Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Commun Biol ; 7(1): 494, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658802

Inflammatory monocytes (iMO) are recruited from the bone marrow to the brain during viral encephalitis. C-C motif chemokine receptor (CCR) 2 deficiency substantially reduces iMO recruitment for most, but not all encephalitic viruses. Here we show CCR7 acts synergistically with CCR2 to control this process. Following Herpes simplex virus type-1 (HSV-1), or La Crosse virus (LACV) infection, we find iMO proportions are reduced by approximately half in either Ccr2 or Ccr7 knockout mice compared to control mice. However, Ccr2/Ccr7 double knockouts eliminate iMO recruitment following infection with either virus, indicating these receptors together control iMO recruitment. We also find that LACV induces a more robust iMO recruitment than HSV-1. However, unlike iMOs in HSV-1 infection, LACV-recruited iMOs do not influence neurological disease development. LACV-induced iMOs have higher expression of proinflammatory and proapoptotic but reduced mitotic, phagocytic and phagolysosomal transcripts compared to HSV-1-induced iMOs. Thus, virus-specific activation of iMOs affects their recruitment, activation, and function.


Brain , Herpesvirus 1, Human , La Crosse virus , Mice, Knockout , Monocytes , Receptors, CCR2 , Receptors, CCR7 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Brain/virology , Brain/metabolism , Brain/immunology , Herpesvirus 1, Human/physiology , La Crosse virus/genetics , La Crosse virus/physiology , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Encephalitis, California/virology , Encephalitis, California/genetics , Encephalitis, California/metabolism , Encephalitis, California/immunology , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/virology , Female , Male
2.
Viruses ; 12(3)2020 02 25.
Article En | MEDLINE | ID: mdl-32106552

Resident cells in the skin serve as the first innate line of defense against insect-borne pathogens, but the role of these cell types in promoting or limiting arbovirus replication is not completely understood. Here, we have examined the outcome of infection of cultured human keratinocyte cells with La Crosse virus (LACV), using a spontaneously transformed cell line, HaCaT. In single cycle infections, keratinocyte HaCaT cells supported rapid and high level LACV replication, resulting in high virus yields and extensive caspase-dependent cell death. By contrast, multi-cycle LACV replication in HaCaT cells was restricted by an antiviral response elicited by the production of both IFN-ß and IFN-λ. During low multiplicity LACV infections, HaCaT cell death was seen in non-infected bystander cells. Media from LACV-infected cells induced caspase-dependent killing of naïve non-infected HaCaT cells, and this bystander cell death was relieved by IFN-ß neutralizing antibodies or by an inhibitor of JAK-STAT signaling. Naïve HaCaT cells showed dose-dependent killing by treatment with exogenous IFN-ß but not IFN-λ. Our data suggest a model whereby keratinocytes produce IFNs which limit virus spread through both antiviral signaling and by induction of bystander cell death of potential new target cells for infection.


Apoptosis , Encephalitis, California/metabolism , Encephalitis, California/virology , Host-Pathogen Interactions , Interferons/metabolism , Keratinocytes/metabolism , Keratinocytes/virology , La Crosse virus/physiology , Bystander Effect , Caspases/metabolism , Cell Line , Cells, Cultured , Host Specificity , Humans , Virus Replication
3.
J Immunol ; 200(2): 471-476, 2018 01 15.
Article En | MEDLINE | ID: mdl-29246952

Inflammatory monocyte (iMO) recruitment to the brain is a hallmark of many neurologic diseases. Prior to entering the brain, iMOs must egress into the blood from the bone marrow through a mechanism, which for known encephalitic viruses, is CCR2 dependent. In this article, we show that during La Crosse Virus-induced encephalitis, egress of iMOs was surprisingly independent of CCR2, with similar percentages of iMOs in the blood and brain of heterozygous and CCR2-/- mice following infection. Interestingly, CCR2 was required for iMO trafficking from perivascular areas to sites of virus infection within the brain. Thus, CCR2 was not essential for iMO trafficking to the blood or the brain but was essential for trafficking within the brain parenchyma. Analysis of other orthobunyaviruses showed that Jamestown Canyon virus also induced CCR2-independent iMO egress to the blood. These studies demonstrate that the CCR2 requirement for iMO egress to the blood is not universal for all viruses.


Antigens, Ly/metabolism , Encephalitis, California/immunology , Encephalitis, California/metabolism , La Crosse virus , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR2/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , Brain/virology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis, California/virology , Female , Male , Mice , Mice, Transgenic , Monocytes/pathology
4.
Acta Neuropathol ; 130(2): 233-45, 2015 Aug.
Article En | MEDLINE | ID: mdl-25956408

Viral neuroinvasion is a critical step in the pathogenesis of viral encephalitis. Multiple mechanisms of neuroinvasion have been identified, but their relative contribution to central nervous system (CNS) infection remains unclear for many viruses. In this study, we examined neuroinvasion of the mosquito-borne bunyavirus La Crosse (LACV), the leading cause of pediatric viral encephalitis in the USA. We found that the olfactory bulb (OB) and tract were the initial areas of CNS virus infection in mice. Removal of the OB reduced the incidence of LACV-induced disease demonstrating the importance of this area to neuroinvasion. However, we determined that infection of the OB was not due to axonal transport of virus from olfactory sensory neurons as ablation of these cells did not affect viral pathogenesis. Instead, we found that OB capillaries were compromised allowing leakage of virus-sized particles into the brain. Analysis of OB capillaries demonstrated specific alterations in cytoskeletal and Rho GTPase protein expression not observed in capillaries from other brain areas such as the cortex where leakage did not occur. Collectively, these findings indicate that LACV neuroinvasion occurs through hematogenous spread in specific brain regions where capillaries are prone to virus-induced activation such as the OB. Capillaries in these areas may be "hot spots" that are more susceptible to neuroinvasion not only for LACV, but other neurovirulent viruses as well.


Capillaries/metabolism , Capillary Permeability/physiology , Cerebral Cortex/metabolism , Encephalitis, California/metabolism , La Crosse virus/pathogenicity , Olfactory Bulb/blood supply , Olfactory Bulb/virology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capillaries/pathology , Capillaries/virology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebral Cortex/virology , Cytoskeleton/metabolism , Disease Models, Animal , Encephalitis, California/pathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Viral Load , Virus Internalization
5.
PLoS One ; 10(4): e0122895, 2015.
Article En | MEDLINE | ID: mdl-25860584

La Crosse encephalitis (LAC) is the leading arboviral disease among children, and was previously limited to the upper Midwest. In 2012 nine pediatric cases of LAC occurred in eastern Tennessee, including one fatal case. In an attempt to identify sites near an active LACv infection and describe the abundance and distribution of potential LACv vectors near a fatal LAC case in the Appalachian region, we initiated an end of season study using a combination of questing and oviposition mosquito traps placed at forty-nine sites consisting of cemeteries and houses within 16 radial kilometers of two pediatric infections. LACv was isolated from three Aedes triseriatus pools collected from cemeteries and spatial clustering analysis identified clusters of Ae. triseriatus and Ae. albopictus populations that overlapped in the same area as the 2012 LACv cases. Results indicate cemeteries are effective sites for monitoring LACv. The role of cemeteries and specific environmental features will be the focus of future investigations.


Aedes/virology , La Crosse virus/isolation & purification , Aedes/growth & development , Animals , Cemeteries , Child , Cluster Analysis , Encephalitis, California/metabolism , Encephalitis, California/pathology , Encephalitis, California/virology , Female , Humans , Insect Vectors/virology , La Crosse virus/genetics , Male , Ovum/virology , RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction
6.
J Virol ; 86(15): 7988-8001, 2012 Aug.
Article En | MEDLINE | ID: mdl-22623766

La Crosse virus (LACV) is a leading cause of pediatric encephalitis and aseptic meningitis in the midwestern and southern United States, where it is considered an emerging human pathogen. No specific therapies or vaccines are available for LACV or any other orthobunyaviruses. Inhibition of LACV entry into cells is a potential target for therapeutic intervention, but this approach is limited by our current knowledge of the entry process. Here, we determined that clathrin-mediated endocytosis is the primary mechanism of orthobunyavirus entry and identified key cellular factors in this process. First, we demonstrated that LACV colocalized with clathrin shortly after infection in HeLa cells; we then confirmed the functional requirement of dynamin- and clathrin-mediated endocytosis for orthobunyavirus entry using several independent assays and, importantly, extended these findings to primary neuronal cultures. We also determined that macropinocytosis and caveolar endocytosis, both established routes of virus entry, are not critical for cellular entry of LACV. Moreover, we demonstrated that LACV infection is dependent on Rab5, which plays an important regulatory role in early endosomes, but not on Rab7, which is associated with late endosomes. These findings provide the first description of bunyavirus entry into cells of the central nervous system, where infection can cause severe neurological disease, and will aid in the design and development of antivirals and therapeutics that may be useful in the treatment of LACV and, more broadly, arboviral infections of the central nervous system.


Clathrin/metabolism , Encephalitis, California/metabolism , Endocytosis , Endosomes/metabolism , La Crosse virus/metabolism , Virus Internalization , Animals , Chlorocebus aethiops , Clathrin/genetics , Cricetinae , Encephalitis, California/drug therapy , Encephalitis, California/genetics , Endosomes/genetics , Endosomes/virology , HeLa Cells , Humans , La Crosse virus/genetics , Vero Cells , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
7.
J Virol ; 70(8): 5329-35, 1996 Aug.
Article En | MEDLINE | ID: mdl-8764043

La Crosse virus causes a highly cytopathic infection in cultured cells and in the murine central nervous system (CNS), with widespread neuronal destruction. In some viral infections of the CNS, apoptosis, or programmed cell death, has been proposed as a mechanism for cytopathology (Y. Shen and T. E. Shenk, Curr. Opin. Genet. Dev. 5:105-111, 1995). To determine whether apoptosis plays a role in La Crosse virus-induced cell death, we performed experiments with newborn mice and two neural tissue culture models. Newborn mice infected with La Crosse virus showed evidence of apoptosis with the terminal deoxynucleotidyl transferase-mediated nicked-end labeling (TUNEL) assay and, concomitantly, histopathological suggestion of neuronal dropout. Infection of tissue culture cells also resulted in DNA fragmentation, TUNEL reactivity, and morphological changes in the nuclei characteristic of apoptotic cells. As in one other system (S. Ubol, P. C. Tucker, D. E. Griffin, and J. M. Hardwick, Proc. Natl. Acad. Sci. USA 91:5202-5206, 1994), expression of the human proto-oncogene bcl-2 was able to protect one neuronal cell line, N18-RE-105, from undergoing apoptosis after La Crosse virus infection and prolonged the survival of infected cells. Nevertheless, expression of bcl-2 did not prevent eventual cytopathicity. However, a human neuronal cell line, NT2N, was resistant to both apoptosis and other types of cytopathicity after infection with La Crosse virus, reaffirming the complexity of cell death. Our results show that apoptosis is an important consequence of La Crosse virus infection in vivo and in vitro.


Apoptosis , Central Nervous System/virology , Encephalitis, California , La Crosse virus , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Animals , Cell Differentiation , Cell Line , Central Nervous System/pathology , Encephalitis, California/metabolism , Encephalitis, California/pathology , Humans , Mice , Proto-Oncogene Mas
...