Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jennifer Govero is active.

Publication


Featured researches published by Jennifer Govero.


Cell Host & Microbe | 2016

A Mouse Model of Zika Virus Pathogenesis

Helen M. Lazear; Jennifer Govero; Amber M. Smith; Derek J. Platt; Estefania Fernandez; Jonathan J. Miner; Michael S. Diamond

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/β, 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.


Nature | 2016

Zika virus infection damages the testes in mice

Jennifer Govero; Prabagaran Esakky; Suzanne Scheaffer; Estefania Fernandez; Andrea Drury; Derek J. Platt; Matthew J. Gorman; Justin M. Richner; Elizabeth A. Caine; Vanessa Salazar; Kelle H. Moley; Michael S. Diamond

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.


Cell Reports | 2016

Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus in Tears.

Jonathan J. Miner; Abdoulaye Sene; Justin M. Richner; Amber M. Smith; Andrea Santeford; Norimitsu Ban; James Weger-Lucarelli; Francesca Manzella; Claudia Rückert; Jennifer Govero; Kevin K. Noguchi; Gregory D. Ebel; Michael S. Diamond; Rajendra S. Apte

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.


Antiviral Research | 2017

The FDA-approved drug sofosbuvir inhibits Zika virus infection

Kristen M. Bullard-Feibelman; Jennifer Govero; Zhe Zhu; Vanessa Salazar; Milena Veselinovic; Michael S. Diamond; Brian J. Geiss

&NA; 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. HighlightsSofosbuvir reduces replication of multiple ZIKV isolates in human liver and placental cells.Sofosbuvir protects human neuronal stem cells from ZIKV infection.Oral administration of sofosbuvir via drinking reduces ZIKV death in mice.Sofosbuvir should be evaluated as an anti‐ZIKV treatment in non‐rodent species.


Journal of Cell Biology | 2008

Binding of cargo sorting signals to AP-1 enhances its association with ADP ribosylation factor 1–GTP

Intaek Lee; Balraj Doray; Jennifer Govero; Stuart Kornfeld

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.


PLOS Pathogens | 2015

Age-dependent cell trafficking defects in draining lymph nodes impair adaptive immunity and control of West Nile virus infection

Justin M. Richner; Grzegorz B. Gmyrek; Jennifer Govero; Yizheng Tu; Gerritje J.W. van der Windt; Talibah Metcalf; Elias K. Haddad; Johannes Textor; Mark J. Miller; Michael S. Diamond

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.


Journal of Clinical Investigation | 2017

Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects

Elena Bekerman; Gregory Neveu; Ana Shulla; Jennifer M. Brannan; Szu-Yuan Pu; Stanley Wang; Fei Xiao; Rina Barouch-Bentov; Russell R. Bakken; Roberto Mateo; Jennifer Govero; Claude M. Nagamine; Michael S. Diamond; Steven De Jonghe; Piet Herdewijn; John M. Dye; Glenn Randall; Shirit Einav

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.


PLOS ONE | 2012

Analysis of Gga null mice demonstrates a non-redundant role for mammalian GGA2 during development.

Jennifer Govero; Balraj Doray; Hongdong Bai; Stuart Kornfeld

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.


G3: Genes, Genomes, Genetics | 2014

Impact of genetic background on neonatal lethality of Gga2 gene-trap mice

Balraj Doray; Jennifer Govero; Stuart Kornfeld

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.


Mbio | 2018

Mouse and human monoclonal antibodies protect against infection by multiple genotypes of japanese encephalitis virus

Estefania Fernandez; Nurgun Kose; Melissa A. Edeling; Jagat Adhikari; Gopal Sapparapu; Susana M. Lazarte; Christopher A. Nelson; Jennifer Govero; Michael L. Gross; Daved H. Fremont; James E. Crowe; Michael S. Diamond

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. 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.

Collaboration


Dive into the Jennifer Govero's collaboration.

Top Co-Authors

Avatar

Michael S. Diamond

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Estefania Fernandez

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Amber M. Smith

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Balraj Doray

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Derek J. Platt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Caine

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jonathan J. Miner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Justin M. Richner

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Kelle H. Moley

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Stuart Kornfeld

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge