immune cells in response to infections by pathogens, a process that must be carefully regulated to prevent potentially life threatening inflammatory responses. Here, we describe a novel regulatory role for the cytokine IFNγ that is critical for preventing fatal encephalitis after viral infection. HSV1 encephalitis (HSE) is triggered by the invasion of the brainstem by inflammatory monocytes and neutrophils. In mice lacking IFNγ (GKO), we observed unrestrained increases in G-CSF levels but not in GM-CSF or IL-17. This resulted in uncontrolled expansion and infiltration of apoptosis-resistant, degranulating neutrophils into the brainstem, causing fatal HSE in GKO but not WT mice. Excessive G-CSF in GKO mice also induced granulocyte derived suppressor cells, which inhibited T-cell proliferation and function, including production of the anti-inflammatory cytokine IL-10. Unexpectedly, we found that IFNγ suppressed G-CSF signaling by increasing SOCS3 expression in neutrophils, resulting in apoptosis. Depletion of G-CSF, but not GM-CSF, in GKO mice induced neutrophil apoptosis and reinstated IL-10 secretion by T cells, which restored their ability to limit innate inflammatory responses resulting in protection from HSE. Our studies reveals a novel, complex interplay among IFNγ, G-CSF and IL-10, which highlights the opposing roles of G-CSF and IFNγ in regulation of innate inflammatory responses in a murine viral encephalitis model and reveals G-CSF as a potential therapeutic target. Thus, the antagonistic G-CSF-IFNγ interactions emerge as a key regulatory node in control of CNS inflammatory responses to virus infection.


Subject(s)
Cell Proliferation/genetics , Central Nervous System/virology , Chemotaxis, Leukocyte/genetics , Encephalitis, Viral/prevention & control , Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Interferon-gamma/physiology , Neutrophils/physiology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Central Nervous System/drug effects , Central Nervous System/metabolism , Chemotaxis, Leukocyte/drug effects , Encephalitis, Viral/genetics , Encephalitis, Viral/immunology , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/physiology , Interferon-gamma/genetics , Interferon-gamma/pharmacology , Male , Mice , Mice, Knockout , Neutrophils/drug effects
4.
Transpl Infect Dis ; 22(4): e13317, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32386074

ABSTRACT

BACKGROUND: Minimal data exist describing the epidemiology, management, and long-term graft outcomes after West Nile viral disease in kidney transplant recipients (KTRs). METHODS: Single-center observational cohort study of patients who received a kidney transplant between 1/1/1994 and 12/31/2018 and developed WNV at any time point after transplantation. RESULTS: During the 24-year study period, 11 patients had documented WNV infection. Seven patients were recipients of a kidney transplant alone, and four had a simultaneous kidney and pancreas transplant. The mean age at the time of transplant was 44.7 ± 17.1 years, and the mean age at the time of WNV infection was 48 ± 17.2 years. All patients received lymphocyte depleting induction at transplant (alemtuzumab (n = 2), OKT3 (n = 1), or anti-thymocyte globulin (n = 8)). The mean time from transplant to WNV infection was 3.4 ± 5.4 years, and none was suspected of having a donor-derived infection. Three patients were treated for rejection in the 6 months before infection. The most common presenting symptom was altered mental status (n = 7), followed by a combination of fever and headache (n = 4). All patients had detectable serum WNV IgM antibodies at the time of diagnosis. All patients had a reduction in their immunosuppression and received supportive care; only two patients were treated with intravenous immunoglobulins. Nine patients recovered with no residual deficit; however, two suffered permanent neurologic damage. The mean estimated glomerular filtration rate drop at 1 year after the infection was 8.4 ± 13 mL/min/1.73 m2 . Three patients suffered acute rejection within 1 year after the infection episode, likely attributable to aggressive immunosuppressive reduction. The mean follow-up after the infection was 5.1 ± 4.3 years. At last follow-up, two patients lost their kidney allograft, and five patients died. None of the graft losses or deaths occurred within a year of the WNV or were directly attributable to WNV. CONCLUSION: The majority of patients with WNV infection after KTR recovered fully with supportive care and immunosuppressive adjustment without residual neurologic sequelae. Additionally, WNV infection was associated with relatively small reductions in eGFR at 1 year.


Subject(s)
Disease Management , Encephalitis, Viral/epidemiology , Kidney Transplantation/adverse effects , Transplant Recipients/statistics & numerical data , West Nile Fever/epidemiology , Adult , Aged , Cohort Studies , Encephalitis, Viral/prevention & control , Female , Humans , Immunocompromised Host , Immunosuppression Therapy , Male , Middle Aged , West Nile Fever/complications , West Nile Fever/prevention & control , West Nile virus/pathogenicity
5.
J Infect Dis ; 220(8): 1302-1306, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31199457

ABSTRACT

We report a case of cytomegalovirus encephalitis in a hematopoietic stem cell transplant recipient. A previously uncharacterized V787E mutation in UL54 was identified in cerebrospinal fluid but not plasma specimens. For the V787E recombinant virus, the half maximal effective concentrations for ganciclovir, foscarnet, and cidofovir were 8.6-, 3.4- and 2.9-fold higher than for wild-type virus, and the replicative capacity was lower. The introduction of a bulkier and negatively charged glutamate residue at position 787 could destabilize the finger domain of UL54 DNA polymerase. Viral genotyping of cerebrospinal fluid is warranted in subjects with cytomegalovirus encephalitis, owing to the low penetration of antivirals in this compartment.


Subject(s)
Cytomegalovirus Infections/diagnosis , Cytomegalovirus/genetics , Drug Resistance, Multiple, Viral/genetics , Encephalitis, Viral/diagnosis , Hematopoietic Stem Cell Transplantation/adverse effects , Acyclovir/pharmacology , Acyclovir/therapeutic use , Antibiotic Prophylaxis/methods , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cerebrospinal Fluid/virology , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/cerebrospinal fluid , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Encephalitis, Viral/cerebrospinal fluid , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Female , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immediate-Early Proteins/genetics , Immunosuppression Therapy/adverse effects , Middle Aged , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Viral Proteins/genetics
6.
J Virol ; 92(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30258000

ABSTRACT

Rift Valley fever virus (RVFV) is an arbovirus that causes disease in livestock and humans in Africa and the Middle East. While human disease is typically mild and self-limiting, some individuals develop severe manifestations, such as hepatitis, hemorrhagic fever, or encephalitis. Encephalitis occurs 2 to 3 weeks after acute illness; therefore, we hypothesized that it was a result of an inadequate adaptive immunity. To test this hypothesis in vivo, we used an attenuated virus (DelNSsRVFV) that does not typically cause disease in mice. We first characterized the normal immune response to infection with DelNSsRVFV in immunocompetent mice and noted expansion of natural killer cells and monocytes, as well as activation of both CD8 and CD4 T cells. Depleting C57BL/6 mice of CD4 T cells prior to DelNSsRVFV infection resulted in encephalitis in 30% of the mice; in encephalitic mice, we noted infiltration of T cells and inflammatory monocytes into the brain. CD4 and CD8 codepletion in C57BL/6 mice, as well as CD4 depletion in CCR2 knockout mice, increased the frequency of encephalitis, demonstrating that these cell types normally contributed to the prevention of disease. Encephalitic mice had similar viral RNA loads in the brain regardless of which cell types were depleted, suggesting that CD4 T cells, CD8 T cells, and inflammatory monocytes did little to control viral replication in the brain. CD4-depleted mice exhibited diminished humoral and T cell memory responses, suggesting that these immune mechanisms contributed to peripheral control of virus, thus preventing infection of the brain.IMPORTANCE RVFV is found in Africa and the Middle East and is transmitted by mosquitos or through contact with infected animals. Infected individuals can develop mild disease or more severe forms, such as hepatitis or encephalitis. In order to understand why some individuals develop encephalitis, we first need to know which immune functions protect those who do not develop this form of disease. In this study, we used a mouse model of RVFV infection to demonstrate that CD4 T cells, CD8 T cells, and monocytes all contribute to prevention of encephalitis. Their likely mechanism of action is preventing RVFV from ever reaching the brain.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Encephalitis, Viral/prevention & control , Monocytes/metabolism , Rift Valley Fever/prevention & control , Rift Valley fever virus/immunology , Animals , Brain/immunology , Brain/virology , Encephalitis, Viral/immunology , Immunity, Humoral , Immunity, Innate , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Rift Valley Fever/immunology , Rift Valley fever virus/pathogenicity
7.
Biol Blood Marrow Transplant ; 24(6): 1264-1273, 2018 06.
Article in English | MEDLINE | ID: mdl-29454651

ABSTRACT

Cord blood transplantation (CBT) is a distinct risk factor for human herpesvirus-6 (HHV-6) reactivation and HHV-6 encephalitis. In a prospective multicenter trial we investigated the effects of prophylactic foscarnet (90 mg/kg i.v. infusion from days 7 to 27 after CBT) on the occurrence of HHV-6 reactivation, HHV-6 encephalitis, and acute graft-versus-host disease (aGVHD) in CBT recipients. Between 2014 and 2016, 57 patients were included in a foscarnet-prophylaxis group. Outcomes were compared with an historical control group who received CBT between 2010 and 2014 (standard-treatment group, n = 63). The cumulative incidence of high-level HHV-6 reactivation, defined as plasma HHV-6 DNA ≥ 104 copies/mL, at 60 days after CBT was significantly lower in the foscarnet-prophylaxis group than in the standard-treatment group (18.3% versus 57.3%, P < .001). Multivariate analysis revealed that myeloablative preconditioning and standard treatment were significant risk factors for high-level HHV-6 reactivation. The cumulative incidence of HHV-6 encephalitis at 60 days after CBT was not different between the groups (foscarnet-prophylaxis group, 12.4%; standard-treatment group, 4.9%; P = .14). The cumulative incidences of grades II to IV and grades III to IV aGVHD at 60 days after CBT were not different between the groups (grades II to IV aGVHD: foscarnet-prophylaxis group, 42.0%; standard-treatment group, 40.5%; P = .96; grades III to IV aGVHD: foscarnet-prophylaxis group, 14.5%; standard-treatment group, 14.5%; P = 1.00). In the setting of this study foscarnet significantly suppressed systemic HHV-6 reactivation in CBT recipients but failed to prevent the development of HHV-6 encephalitis. Suppression of HHV-6 reactivation by foscarnet did not show any effects against the incidence of aGVHD.


Subject(s)
Encephalitis, Viral/prevention & control , Foscarnet/pharmacology , Herpesvirus 6, Human/drug effects , Virus Activation/drug effects , Adolescent , Adult , Aged , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , DNA, Viral/blood , Encephalitis, Viral/drug therapy , Female , Fetal Blood/transplantation , Foscarnet/therapeutic use , Graft vs Host Disease , Historically Controlled Study , Humans , Middle Aged , Myeloablative Agonists/pharmacology , Premedication/methods , Prospective Studies , Risk Factors , Treatment Outcome , Young Adult
9.
J Neuroinflammation ; 11: 180, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25391297

ABSTRACT

BACKGROUND: Theiler's murine encephalomyelitis virus (TMEV) infection represents a commonly used infectious animal model to study various aspects of the pathogenesis of multiple sclerosis (MS). In susceptible SJL mice, dominant activity of Foxp3(+) CD4(+) regulatory T cells (Tregs) in the CNS partly contributes to viral persistence and progressive demyelination. On the other hand, resistant C57BL/6 mice rapidly clear the virus by mounting a strong antiviral immune response. However, very little is known about the role of Tregs in regulating antiviral responses during acute encephalitis in resistant mouse strains. METHODS: In this study, we used DEREG mice that express the diphtheria toxin (DT) receptor under control of the foxp3 locus to selectively deplete Foxp3(+) Tregs by injection of DT prior to infection and studied the effect of Treg depletion on the course of acute Theiler's murine encephalomyelitis (TME). RESULTS: As expected, DEREG mice that are on a C57BL/6 background were resistant to TMEV infection and cleared the virus within days of infection, regardless of the presence or absence of Tregs. Nevertheless, in the absence of Tregs we observed priming of stronger effector T cell responses in the periphery, which subsequently resulted in a transient increase in the frequency of IFNγ-producing T cells in the brain at an early stage of infection. Histological and flow cytometric analysis revealed that this transiently increased frequency of brain-infiltrating IFNγ-producing T cells in Treg-depleted mice neither led to an augmented antiviral response nor enhanced inflammation-mediated tissue damage. Intriguingly, Treg depletion did not change the expression of IL-10 in the infected brain, which might play a role for dampening the inflammatory damage caused by the increased number of effector T cells. CONCLUSION: We therefore propose that unlike susceptible mice strains, interfering with the Treg compartment of resistant mice only has negligible effects on virus-induced pathologies in the CNS. Furthermore, in the absence of Tregs, local anti-inflammatory mechanisms might limit the extent of damage caused by strong anti-viral response in the CNS.


Subject(s)
Cardiovirus Infections/immunology , Disease Resistance/immunology , Encephalitis, Viral/immunology , T-Lymphocytes, Regulatory/physiology , Theilovirus/immunology , Acute Disease , Animals , Cardiovirus Infections/prevention & control , Encephalitis, Viral/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Med J Aust ; 201(11): 663-6, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25495312

ABSTRACT

OBJECTIVE: To report the findings of the enhanced surveillance set up in New South Wales in response to the recent outbreak of human enterovirus 71 (EV71) infection. DESIGN AND SETTING: A two-armed enhanced public health surveillance system including statewide emergency department surveillance and clinical surveillance at the Sydney Children's Hospitals Network. PARTICIPANTS: Children aged less than 10 years with suspected or confirmed enterovirus infection. MAIN OUTCOME MEASURES: Epidemiology of the outbreak, including weekly case counts, demographic information, geographic spread of the outbreak, and clinical presentation and progression. RESULTS: Statewide weekly case counts indicate that an epidemic of EV71 infection occurred in NSW from December 2012 until May 2013. Around 119 children were reported with disease severe enough to warrant admission to a tertiary Sydney children's hospital. Cases were spread throughout the Sydney metropolitan area and there is some evidence of geographic migration of the outbreak. Presenting features included fever, lethargy, myoclonus and skin rash. Only 24% of cases presented with classical hand, foot and mouth disease. CONCLUSIONS: EV71 infection is likely to continue to be a public health problem in Australia. Surveillance of routinely collected emergency department data can provide a useful indication of its activity in the community.


Subject(s)
Disease Outbreaks/statistics & numerical data , Encephalitis, Viral/epidemiology , Enterovirus A, Human , Enterovirus Infections/epidemiology , Child , Child, Preschool , Cities/epidemiology , Disease Outbreaks/prevention & control , Emergency Service, Hospital/statistics & numerical data , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Enterovirus Infections/prevention & control , Enterovirus Infections/virology , Female , Humans , Infant , Infant, Newborn , Male , New South Wales/epidemiology , Population Surveillance
11.
J Infect Dis ; 207(1): 142-51, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23089589

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are closely related, recently emerged paramyxoviruses that form Henipavirus genus and are capable of causing considerable morbidity and mortality in a number of mammalian species, including humans. However, in contrast to many other species and despite expression of functional virus entry receptors, mice are resistant to henipavirus infection. We report here the susceptibility of mice deleted for the type I interferon receptor (IFNAR-KO) to both HeV and NiV. Intraperitoneally infected mice developed fatal encephalitis, with pathology and immunohistochemical features similar to what was found in humans. Viral RNA was found in the majority of analyzed organs, and sublethally infected animals developed virus-specific neutralizing antibodies. Altogether, these results reveal IFNAR-KO mice as a new small animal model to study HeV and NiV pathogenesis, prophylaxis, and treatment and suggest the critical role of type I interferon signaling in the control of henipavirus infection.


Subject(s)
Antibodies, Viral/immunology , Encephalitis, Viral/prevention & control , Henipavirus Infections/prevention & control , Henipavirus/immunology , Interferon Type I/genetics , Animals , Antibodies, Neutralizing , Antibody Specificity , Brain/virology , Cells, Cultured , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/mortality , Encephalitis, Viral/virology , Hendra Virus/genetics , Hendra Virus/immunology , Hendra Virus/pathogenicity , Henipavirus/genetics , Henipavirus/pathogenicity , Henipavirus Infections/immunology , Henipavirus Infections/mortality , Henipavirus Infections/virology , Humans , Interferon Type I/immunology , Mice , Mice, Knockout , Neuroglia/virology , Nipah Virus/genetics , Nipah Virus/immunology , Nipah Virus/pathogenicity , RNA, Viral/analysis , Signal Transduction , Survival Analysis , Virulence , Virus Internalization , Virus Replication
12.
Bone Marrow Transplant ; 59(9): 1224-1231, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38796633

ABSTRACT

The impact of letermovir (LTV)-an anti-cytomegalovirus (CMV) drug-on human herpesvirus-6 (HHV-6) encephalitis is unclear. We hypothesized that LTV prophylaxis may increase the incidence of HHV-6 encephalitis by reducing anti-CMV therapies after allogeneic hematopoietic stem cell transplantation (HSCT). To evaluate the association between HHV-6 encephalitis and antiviral prophylaxis, 7985 adult patients from a nationwide registry who underwent their first HSCT between January 2019 and December 2021 were analyzed. The incidence of HHV-6 encephalitis on day 100 after HSCT was 3.6%; 11.5% for the broad-spectrum antiviral group (foscarnet, ganciclovir, or valganciclovir); 2.8% for the LTV group, and 3.8% for the other antiviral group (p < 0.001). These differences persisted when cord blood transplantation (CBT) was analyzed separately (14.1%, 5.9%, and 7.4%, p < 0.001). In the multivariate analysis, CBT (hazard ratio [HR]: 2.90), broad-spectrum antiviral prophylaxis (HR: 1.91), and grade II-IV acute graft-versus-host disease requiring systemic corticosteroids (HR: 2.42) were independent risk factors for encephalitis (all p < 0.001). The findings of this large modern database study indicate that broad-spectrum antiviral prophylaxis, rather than LTV prophylaxis, is paradoxically associated with HHV-6 encephalitis in the LTV era. This paradoxical finding needs to be further explored in future studies.


Subject(s)
Antiviral Agents , Encephalitis, Viral , Hematopoietic Stem Cell Transplantation , Herpesvirus 6, Human , Roseolovirus Infections , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Antiviral Agents/therapeutic use , Male , Encephalitis, Viral/prevention & control , Encephalitis, Viral/etiology , Adult , Female , Roseolovirus Infections/prevention & control , Roseolovirus Infections/etiology , Middle Aged , Quinazolines/therapeutic use , Triazoles/therapeutic use , Transplantation, Homologous , Acetates
14.
Arch Virol ; 157(8): 1423-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22592957

ABSTRACT

Members of the genus Flavivirus are responsible for a spectrum of important neurological syndromes in humans and animals. Rodent models have been used extensively to model flavivirus neurological disease, to discover host-pathogen interactions that influence disease outcome, and as surrogates to determine the efficacy and safety of vaccines and therapeutics. In this review, we discuss the current understanding of flavivirus neuroinvasive disease and outline the host, viral and experimental factors that influence the outcome and reliability of virus infection of small-animal models.


Subject(s)
Central Nervous System/virology , Disease Models, Animal , Encephalitis, Viral , Flavivirus Infections , Flavivirus/pathogenicity , Rodentia , Animals , Cricetinae , Encephalitis, Viral/drug therapy , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Flavivirus Infections/drug therapy , Flavivirus Infections/prevention & control , Flavivirus Infections/virology , Host-Pathogen Interactions , Insect Vectors/virology , Mice
15.
Transpl Infect Dis ; 14(1): 33-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21794043

ABSTRACT

Human herpesvirus-6 (HHV-6) is a major cause of limbic encephalitis with a dismal prognosis after allogeneic hematopoietic stem cell transplantation (SCT). Because our previous trial of preemptive therapy with foscarnet sodium (phosphonoformic acid; PFA) failed to prevent HHV-6 encephalitis, we conducted a prospective study to examine the safety of prophylactic PFA administration and elucidate the changes in the plasma HHV-6 DNA levels in the early post-SCT period. Plasma HHV-6 DNA was measured thrice weekly from day 6. PFA, 90 mg/kg/day, was administered from days 7 to 21 after bone marrow or peripheral blood SCT and to day 25 after umbilical cord blood transplantation. Of the 10 patients enrolled, 2 dropped out of the study, 1 because of early death, and 1 with a low glomerular filtration rate. Grade 3 or greater adverse events occurred in 9 of the 10 prophylactic PFA patients and in 7 of the 10 control patients who had clinical backgrounds similar to the study subjects and underwent SCT during the same period. Neurological disorders developed in none of the study subjects but in 4 of the 10 control patients, including 2 with HHV-6 encephalitis. HHV-6 reactivation occurred in 3 of the 10 study subjects. The prophylactic PFA regimen was thus safe and it may reduce the risk of limbic encephalitis, but is not considered to be potent enough to prevent HHV-6 reactivation.


Subject(s)
Antiviral Agents/adverse effects , Encephalitis, Viral/prevention & control , Foscarnet/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Herpesvirus 6, Human/drug effects , Adolescent , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , DNA, Viral/blood , Encephalitis, Viral/epidemiology , Encephalitis, Viral/virology , Female , Foscarnet/administration & dosage , Foscarnet/therapeutic use , Humans , Incidence , Male , Middle Aged , Peripheral Blood Stem Cell Transplantation/adverse effects , Roseolovirus Infections/epidemiology , Roseolovirus Infections/prevention & control , Roseolovirus Infections/virology , Transplantation, Homologous , Treatment Outcome , Viremia/epidemiology , Viremia/prevention & control , Viremia/virology , Young Adult
16.
Sci Immunol ; 7(67): eabl9929, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-34812647

ABSTRACT

The development of a tractable small animal model faithfully reproducing human coronavirus disease 2019 pathogenesis would arguably meet a pressing need in biomedical research. Thus far, most investigators have used transgenic mice expressing the human ACE2 in epithelial cells (K18-hACE2 transgenic mice) that are intranasally instilled with a liquid severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) suspension under deep anesthesia. Unfortunately, this experimental approach results in disproportionate high central nervous system infection leading to fatal encephalitis, which is rarely observed in humans and severely limits this model's usefulness. Here, we describe the use of an inhalation tower system that allows exposure of unanesthetized mice to aerosolized virus under controlled conditions. Aerosol exposure of K18-hACE2 transgenic mice to SARS-CoV-2 resulted in robust viral replication in the respiratory tract, anosmia, and airway obstruction but did not lead to fatal viral neuroinvasion. When compared with intranasal inoculation, aerosol infection resulted in a more pronounced lung pathology including increased immune infiltration, fibrin deposition, and a transcriptional signature comparable to that observed in SARS-CoV-2­infected patients. This model may prove useful for studies of viral transmission, disease pathogenesis (including long-term consequences of SARS-CoV-2 infection), and therapeutic interventions.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/physiopathology , Disease Models, Animal , Encephalitis, Viral/prevention & control , Keratin-18/genetics , Nasal Sprays , SARS-CoV-2/physiology , Administration, Inhalation , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Encephalitis, Viral/mortality , Epithelial Cells/metabolism , Female , Humans , Keratin-18/metabolism , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Transcriptome , Virus Replication
17.
J Immunol ; 182(6): 3855-65, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265165

ABSTRACT

HIV-1-associated neurocognitive impairments are intrinsically linked to microglial immune activation, persistent viral infection, and inflammation. In the era of antiretroviral therapy, more subtle cognitive impairments occur without adaptive immune compromise. We posit that adaptive immunity is neuroprotective, serving in both the elimination of infected cells through CD8(+) cytotoxic T cell activities and the regulation of neuroinflammatory responses of activated microglia. For the latter, little is known. Thus, we studied the neuromodulatory effects of CD4(+) regulatory T cells (Treg; CD4(+)CD25(+)) or effector T cells in HIV-1-associated neurodegeneration. A newly developed HIV-1 encephalitis mouse model was used wherein murine bone marrow-derived macrophages are infected with a full-length HIV-1(YU2)/vesicular stomatitis viral pseudotype and injected into basal ganglia of syngeneic immunocompetent mice. Adoptive transfer of CD3-activated Treg attenuated astrogliosis and microglia inflammation with concomitant neuroprotection. Moreover, Treg-mediated anti-inflammatory activities and neuroprotection were associated with up-regulation of brain-derived neurotrophic factor and glial cell-derived neurotrophic factor expression and down-regulation of proinflammatory cytokines, oxidative stress, and viral replication. Effector T cells showed contrary effects. These results, taken together, demonstrate the importance of Treg in disease control and raise the possibility of their utility for therapeutic strategies.


Subject(s)
AIDS Dementia Complex/prevention & control , Encephalitis, Viral/prevention & control , HIV Infections/prevention & control , HIV-1/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Neuroimmunomodulation/immunology , T-Lymphocytes, Regulatory/immunology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/pathology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Cells/virology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/pathology , HIV Infections/immunology , HIV Infections/pathology , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Microglia/virology , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/physiology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology
18.
Res Vet Sci ; 136: 185-191, 2021 May.
Article in English | MEDLINE | ID: mdl-33677208

ABSTRACT

The Bovine herpes virus type 5 glycoprotein D (gD) is essential for viral penetration into host permissive cells. The Herpes virus gD glycoprotein has been used for bovine immunization, being efficient in reduction of viral replication, shedding and clinical signs, however sterilizing immunity is still not achieved. Recombinant subunit vaccines are, in general, poorly immunogenic requiring additional adjuvant components. Interleukin 17A (IL17A) is a pro-inflammatory cytokine produced by T helper 17 cells that mediate mucosal immunity. IL17 production during vaccine-induced immunity is a requirement for mucosal protection to several agents. In this study, we investigated the potential of a recombinant IL17A to act as an adjuvant for a recombinant BoHV-5 glycoprotein D vaccine in cattle. Three cattle groups were divided as: group 1) rgD5 + alumen + rIL-17A; 2) rgD5 + alumen; and 3) PBS + alumen. The cattle (3 per group) received two doses of their respective vaccines at an interval of 21 days. The group that received rIL17 in its vaccine formulation at the 7th day after the prime immunization had significant higher levels of specific rgD-IgG than the alumen group. Addition of rIL17 also led to a significant fold increase in specific anti-rgD IgG and neutralizing antibodies to the virus, respectively, when compared with the alumen group. Cells stimulated with rIL17A responded with IL17 transcription, as well IL2, IL4, IL10, IL15, Bcl6 and CXCR5. Our findings suggest that the rIL17A has adjuvant potential for use in vaccines against BoHV-5 as well as potentially other pathogens of cattle.


Subject(s)
Antibodies, Viral/immunology , Cattle Diseases/prevention & control , Encephalitis, Viral/veterinary , Herpesviridae Infections/veterinary , Herpesvirus 5, Bovine/immunology , Herpesvirus Vaccines/immunology , Meningoencephalitis/veterinary , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/immunology , Cattle , Encephalitis, Viral/prevention & control , Herpesviridae Infections/prevention & control , Herpesvirus 5, Bovine/genetics , Immunization/veterinary , Interleukin-17/genetics , Interleukin-17/immunology , Meningoencephalitis/prevention & control , Vaccines, Synthetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
19.
Ocul Immunol Inflamm ; 29(6): 1225-1233, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34648417

ABSTRACT

The novel pandemic coronavirus disease 2019 (COVID-19) leading to health and economic problems worldwide is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 mainly occurs as a lower respiratory tract infection, there is multiorgan involvement in infected patients. The disease is transmitted from person to person through air droplets or contact with contaminated surfaces. SARS-CoV-2 leads to this systemic involvement by attaching to angiotensin-converting enzyme 2 (ACE2) receptors located on several human cells. Since SARS-CoV-2 RNA has been found in tears of infected patients, ocular surface may allow the virus to transmit to nasopharynx via the nasolacrimal duct. This narrative review aims to sum up all segmental ocular complications, ocular adverse effects of COVID-19 treatment, and preventive measures suggested to minimize the SARS-CoV-2 transmission between patients and ophthalmologists by reviewing currently available literature.


Subject(s)
COVID-19/diagnosis , Eye Infections, Viral/diagnosis , SARS-CoV-2 , Tears/virology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Nucleic Acid Testing , Conjunctivitis, Viral/diagnosis , Conjunctivitis, Viral/prevention & control , Conjunctivitis, Viral/virology , Encephalitis, Viral/diagnosis , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Eye Infections, Viral/prevention & control , Eye Infections, Viral/virology , Humans , Preventive Medicine/methods , Retinal Diseases/diagnosis , Retinal Diseases/prevention & control , Retinal Diseases/virology , SARS-CoV-2/pathogenicity
20.
Front Immunol ; 12: 667478, 2021.
Article in English | MEDLINE | ID: mdl-34025669

ABSTRACT

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antigen-Presenting Cells/drug effects , Brain/drug effects , Encephalitis, Viral/prevention & control , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Macrophages/drug effects , Rhabdoviridae Infections/prevention & control , Vesiculovirus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Brain/immunology , Brain/metabolism , Brain/virology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RAW 264.7 Cells , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Ubiquitination , Vero Cells , Vesiculovirus/pathogenicity , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL