Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(3): e2218899120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36638211

ABSTRACT

Cleavage of the flavivirus premembrane (prM) structural protein during maturation can be inefficient. The contribution of partially mature flavivirus virions that retain uncleaved prM to pathogenesis during primary infection is unknown. To investigate this question, we characterized the functional properties of newly-generated dengue virus (DENV) prM-reactive monoclonal antibodies (mAbs) in vitro and using a mouse model of DENV disease. Anti-prM mAbs neutralized DENV infection in a virion maturation state-dependent manner. Alanine scanning mutagenesis and cryoelectron microscopy of anti-prM mAbs in complex with immature DENV defined two modes of attachment to a single antigenic site. In vivo, passive transfer of intact anti-prM mAbs resulted in an antibody-dependent enhancement of disease. However, protection against DENV-induced lethality was observed when the transferred mAbs were genetically modified to inhibit their ability to interact with Fcγ receptors. These data establish that in addition to mature forms of the virus, partially mature infectious prM+ virions can also contribute to pathogenesis during primary DENV infections.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Dengue Virus , Dengue , Cryoelectron Microscopy , Viral Envelope Proteins/metabolism , Virion/metabolism , Animals , Mice
2.
JCI Insight ; 6(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33232299

ABSTRACT

Glioblastoma multiforme (GBM) is a fatal human cancer in part because GBM stem cells are resistant to therapy and recurrence is inevitable. Previously, we demonstrated Zika virus (ZIKV) targets GBM stem cells and prevents death of mice with gliomas. Here, we evaluated the immunological basis of ZIKV-mediated protection against GBM. Introduction of ZIKV into the brain tumor increased recruitment of CD8+ T and myeloid cells to the tumor microenvironment. CD8+ T cells were required for ZIKV-dependent tumor clearance because survival benefits were lost with CD8+ T cell depletion. Moreover, while anti-PD-1 antibody monotherapy moderately improved tumor survival, when coadministered with ZIKV, survival increased. ZIKV-mediated tumor clearance also resulted in durable protection against syngeneic tumor rechallenge, which also depended on CD8+ T cells. To address safety concerns, we generated an immune-sensitized ZIKV strain, which was effective alone or in combination with immunotherapy. Thus, oncolytic ZIKV treatment can be leveraged by immunotherapies, which may prompt combination treatment paradigms for adult patients with GBM.


Subject(s)
Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Glioblastoma/therapy , Immune Checkpoint Inhibitors/administration & dosage , Oncolytic Virotherapy/methods , Oncolytic Viruses/immunology , Zika Virus/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Combined Modality Therapy , Female , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Tumor Microenvironment/immunology
3.
Immunity ; 53(5): 1078-1094.e7, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33010224

ABSTRACT

Memory B cells (MBCs) can respond to heterologous antigens either by molding new specificities through secondary germinal centers (GCs) or by selecting preexisting clones without further affinity maturation. To distinguish these mechanisms in flavivirus infections and immunizations, we studied recall responses to envelope protein domain III (DIII). Conditional deletion of activation-induced cytidine deaminase (AID) between heterologous challenges of West Nile, Japanese encephalitis, Zika, and dengue viruses did not affect recall responses. DIII-specific MBCs were contained mostly within the plasma-cell-biased CD80+ subset, and few GCs arose following heterologous boosters, demonstrating that recall responses are confined by preexisting clonal diversity. Measurement of monoclonal antibody (mAb) binding affinity to DIII proteins, timed AID deletion, single-cell RNA sequencing, and lineage tracing experiments point to selection of relatively low-affinity MBCs as a mechanism to promote diversity. Engineering immunogens to avoid this MBC diversity may facilitate flavivirus-type-specific vaccines with minimized potential for infection enhancement.


Subject(s)
B-Lymphocytes/immunology , Cross Reactions/immunology , Flavivirus Infections/immunology , Flavivirus Infections/virology , Flavivirus/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , Animals , B-Lymphocytes/metabolism , Disease Models, Animal , Dose-Response Relationship, Immunologic , Flavivirus Infections/metabolism , Immunization , Mice , Mice, Knockout , Mice, Transgenic , Plasma Cells/immunology , Plasma Cells/metabolism , Species Specificity
4.
Mol Ther Methods Clin Dev ; 18: 402-414, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32695842

ABSTRACT

Monoclonal antibody (mAb) therapeutics are an effective modality for the treatment of infectious, autoimmune, and cancer-related diseases. However, the discovery, development, and manufacturing processes are complex, resource-consuming activities that preclude the rapid deployment of mAbs in outbreaks of emerging infectious diseases. Given recent advances in nucleic acid delivery technology, it is now possible to deliver exogenous mRNA encoding mAbs for in situ expression following intravenous (i.v.) infusion of lipid nanoparticle-encapsulated mRNA. However, the requirement for i.v. administration limits the application to settings where infusion is an option, increasing the cost of treatment. As an alternative strategy, and to enable intramuscular (IM) administration of mRNA-encoded mAbs, we describe a nanostructured lipid carrier for delivery of an alphavirus replicon encoding a previously described highly neutralizing human mAb, ZIKV-117. Using a lethal Zika virus challenge model in mice, our studies show robust protection following alphavirus-driven expression of ZIKV-117 mRNA when given by IM administration as pre-exposure prophylaxis or post-exposure therapy.

5.
J Infect Dis ; 220(12): 1904-1914, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31063544

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has become a global concern because infection of pregnant mothers was linked to congenital birth defects. Zika virus is unique from other flaviviruses, because it is transmitted vertically and sexually in addition to by mosquito vectors. Prior studies in mice, nonhuman primates, and humans have shown that ZIKV targets the testis in males, resulting in persistent infection and oligospermia. However, its effects on the corresponding female gonads have not been evaluated. METHODS: In this study, we assessed the effects of ZIKV on the ovary in nonpregnant mice. RESULTS: During the acute phase, ZIKV productively infected the ovary causing accumulation of CD4+ and virus-specific CD8+ T cells. T cells protected against ZIKV infection in the ovary, as higher viral burden was measured in CD8-/- and TCRßδ-/- mice. Increased cell death and tissue inflammation in the ovary was observed during the acute phase of infection, but this normalized over time. CONCLUSIONS: In contrast to that observed with males, minimal persistence and no long-term consequences of ZIKV infection on ovarian follicular reserve or fertility were demonstrated in this model. Thus, although ZIKV replicates in cells of the ovary and causes acute oophoritis, there is rapid resolution and no long-term effects on fertility, at least in mice.


Subject(s)
Fertility , Oophoritis/physiopathology , Oophoritis/virology , Zika Virus Infection/physiopathology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Biomarkers , Disease Models, Animal , Female , Infertility, Female/etiology , Mice , Mice, Knockout , Oophoritis/complications , Oophoritis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viral Load , Viral Tropism , Zika Virus Infection/complications , Zika Virus Infection/pathology
6.
Cell Host Microbe ; 24(5): 743-750.e5, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30439343

ABSTRACT

Antibody (Ab)-dependent enhancement can exacerbate dengue virus (DENV) infection due to cross-reactive Abs from an initial DENV infection, facilitating replication of a second DENV. Zika virus (ZIKV) emerged in DENV-endemic areas, raising questions about whether existing immunity could affect these related flaviviruses. We show that mice born with circulating maternal Abs against ZIKV develop severe disease upon DENV infection. Compared with pups of naive mothers, those born to ZIKV-immune mice lacking type I interferon receptor in myeloid cells (LysMCre+Ifnar1fl/fl) exhibit heightened disease and viremia upon DENV infection. Passive transfer of IgG isolated from mice born to ZIKV-immune mothers resulted in increased viremia in naive recipient mice. Treatment with Abs blocking inflammatory cytokine tumor necrosis factor linked to DENV disease or Abs blocking DENV entry improved survival of DENV-infected mice born to ZIKV-immune mothers. Thus, the maternal Ab response to ZIKV infection or vaccination might predispose to severe dengue disease in infants.


Subject(s)
Antibodies, Viral/immunology , Antibody-Dependent Enhancement/immunology , Dengue Virus/immunology , Dengue/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibody Formation , Cell Line , Cross Reactions/immunology , Culicidae , Cytokines/metabolism , Dengue Virus/pathogenicity , Disease Models, Animal , Female , Humans , Immunity , Immunoglobulin G , Male , Mice , Mice, Inbred C57BL , Myeloid Cells , Receptor, Interferon alpha-beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Viremia , Virus Internalization , Zika Virus/pathogenicity , Zika Virus Infection/virology
7.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29976679

ABSTRACT

Dengue virus is the most globally prevalent mosquito-transmitted virus. Primary infection with one of four cocirculating serotypes (DENV-1 to -4) causes a febrile illness, but secondary infection with a heterologous serotype can result in severe disease, due in part to antibody-dependent enhancement of infection (ADE). In ADE, cross-reactive but nonneutralizing antibodies, or subprotective levels of neutralizing antibodies, promote uptake of antibody-opsonized virus in Fc-γ receptor-positive cells. Thus, elicitation of broadly neutralizing antibodies (bNAbs), but not nonneutralizing antibodies, is desirable for dengue vaccine development. Domain III of the envelope glycoprotein (EDIII) is targeted by bNAbs and thus is an attractive immunogen. However, immunization with EDIII results in sera with limited neutralization breadth. We developed "resurfaced" EDIII immunogens (rsDIIIs) in which the A/G strand epitope that is targeted by bNAb 4E11 is maintained but less desirable epitopes are masked. RsDIIIs bound 4E11, but not serotype-specific or nonneutralizing antibodies. One rsDIII and, unexpectedly, wild-type (WT) DENV-2 EDIII elicited cross-neutralizing antibody responses against DENV-1 to -3 in mice. While these sera were cross-neutralizing, they were not sufficiently potent to protect AG129 immunocompromised mice at a dose of 200 µl (50% focus reduction neutralization titer [FRNT50], ∼1:60 to 1:130) against mouse-adapted DENV-2. Our results provide insight into immunogen design strategies based on EDIII.IMPORTANCE Dengue virus causes approximately 390 million infections per year. Primary infection by one serotype causes a self-limiting febrile illness, but secondary infection by a heterologous serotype can result in severe dengue syndrome, which is characterized by hemorrhagic fever and shock syndrome. This severe disease is thought to arise because of cross-reactive, non- or poorly neutralizing antibodies from the primary infection that are present in serum at the time of secondary infection. These cross-reactive antibodies enhance the infection rather than controlling it. Therefore, induction of a broadly and potently neutralizing antibody response is desirable for dengue vaccine development. Here, we explore a novel strategy for developing immunogens based on domain III of the E glycoprotein, where undesirable epitopes (nonneutralizing or nonconserved) are masked by mutation. This work provides fundamental insight into the immune response to domain III that can be leveraged for future immunogen design.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/immunology , Dengue Virus/genetics , Protein Domains/genetics , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/administration & dosage , Antibodies, Viral/adverse effects , Antibody-Dependent Enhancement , Cell Surface Display Techniques , Cross Reactions , Dengue/virology , Dengue Vaccines/immunology , Dengue Virus/chemistry , Dengue Virus/immunology , Epitopes/immunology , Mice , Protein Domains/immunology , Protein Engineering/methods , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Proteins/chemistry , Viral Proteins/genetics
8.
mBio ; 9(1)2018 02 27.
Article in English | MEDLINE | ID: mdl-29487230

ABSTRACT

Japanese encephalitis virus (JEV) remains a leading cause of viral encephalitis worldwide. Although JEV-specific antibodies have been described, an assessment of their ability to neutralize multiple genotypes of JEV has been limited. Here, we describe the development of a panel of mouse and human neutralizing monoclonal antibodies (MAbs) that inhibit infection in cell culture of four different JEV genotypes tested. Mechanism-of-action studies showed that many of these MAbs inhibited infection at a postattachment step, including blockade of virus fusion. Mapping studies using site-directed mutagenesis and hydrogen-deuterium exchange with mass spectrometry revealed that the lateral ridge on domain III of the envelope protein was a primary recognition epitope for our panel of strongly neutralizing MAbs. Therapeutic studies in mice demonstrated protection against lethality caused by genotype I and III strains when MAbs were administered as a single dose even 5 days after infection. This information may inform the development of vaccines and therapeutic antibodies as emerging strains and genotypic shifts become more prevalent.IMPORTANCE Although Japanese encephalitis virus (JEV) is a vaccine-preventable cause of viral encephalitis, the inactivated and live attenuated platforms available are derived from strains belonging to a single genotype (GIII) due to its historical prevalence in areas of JEV epidemics. Related to this, studies with vaccines and antibodies have focused on assessing the in vitro and in vivo protective responses to homologous or heterologous GIII strains. An epidemiological shift in JEV genotype distribution warrants the induction of broadly neutralizing antibody responses that inhibit infection of multiple JEV genotypes. Here, we generated a panel of mouse and human neutralizing monoclonal antibodies and evaluated their inhibitory activity, epitope location, and capacity for protection against multiple JEV genotypes in mice.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/prevention & control , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis Virus, Japanese/classification , Encephalitis Virus, Japanese/genetics , Epitopes/immunology , Genotype , Humans , Mice , Models, Biological , Treatment Outcome , Vero Cells , Viral Envelope Proteins/immunology
9.
J Clin Invest ; 127(4): 1338-1352, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28240606

ABSTRACT

Global health is threatened by emerging viral infections, which largely lack effective vaccines or therapies. Targeting host pathways that are exploited by multiple viruses could offer broad-spectrum solutions. We previously reported that AAK1 and GAK, kinase regulators of the host adaptor proteins AP1 and AP2, are essential for hepatitis C virus (HCV) infection, but the underlying mechanism and relevance to other viruses or in vivo infections remained unknown. Here, we have discovered that AP1 and AP2 cotraffic with HCV particles in live cells. Moreover, we found that multiple viruses, including dengue and Ebola, exploit AAK1 and GAK during entry and infectious virus production. In cultured cells, treatment with sunitinib and erlotinib, approved anticancer drugs that inhibit AAK1 or GAK activity, or with more selective compounds inhibited intracellular trafficking of HCV and multiple unrelated RNA viruses with a high barrier to resistance. In murine models of dengue and Ebola infection, sunitinib/erlotinib combination protected against morbidity and mortality. We validated sunitinib- and erlotinib-mediated inhibition of AAK1 and GAK activity as an important mechanism of antiviral action. Additionally, we revealed potential roles for additional kinase targets. These findings advance our understanding of virus-host interactions and establish a proof of principle for a repurposed, host-targeted approach to combat emerging viruses.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Erlotinib Hydrochloride/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cell Line, Tumor , Dengue/prevention & control , Dengue/virology , Dengue Virus/drug effects , Dengue Virus/metabolism , Drug Evaluation, Preclinical , Drug Synergism , Ebolavirus/drug effects , Ebolavirus/metabolism , Female , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Hepacivirus/drug effects , Hepacivirus/metabolism , Hepatitis C/prevention & control , Hepatitis C/virology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Sunitinib , Virus Internalization/drug effects
10.
Antiviral Res ; 137: 134-140, 2017 01.
Article in English | MEDLINE | ID: mdl-27902933

ABSTRACT

The rapidly expanding Zika virus (ZIKV) epidemic has affected thousands of individuals with severe cases causing Guillain-Barré syndrome, congenital malformations, and microcephaly. Currently, there is no available vaccine or therapy to prevent or treat ZIKV infection. We evaluated whether sofosbuvir, an FDA-approved nucleotide polymerase inhibitor for the distantly related hepatitis C virus, could have antiviral activity against ZIKV infection. Cell culture studies established that sofosbuvir efficiently inhibits replication and infection of several ZIKV strains in multiple human tumor cell lines and isolated human fetal-derived neuronal stem cells. Moreover, oral treatment with sofosbuvir protected against ZIKV-induced death in mice. These results suggest that sofosbuvir may be a candidate for further evaluation as a therapy against ZIKV infection in humans.


Subject(s)
Antiviral Agents/pharmacology , Sofosbuvir/pharmacology , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Administration, Oral , Animals , Antiviral Agents/therapeutic use , Cell Line , Drug Approval , Drug Evaluation, Preclinical , Humans , Mice , Sofosbuvir/administration & dosage , Sofosbuvir/therapeutic use , United States , United States Food and Drug Administration , Zika Virus Infection/virology
11.
Nature ; 540(7633): 438-442, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798603

ABSTRACT

Infection of pregnant women with Zika virus (ZIKV) can cause congenital malformations including microcephaly, which has focused global attention on this emerging pathogen. In addition to transmission by mosquitoes, ZIKV can be detected in the seminal fluid of affected males for extended periods of time and transmitted sexually. Here, using a mouse-adapted African ZIKV strain (Dakar 41519), we evaluated the consequences of infection in the male reproductive tract of mice. We observed persistence of ZIKV, but not the closely related dengue virus (DENV), in the testis and epididymis of male mice, and this was associated with tissue injury that caused diminished testosterone and inhibin B levels and oligospermia. ZIKV preferentially infected spermatogonia, primary spermatocytes and Sertoli cells in the testis, resulting in cell death and destruction of the seminiferous tubules. Less damage was caused by a contemporary Asian ZIKV strain (H/PF/2013), in part because this virus replicates less efficiently in mice. The extent to which these observations in mice translate to humans remains unclear, but longitudinal studies of sperm function and viability in ZIKV-infected humans seem warranted.


Subject(s)
Testis/pathology , Testis/virology , Zika Virus Infection/pathology , Zika Virus/pathogenicity , Animals , Cell Death , Dengue Virus/physiology , Epididymis/pathology , Epididymis/virology , Humans , Inhibins/metabolism , Male , Mice , Mice, Inbred C57BL , Oligospermia/pathology , Oligospermia/virology , Seminiferous Tubules/pathology , Seminiferous Tubules/virology , Sertoli Cells/virology , Spermatocytes/virology , Spermatogonia/virology , Testosterone/metabolism , Time Factors
12.
Cell Rep ; 16(12): 3208-3218, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27612415

ABSTRACT

Zika virus (ZIKV) is an emerging flavivirus that causes congenital abnormalities and Guillain-Barré syndrome. ZIKV infection also results in severe eye disease characterized by optic neuritis, chorioretinal atrophy, and blindness in newborns and conjunctivitis and uveitis in adults. We evaluated ZIKV infection of the eye by using recently developed mouse models of pathogenesis. ZIKV-inoculated mice developed conjunctivitis, panuveitis, and infection of the cornea, iris, optic nerve, and ganglion and bipolar cells in the retina. This phenotype was independent of the entry receptors Axl or Mertk, given that Axl(-/-), Mertk(-/-), and Axl(-/-)Mertk(-/-) double knockout mice sustained levels of infection similar to those of control animals. We also detected abundant viral RNA in tears, suggesting that virus might be secreted from lacrimal glands or shed from the cornea. This model provides a foundation for studying ZIKV-induced ocular disease, defining mechanisms of viral persistence, and developing therapeutic approaches for viral infections of the eye.


Subject(s)
Panuveitis/virology , Tears/virology , Virus Shedding/physiology , Zika Virus Infection/virology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , c-Mer Tyrosine Kinase/deficiency , Axl Receptor Tyrosine Kinase
13.
Cell ; 166(4): 1016-1027, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27475895

ABSTRACT

Zika virus (ZIKV) infection during pregnancy has emerged as a global public health problem because of its ability to cause severe congenital disease. Here, we developed six mouse monoclonal antibodies (mAbs) against ZIKV including four (ZV-48, ZV-54, ZV-64, and ZV-67) that were ZIKV specific and neutralized infection of African, Asian, and American strains to varying degrees. X-ray crystallographic and competition binding analyses of Fab fragments and scFvs defined three spatially distinct epitopes in DIII of the envelope protein corresponding to the lateral ridge (ZV-54 and ZV-67), C-C' loop (ZV-48 and ZV-64), and ABDE sheet (ZV-2) regions. In vivo passive transfer studies revealed protective activity of DIII-lateral ridge specific neutralizing mAbs in a mouse model of ZIKV infection. Our results suggest that DIII is targeted by multiple type-specific antibodies with distinct neutralizing activity, which provides a path for developing prophylactic antibodies for use in pregnancy or designing epitope-specific vaccines against ZIKV.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , Viral Envelope Proteins/chemistry , Zika Virus/chemistry , Zika Virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Epitope Mapping , Epitopes , Mice , Mice, Inbred C57BL , Models, Molecular , Zika Virus/classification , Zika Virus Infection/immunology , Zika Virus Infection/virology
14.
Cell ; 165(5): 1081-1091, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27180225

ABSTRACT

Zika virus (ZIKV) infection in pregnant women causes intrauterine growth restriction, spontaneous abortion, and microcephaly. Here, we describe two mouse models of placental and fetal disease associated with in utero transmission of ZIKV. Female mice lacking type I interferon signaling (Ifnar1(-/-)) crossed to wild-type (WT) males produced heterozygous fetuses resembling the immune status of human fetuses. Maternal inoculation at embryonic day 6.5 (E6.5) or E7.5 resulted in fetal demise that was associated with ZIKV infection of the placenta and fetal brain. We identified ZIKV within trophoblasts of the maternal and fetal placenta, consistent with a trans-placental infection route. Antibody blockade of Ifnar1 signaling in WT pregnant mice enhanced ZIKV trans-placental infection although it did not result in fetal death. These models will facilitate the study of ZIKV pathogenesis, in utero transmission, and testing of therapies and vaccines to prevent congenital malformations.


Subject(s)
Disease Models, Animal , Fetal Diseases/virology , Placenta Diseases/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/pathology , Zika Virus/physiology , Animals , Apoptosis , Brain/embryology , Brain/pathology , Brain/virology , Female , Fetal Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Placenta Diseases/pathology , Pregnancy , Pregnancy Complications, Infectious/pathology , RNA, Viral/isolation & purification , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Zika Virus Infection/virology
15.
Cell Host Microbe ; 19(5): 720-30, 2016 May 11.
Article in English | MEDLINE | ID: mdl-27066744

ABSTRACT

The ongoing Zika virus (ZIKV) epidemic and unexpected clinical outcomes, including Guillain-Barré syndrome and birth defects, has brought an urgent need for animal models. We evaluated infection and pathogenesis with contemporary and historical ZIKV strains in immunocompetent mice and mice lacking components of the antiviral response. Four- to six-week-old Irf3(-/-)Irf5(-/-)Irf7(-/-) triple knockout mice, which produce little interferon α/ß, and mice lacking the interferon receptor (Ifnar1(-/-)) developed neurological disease and succumbed to ZIKV infection, whereas single Irf3(-/-), Irf5(-/-), and Mavs(-/-) knockout mice exhibited no overt illness. Ifnar1(-/-) mice sustained high viral loads in the brain and spinal cord, consistent with evidence that ZIKV causes neurodevelopmental defects in human fetuses. The testes of Ifnar1(-/-) mice had the highest viral loads, which is relevant to sexual transmission of ZIKV. This model of ZIKV pathogenesis will be valuable for evaluating vaccines and therapeutics as well as understanding disease pathogenesis.


Subject(s)
Disease Models, Animal , Zika Virus Infection/virology , Zika Virus/pathogenicity , Animals , Chlorocebus aethiops , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vero Cells , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control
16.
PLoS Pathog ; 11(7): e1005027, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26204259

ABSTRACT

Impaired immune responses in the elderly lead to reduced vaccine efficacy and increased susceptibility to viral infections. Although several groups have documented age-dependent defects in adaptive immune priming, the deficits that occur prior to antigen encounter remain largely unexplored. Herein, we identify novel mechanisms for compromised adaptive immunity that occurs with aging in the context of infection with West Nile virus (WNV), an encephalitic flavivirus that preferentially causes disease in the elderly. An impaired IgM and IgG response and enhanced vulnerability to WNV infection during aging was linked to delayed germinal center formation in the draining lymph node (DLN). Adoptive transfer studies and two-photon intravital microscopy revealed a decreased trafficking capacity of donor naïve CD4+ T cells from old mice, which manifested as impaired T cell diapedesis at high endothelial venules and reduced cell motility within DLN prior to antigen encounter. Furthermore, leukocyte accumulation in the DLN within the first few days of WNV infection or antigen-adjuvant administration was diminished more generally in old mice and associated with a second aging-related defect in local cytokine and chemokine production. Thus, age-dependent cell-intrinsic and environmental defects in the DLN result in delayed immune cell recruitment and antigen recognition. These deficits compromise priming of early adaptive immune responses and likely contribute to the susceptibility of old animals to acute WNV infection.


Subject(s)
Adaptive Immunity/immunology , Antibodies, Viral/immunology , Lymph Nodes/virology , West Nile Fever/virology , West Nile virus/isolation & purification , Aging , Animals , Brain/immunology , Cytokines/metabolism , Lymph Nodes/immunology , Mice , West Nile Fever/immunology , West Nile virus/immunology
17.
G3 (Bethesda) ; 4(5): 885-90, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24637350

ABSTRACT

The functional redundancy of the three mammalian Golgi-localized, γ-ear-containing, ADP-ribosylation factor-binding proteins (GGAs) was addressed in a previous study. Using insertional mutagenesis, we found that Gga1 or Gga3 homozygous knockout mice were for the most part normal, whereas mice homozygous for two different Gga2 gene-trap alleles exhibited either embryonic or neonatal lethality in the C57BL/6 background, depending on the source of the vector utilized (Byg vs. Tigm, respectively). We now show that the Byg strain harbors a disrupted Gga2 allele that is hypomorphic, indicating that the Byg lethality is attributable to a mechanism independent of GGA2. This is in contrast to the Tigm Gga2 allele, which is a true knockout and establishes a role for GGA2 during the neonatal period. Placement of the Tigm Gga2 allele into the C57BL6/Ola129Sv mixed background results in a lower incidence of neonatal lethality, showing the importance of genetic background in determining the requirement for GGA2 during this period. The Gga2(-/-) mice that survive have reduced body weight at birth and this runted phenotype is maintained through adulthood.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Genes, Lethal , Alleles , Animals , Female , Genotype , Male , Mice , Mice, Knockout , Phenotype
18.
PLoS One ; 7(1): e30184, 2012.
Article in English | MEDLINE | ID: mdl-22291915

ABSTRACT

Numerous studies using cultured mammalian cells have shown that the three GGAs (Golgi-localized, gamma-ear containing, ADP-ribosylation factor- binding proteins) function in the transport of cargo proteins between the trans- Golgi network and endosomes. However, the in vivo role(s) of these adaptor proteins and their possible functional redundancy has not been analyzed. In this study, the genes encoding GGAs1-3 were disrupted in mice by insertional mutagenesis. Loss of GGA1 or GGA3 alone was well tolerated whereas the absence of GGA2 resulted in embryonic or neonatal lethality, depending on the genetic background of the mice. Thus, GGA2 mediates a vital function that cannot be compensated for by GGA1and/or GGA3. The combined loss of GGA1 and GGA3 also resulted in a high incidence of neonatal mortality but in this case the expression level of GGA2 may be inadequate to compensate for the loss of the other two GGAs. We conclude that the three mammalian GGAs are essential proteins that are not fully redundant.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/physiology , Growth and Development/genetics , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian , Female , Humans , Male , Mammals/embryology , Mammals/genetics , Mammals/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family/physiology , Substrate Specificity/genetics
19.
J Cell Biol ; 180(3): 467-72, 2008 Feb 11.
Article in English | MEDLINE | ID: mdl-18250197

ABSTRACT

The adaptor protein AP-1 is the major coat protein involved in the formation of clathrin-coated vesicles at the trans-Golgi network. The prevailing view is that AP-1 recruitment involves coincident binding to multiple low-affinity sites comprising adenosine diphosphate ribosylation factor 1 (Arf-1)-guanosine triphosphate (GTP), cargo sorting signals, and phosphoinositides. We now show that binding of cargo signal peptides to AP-1 induces a conformational change in its core domain that greatly enhances its interaction with Arf-1-GTP. In addition, we provide evidence for cross talk between the dileucine and tyrosine binding sites within the AP-1 core domain such that binding of a cargo signal to one site facilitates binding to the other site. The stable association of AP-1 with Arf-1-GTP, which is induced by cargo signals, would serve to provide sufficient time for adaptor polymerization and clathrin recruitment while ensuring the packaging of cargo molecules into the forming transport vesicles.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Guanosine Triphosphate/metabolism , Signal Transduction/genetics , Transcription Factor AP-1/metabolism , Transport Vesicles/genetics , ADP-Ribosylation Factor 1/genetics , Animals , Binding Sites/genetics , Cattle , Clathrin/metabolism , Clathrin-Coated Vesicles/metabolism , Clathrin-Coated Vesicles/ultrastructure , Guanosine Triphosphate/genetics , Humans , Leucine/metabolism , Polymers/metabolism , Protein Binding/genetics , Protein Conformation , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Subcellular Fractions , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/genetics , Tyrosine/metabolism
20.
Virology ; 358(2): 283-90, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17010406

ABSTRACT

VZV gK, an essential glycoprotein that is conserved among the alphaherpesviruses, is believed to participate in membrane fusion and cytoplasmic virion morphogenesis based on analogy to its HSV-1 homolog. However, the production of VZV gK-specific antibodies has proven difficult presumably due to its highly hydrophobic nature and, therefore, VZV gK has received limited study. To overcome this obstacle, we inserted a FLAG epitope into gK near its amino terminus and produced VZV recombinants expressing epitope-tagged gK (VZV gK-F). These recombinants grew indistinguishably from native VZV, and FLAG-tagged gK could be readily detected in VZV gK-F-infected cells. FACS analysis established that gK is transported to the plasma membrane of infected cells, while indirect immunofluorescence demonstrated that gK accumulates predominately in the Golgi. Using VZV gK-F-infected cells we demonstrated that VZV gK, like several other herpesvirus glycoproteins, is efficiently endocytosed from the plasma membrane. However, pulse-labeling experiments revealed that the half-life of gK is considerably shorter than that of other VZV glycoproteins including gB, gE and gH. This finding suggests that gK may be required in lower abundance than other viral glycoproteins during virion morphogenesis or viral entry.


Subject(s)
Cell Membrane/metabolism , Glycoproteins/metabolism , Herpesvirus 3, Human/physiology , Viral Envelope Proteins/metabolism , Animals , Biological Transport , Cell Line , Golgi Apparatus/metabolism , Half-Life , Humans , Reassortant Viruses , Recombinant Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...