Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Nat Immunol ; 20(5): 559-570, 2019 05.
Article in English | MEDLINE | ID: mdl-30996332

ABSTRACT

The C-type lectin receptor-Syk (spleen tyrosine kinase) adaptor CARD9 facilitates protective antifungal immunity within the central nervous system (CNS), as human deficiency in CARD9 causes susceptibility to fungus-specific, CNS-targeted infection. CARD9 promotes the recruitment of neutrophils to the fungus-infected CNS, which mediates fungal clearance. In the present study we investigated host and pathogen factors that promote protective neutrophil recruitment during invasion of the CNS by Candida albicans. The cytokine IL-1ß served an essential function in CNS antifungal immunity by driving production of the chemokine CXCL1, which recruited neutrophils expressing the chemokine receptor CXCR2. Neutrophil-recruiting production of IL-1ß and CXCL1 was induced in microglia by the fungus-secreted toxin Candidalysin, in a manner dependent on the kinase p38 and the transcription factor c-Fos. Notably, microglia relied on CARD9 for production of IL-1ß, via both transcriptional regulation of Il1b and inflammasome activation, and of CXCL1 in the fungus-infected CNS. Microglia-specific Card9 deletion impaired the production of IL-1ß and CXCL1 and neutrophil recruitment, and increased fungal proliferation in the CNS. Thus, an intricate network of host-pathogen interactions promotes antifungal immunity in the CNS; this is impaired in human deficiency in CARD9, which leads to fungal disease of the CNS.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Candidiasis/immunology , Chemokine CXCL1/immunology , Interleukin-1beta/immunology , Microglia/immunology , Neutrophils/immunology , Animals , Brain/immunology , Brain/metabolism , Brain/microbiology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Candida albicans/immunology , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/microbiology , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/microbiology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Neutrophils/microbiology
2.
PLoS Pathog ; 17(10): e1009967, 2021 10.
Article in English | MEDLINE | ID: mdl-34648590

ABSTRACT

Cell death plays a critical role in inflammatory responses. During pyroptosis, inflammatory caspases cleave Gasdermin D (GSDMD) to release an N-terminal fragment that generates plasma membrane pores that mediate cell lysis and IL-1 cytokine release. Terminal cell lysis and IL-1ß release following caspase activation can be uncoupled in certain cell types or in response to particular stimuli, a state termed hyperactivation. However, the factors and mechanisms that regulate terminal cell lysis downstream of GSDMD cleavage remain poorly understood. In the course of studies to define regulation of pyroptosis during Yersinia infection, we identified a line of Card19-deficient mice (Card19lxcn) whose macrophages were protected from cell lysis and showed reduced apoptosis and pyroptosis, yet had wild-type levels of caspase activation, IL-1 secretion, and GSDMD cleavage. Unexpectedly, CARD19, a mitochondrial CARD-containing protein, was not directly responsible for this, as an independently-generated CRISPR/Cas9 Card19 knockout mouse line (Card19Null) showed no defect in macrophage cell lysis. Notably, Card19 is located on chromosome 13, immediately adjacent to Ninj1, which was recently found to regulate cell lysis downstream of GSDMD activation. RNA-seq and western blotting revealed that Card19lxcn BMDMs have significantly reduced NINJ1 expression, and reconstitution of Ninj1 in Card19lxcn immortalized BMDMs restored their ability to undergo cell lysis in response to caspase-dependent cell death stimuli. Card19lxcn mice exhibited increased susceptibility to Yersinia infection, whereas independently-generated Card19Null mice did not, demonstrating that cell lysis itself plays a key role in protection against bacterial infection, and that the increased infection susceptibility of Card19lxcn mice is attributable to loss of NINJ1. Our findings identify genetic targeting of Card19 being responsible for off-target effects on the adjacent gene Ninj1, disrupting the ability of macrophages to undergo plasma membrane rupture downstream of gasdermin cleavage and impacting host survival and bacterial control during Yersinia infection.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Macrophages/metabolism , Nerve Growth Factors/metabolism , Yersinia Infections/pathology , Animals , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Pyroptosis/physiology , Yersinia Infections/metabolism
3.
PLoS Comput Biol ; 17(5): e1007986, 2021 05.
Article in English | MEDLINE | ID: mdl-34014917

ABSTRACT

The adaptive immune system serves as a potent and highly specific defense mechanism against pathogen infection. One component of this system, the effector T cell, facilitates pathogen clearance upon detection of specific antigens by the T cell receptor (TCR). A critical process in effector T cell activation is transmission of signals from the TCR to a key transcriptional regulator, NF-κB. The transmission of this signal involves a highly dynamic process in which helical filaments of Bcl10, a key protein constituent of the TCR signaling cascade, undergo competing processes of polymeric assembly and macroautophagy-dependent degradation. Through computational analysis of three-dimensional, super-resolution optical micrographs, we quantitatively characterize TCR-stimulated Bcl10 filament assembly and length dynamics, and demonstrate that filaments become shorter over time. Additionally, we develop an image-based, bootstrap-like resampling method that demonstrates the preferred association between autophagosomes and both Bcl10-filament ends and punctate-Bcl10 structures, implying that autophagosome-driven macroautophagy is directly responsible for Bcl10 filament shortening. We probe Bcl10 polymerization-depolymerization dynamics with a stochastic Monte-Carlo simulation of nucleation-limited filament assembly and degradation, and we show that high probabilities of filament nucleation in response to TCR engagement could provide the observed robust, homogeneous, and tunable response dynamic. Furthermore, we demonstrate that the speed of filament disassembly preferentially at filament ends provides effective regulatory control. Taken together, these data suggest that Bcl10 filament growth and degradation act as an excitable system that provides a digital response mechanism and the reliable timing critical for T cell activation and regulatory processes.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Algorithms , Animals , Autophagosomes/immunology , Autophagosomes/metabolism , B-Cell CLL-Lymphoma 10 Protein/chemistry , B-Cell CLL-Lymphoma 10 Protein/genetics , Cell Line , Computational Biology , Computer Simulation , Mice , Models, Biological , Monte Carlo Method , Polymerization , Proteolysis , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
4.
Immunity ; 36(6): 947-58, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22658522

ABSTRACT

The adaptor protein Bcl10 is a critically important mediator of T cell receptor (TCR)-to-NF-κB signaling. Bcl10 degradation is a poorly understood biological phenomenon suggested to reduce TCR activation of NF-κB. Here we have shown that TCR engagement triggers the degradation of Bcl10 in primary effector T cells but not in naive T cells. TCR engagement promoted K63 polyubiquitination of Bcl10, causing Bcl10 association with the autophagy adaptor p62. Paradoxically, p62 binding was required for both Bcl10 signaling to NF-κB and gradual degradation of Bcl10 by autophagy. Bcl10 autophagy was highly selective, as shown by the fact that it spared Malt1, a direct Bcl10 binding partner. Blockade of Bcl10 autophagy enhanced TCR activation of NF-κB. Together, these data demonstrate that selective autophagy of Bcl10 is a pathway-intrinsic homeostatic mechanism that modulates TCR signaling to NF-κB in effector T cells. This homeostatic process may protect T cells from adverse consequences of unrestrained NF-κB activation, such as cellular senescence.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autophagy/physiology , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy-Related Proteins , B-Cell CLL-Lymphoma 10 Protein , Caspases/physiology , Cell Differentiation , Cytosol/immunology , Cytosol/ultrastructure , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Homeostasis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/physiology , Phagosomes/physiology , Phagosomes/ultrastructure , Protein Interaction Mapping , Sequestosome-1 Protein , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/ultrastructure , Th2 Cells/immunology , Th2 Cells/ultrastructure , Ubiquitin-Conjugating Enzymes/physiology
5.
Cell Immunol ; 356: 104161, 2020 10.
Article in English | MEDLINE | ID: mdl-32768663

ABSTRACT

T cell responses to antigen are initiated by engagement of the T cell receptor (TCR)1, leading to activation of diverse signaling cascades, including an incompletely defined pathway that triggers rapid remodeling of the actin cytoskeleton. Defects in the control of actin dynamics and organization are associated with several human immunodeficiency diseases, emphasizing the importance of cytoskeletal remodeling in the functioning of the adaptive immune system. Here, we investigate the role of the adaptor protein Bcl102 in the control of actin dynamics. Although Bcl10 is primarily known as a component of the pathway connecting the TCR to activation of the NF-κB3 transcription factor, a few studies have implicated Bcl10 in antigen receptor-dependent control of actin polymerization and F-actin-dependent functional responses. However, the role of Bcl10 in the regulation of cytoskeletal dynamics remains largely undefined. To investigate the contribution of Bcl10 in the regulation of TCR-dependent cytoskeletal dynamics, we monitored actin dynamics at the immune synapse of primary murine CD8 effector T cells. Quantification of these dynamics reveals two distinct temporal phases distinguished by differences in speed and directionality. Our results indicate that effector CD8 T cells lacking Bcl10 display faster actin flows and more dynamic lamellipodia, compared to wild-type cells. These studies define a role for Bcl10 in TCR-dependent actin dynamics, emphasizing that Bcl10 has important cytoskeleton-directed functions that are likely independent of its role in transmission of NF-κB -activating signals.


Subject(s)
Actins/metabolism , B-Cell CLL-Lymphoma 10 Protein/metabolism , Receptors, Antigen, T-Cell/immunology , Actins/immunology , Animals , B-Cell CLL-Lymphoma 10 Protein/immunology , Male , Mice , Mice, Inbred C57BL , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/immunology , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Synapses/metabolism
6.
Cell Immunol ; 356: 104179, 2020 10.
Article in English | MEDLINE | ID: mdl-32763502

ABSTRACT

After T cell receptor (TCR) engagement, the CARD11-Bcl10-Malt1 (CBM) complex oligomerizes to transduce NF-κB activating signals. Bcl10 is then degraded to limit NF-κB activation. The cDNA AK057716 (BinCARD-1) was reported to encode a novel CARD protein that interacts with Bcl10 and modestly inhibits NF-κB activation. In a later study, a second isoform, BinCARD-2, was identified. Here, we report that the cDNA AK057716 (BinCARD-1) is an incompletely spliced derivative of the gene product of C9orf89, whereas CARD19 (BinCARD-2) represents the properly spliced isoform, with conservation across diverse species. Immunoblotting revealed expression of CARD19 in T cells, but no evidence of BinCARD-1 expression, and microscopy demonstrated that endogenous CARD19 localizes to mitochondria. Although we confirmed that both BinCARD-1 and CARD19 can inhibit NF-κB activation and promote Bcl10 degradation when transiently overexpressed in HEK293T cells, loss of endogenous CARD19 expression had little effect on Bcl10-dependent NF-κB activation, activation of Malt1 protease function, or Bcl10 degradation after TCR engagement in primary murine CD8 T cells. Together, these data indicate that the only detectable translated product of C9orf89 is the mitochondrial protein CARD19, which does not play a discernible role in TCR-dependent, Bcl10-mediated signal transduction to Malt1 or NF-κB.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/physiology , Receptors, Antigen, T-Cell/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , Databases, Genetic , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Jurkat Cells , Mice , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics
7.
Brain Behav Immun ; 61: 96-109, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27939247

ABSTRACT

Neuroinflammation plays a critical role in the pathogenesis of traumatic brain injury (TBI). TBI induces rapid activation of astrocytes and microglia, infiltration of peripheral leukocytes, and secretion of inflammatory cytokines. In the context of modest or severe TBI, such inflammation contributes to tissue destruction and permanent brain damage. However, it is clear that the inflammatory response is also necessary to promote post-injury healing. To date, anti-inflammatory therapies, including the broad class of non-steroidal anti-inflammatory drugs (NSAIDs), have met with little success in treatment of TBI, perhaps because these drugs have inhibited both the tissue-damaging and repair-promoting aspects of the inflammatory response, or because inhibition of inflammation alone is insufficient to yield therapeutic benefit. Salsalate is an unacetylated salicylate with long history of use in limiting inflammation. This drug is known to block activation of NF-κB, and recent data suggest that salsalate has a number of additional biological activities, which may also contribute to its efficacy in treatment of human disease. Here, we show that salsalate potently blocks pro-inflammatory gene expression and nitrite secretion by microglia in vitro. Using the controlled cortical impact (CCI) model in mice, we find that salsalate has a broad anti-inflammatory effect on in vivo TBI-induced gene expression, when administered post-injury. Interestingly, salsalate also elevates expression of genes associated with neuroprotection and neurogenesis, including the neuropeptides, oxytocin and thyrotropin releasing hormone. Histological analysis reveals salsalate-dependent decreases in numbers and activation-associated morphological changes in microglia/macrophages, proximal to the injury site. Flow cytometry data show that salsalate changes the kinetics of CCI-induced accumulation of various populations of CD11b-positive myeloid cells in the injured brain. Behavioral assays demonstrate that salsalate treatment promotes significant recovery of function following CCI. These pre-clinical data suggest that salsalate may show promise as a TBI therapy with a multifactorial mechanism of action to enhance functional recovery.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain/drug effects , Inflammation/drug therapy , Neuroprotective Agents/therapeutic use , Recovery of Function/drug effects , Salicylates/therapeutic use , Animals , Brain/pathology , Brain Injuries, Traumatic/pathology , Cell Line , Gene Flow/drug effects , Inflammation/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Neuroprotective Agents/pharmacology , Oxytocin/genetics , Salicylates/pharmacology , Thyrotropin-Releasing Hormone/genetics
8.
J Immunol ; 195(7): 2985-3000, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26320249

ABSTRACT

Using the parent-into-F1 model of induced lupus and (C57BL/6 × DBA2) F1 mice as hosts, we compared the inherent lupus-inducing properties of the two parental strain CD4 T cells. To control for donor CD4 recognition of alloantigen, we used H-2(d) identical DBA/2 and B10.D2 donor T cells. We demonstrate that these two normal, nonlupus-prone parental strains exhibit two different T cell activation pathways in vivo. B10.D2 CD4 T cells induce a strong Th1/CMI pathway that is characterized by IL-2/IFN-γ expression, help for CD8 CTLs, and skewing of dendritic cell (DC) subsets toward CD8a DCs, coupled with reduced CD4 T follicular helper cells and transient B cell help. In contrast, DBA/2 CD4 T cells exhibit a reciprocal, lupus-inducing pathway that is characterized by poor IL-2/IFN-γ expression, poor help for CD8 CTLs, and skewing of DC subsets toward plasmacytoid DCs, coupled with greater CD4 T follicular helper cells, prolonged B cell activation, autoantibody formation, and lupus-like renal disease. Additionally, two distinct in vivo splenic gene-expression signatures were induced. In vitro analysis of TCR signaling revealed defective DBA CD4 T cell induction of NF-κB, reduced degradation of IκBα, and increased expression of the NF-κB regulator A20. Thus, attenuated NF-κB signaling may lead to diminished IL-2 production by DBA CD4 T cells. These results indicate that intrinsic differences in donor CD4 IL-2 production and subsequent immune skewing could contribute to lupus susceptibility in humans. Therapeutic efforts to skew immune function away from excessive help for B cells and toward help for CTLs may be beneficial.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Lupus Erythematosus, Systemic/immunology , Th1 Cells/immunology , Animals , Autoantibodies/immunology , Cysteine Endopeptidases/biosynthesis , Dendritic Cells/classification , Dendritic Cells/immunology , Disease Models, Animal , Female , I-kappa B Proteins/metabolism , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/immunology , Tumor Necrosis Factor alpha-Induced Protein 3
9.
Trends Immunol ; 34(6): 269-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23474202

ABSTRACT

Antigen stimulation of T cell receptor (TCR) signaling to nuclear factor (NF)-κB is required for T cell proliferation and differentiation of effector cells. The TCR-to-NF-κB pathway is generally viewed as a linear sequence of events in which TCR engagement triggers a cytoplasmic cascade of protein-protein interactions and post-translational modifications, ultimately culminating in the nuclear translocation of NF-κB. However, recent findings suggest a more complex picture in which distinct signalosomes, previously unrecognized proteins, and newly identified regulatory mechanisms play key roles in signal transmission. In this review, we evaluate recent data and suggest areas of future emphasis in the study of this important pathway.


Subject(s)
Cell Nucleus/metabolism , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Active Transport, Cell Nucleus , Animals , Humans , Lymphocyte Activation , Multiprotein Complexes/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology
11.
Article in English | MEDLINE | ID: mdl-38770087

ABSTRACT

Henipaviruses are enveloped single-stranded, negative-sense RNA viruses of the paramyxovirus family. Two henipaviruses, Nipah virus and Hendra virus, cause a systemic respiratory and/or neurological disease in humans and ten additional species of mammals, with a high fatality rate. Because of their highly pathogenic nature, Nipah virus and Hendra virus are categorized as BSL-4 pathogens, which limits the number and scope of translational research studies on these important human pathogens. To begin to address this limitation, we are developing a BSL-2 model of authentic henipavirus infection in mice, using the non-pathogenic henipavirus, Cedar virus. Notably, wild-type mice are highly resistant to Hendra virus and Nipah virus infection. However, previous work has shown that mice lacking expression of the type I interferon receptor (IFNAR-KO mice) are susceptible to both viruses. Here, we show that luciferase-expressing recombinant Cedar virus (rCedV-luc) is also able to replicate and establish a transient infection in IFNAR-KO mice, but not in wild-type mice. Using longitudinal bioluminescence imaging (BLI) of luciferase expression, we detected rCedV-luc replication as early as 10 h post-infection. Viral replication peaks between days 1 and 3 post-infection, and declines to levels undetectable by BLI by 7 days post-infection. Immunohistochemistry is consistent with viral infection and replication in endothelial cells and other non-immune cell types within tissue parenchyma. Serology analyses demonstrate significant IgG responses to the Cedar virus surface glycoprotein with potent neutralizing activity in IFNAR-KO mice, whereas antibody responses in wild-type animals were non-significant. Overall, these data suggest that rCedV-luc infection of IFNAR-KO mice represents a viable platform for the study of in vivo henipavirus replication, anti-henipavirus host responses and henipavirus-directed therapeutics.

12.
EMBO Mol Med ; 15(10): e16394, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37767784

ABSTRACT

Infections with rabies virus (RABV) and related lyssaviruses are uniformly fatal once virus accesses the central nervous system (CNS) and causes disease signs. Current immunotherapies are thus focused on the early, pre-symptomatic stage of disease, with the goal of peripheral neutralization of virus to prevent CNS infection. Here, we evaluated the therapeutic efficacy of F11, an anti-lyssavirus human monoclonal antibody (mAb), on established lyssavirus infections. We show that a single dose of F11 limits viral load in the brain and reverses disease signs following infection with a lethal dose of lyssavirus, even when administered after initiation of robust virus replication in the CNS. Importantly, we found that F11-dependent neutralization is not sufficient to protect animals from mortality, and a CD4 T cell-dependent adaptive immune response is required for successful control of infection. F11 significantly changes the spectrum of leukocyte populations in the brain, and the FcRγ-binding function of F11 contributes to therapeutic efficacy. Thus, mAb therapy can drive potent neutralization-independent T cell-mediated effects, even against an established CNS infection by a lethal neurotropic virus.


Subject(s)
Central Nervous System Infections , Chiroptera , Lyssavirus , Rabies virus , Rabies , Rhabdoviridae Infections , Animals , Humans , Rhabdoviridae Infections/drug therapy , Rhabdoviridae Infections/prevention & control , CD4-Positive T-Lymphocytes , Immunotherapy , Antibodies, Monoclonal/therapeutic use , Rabies/prevention & control
13.
Viruses ; 15(9)2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37766204

ABSTRACT

Rabies is a fatal zoonosis that is considered a re-emerging infectious disease. Although rabies remains endemic in canines throughout much of the world, vaccination programs have essentially eliminated dog rabies in the Americas and much of Europe. However, despite the goal of eliminating dog rabies in the European Union by 2020, sporadic cases of dog rabies still occur in Eastern Europe, including Georgia. To assess the genetic diversity of the strains recently circulating in Georgia, we sequenced seventy-eight RABV-positive samples from the brain tissues of rabid dogs and jackals using Illumina short-read sequencing of total RNA shotgun libraries. Seventy-seven RABV genomes were successfully assembled and annotated, with seventy-four of them reaching the coding-complete status. Phylogenetic analyses of the nucleoprotein (N) and attachment glycoprotein (G) genes placed all the assembled genomes into the Cosmopolitan clade, consistent with the Georgian origin of the samples. An amino acid alignment of the G glycoprotein ectodomain identified twelve different sequences for this domain among the samples. Only one of the ectodomain groups contained a residue change in an antigenic site, an R264H change in the G5 antigenic site. Three isolates were cultured, and these were found to be efficiently neutralized by the human monoclonal antibody A6. Overall, our data show that recently circulating RABV isolates from Georgian canines are predominantly closely related phylogroup I viruses of the Cosmopolitan clade. Current human rabies vaccines should offer protection against infection by Georgian canine RABVs. The genomes have been deposited in GenBank (accessions: OQ603609-OQ603685).


Subject(s)
Rabies Vaccines , Rabies virus , Rabies , Dogs , Animals , Humans , Phylogeny , Jackals , Glycoproteins/genetics , Genomics
14.
PLoS Pathog ; 6(4): e1000892, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20442785

ABSTRACT

Schistosoma blood flukes, which infect over 200 million people globally, co-opt CD4+ T cell-dependent mechanisms to facilitate parasite development and egg excretion. The latter requires Th2 responses, while the mechanism underpinning the former has remained obscure. Using mice that are either defective in T cell receptor (TCR) signaling or that lack TCRs that can respond to schistosomes, we show that naïve CD4+ T cells facilitate schistosome development in the absence of T cell receptor signaling. Concurrently, the presence of naïve CD4+ T cells correlates with both steady-state changes in the expression of genes that are critical for the development of monocytes and macrophages and with significant changes in the composition of peripheral mononuclear phagocyte populations. Finally, we show that direct stimulation of the mononuclear phagocyte system restores blood fluke development in the absence of CD4+ T cells. Thus we conclude that schistosomes co-opt innate immune signals to facilitate their development and that the role of CD4+ T cells in this process may be limited to the provision of non-cognate help for mononuclear phagocyte function. Our findings have significance for understanding interactions between schistosomiasis and other co-infections, such as bacterial infections and human immunodeficiency virus infection, which potently stimulate innate responses or interfere with T cell help, respectively. An understanding of immunological factors that either promote or inhibit schistosome development may be valuable in guiding the development of efficacious new therapies and vaccines for schistosomiasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Schistosoma mansoni/immunology , Schistosomiasis/immunology , Animals , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Gene Expression Profiling , Helminth Proteins/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell/immunology
15.
J Immunol ; 185(8): 4520-4, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20855880

ABSTRACT

TCR-mediated activation of the transcription factor NF-κB is required for T cell proliferation, survival, and effector differentiation. Although this pathway is the subject of intense study, it is not known whether TCR signaling to NF-κB is digital (switch-like) or analog in nature. Through analysis of the phosphorylation and degradation of IκBα and the nuclear translocation and phosphorylation of the NF-κB subunit RelA, we show that TCR-directed NF-κB activation is digital. Furthermore, digitization occurs well upstream of the IκB kinase complex, as protein kinase C translocation to the immunologic synapse and activation-associated aggregation of Bcl10 and Malt1 also demonstrate both digital behavior and high correlation with RelA nuclear translocation. Thus, similar to the TCR-to-MAPK signaling cascade, analog Ag inputs are converted to digital activation outputs to NF-κB at an early step downstream of TCR ligation.


Subject(s)
Lymphocyte Activation/immunology , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Animals , Cell Separation , Enzyme Activation , Flow Cytometry , Humans , I-kappa B Proteins/immunology , I-kappa B Proteins/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , Phosphorylation , Protein Transport/immunology , Receptors, Antigen, T-Cell/immunology
16.
Methods Mol Biol ; 2524: 369-394, 2022.
Article in English | MEDLINE | ID: mdl-35821488

ABSTRACT

Bioluminescence imaging (BLI) is a technique that can be employed to quantify biological processes in living cells. When used in small animal models such as mice, BLI can provide both longitudinal and positional information regarding the biological process under investigation. Although perhaps best known for its utility in non-invasively quantifying tumor burden over time in experimental animals, BLI has also been applied in many pathogenesis models to track pathogen burden and responses to therapeutic interventions. In this chapter, we present a BLI-based method for tracing anatomical progression of lyssavirus infection in a mouse model. We also include validation methods to ensure that semiquantitative BLI data correlate well with viral load. Due to the longitudinal nature of this approach, lyssavirus pathogenesis and therapeutic intervention studies can be performed with far fewer animals than more traditional approaches, which typically require euthanasia of large animal groups at every data collection time point.


Subject(s)
Luminescent Measurements , Lyssavirus , Animals , Diagnostic Imaging , Disease Models, Animal , Luminescent Measurements/methods , Mice
17.
Cells ; 11(7)2022 03 31.
Article in English | MEDLINE | ID: mdl-35406738

ABSTRACT

CARD19 is a mitochondrial protein of unknown function. While CARD19 was originally reported to regulate TCR-dependent NF-κB activation via interaction with BCL10, this function is not recapitulated ex vivo in primary murine CD8+ T cells. Here, we employ a combination of SIM, TEM, and confocal microscopy, along with proteinase K protection assays and proteomics approaches, to identify interacting partners of CARD19 in macrophages. Our data show that CARD19 is specifically localized to the outer mitochondrial membrane. Through deletion of functional domains, we demonstrate that both the distal C-terminus and transmembrane domain are required for mitochondrial targeting, whereas the CARD is not. Importantly, mass spectrometry analysis of 3×Myc-CARD19 immunoprecipitates reveals that CARD19 interacts with the components of the mitochondrial intermembrane bridge (MIB), consisting of mitochondrial contact site and cristae organizing system (MICOS) components MIC19, MIC25, and MIC60, and MICOS-interacting proteins SAMM50 and MTX2. These CARD19 interactions are in part dependent on a properly folded CARD. Consistent with previously reported phenotypes upon siRNA silencing of MICOS subunits, absence of CARD19 correlates with irregular cristae morphology. Based on these data, we propose that CARD19 is a previously unknown interacting partner of the MIB and the MIC19-MIC25-MIC60 MICOS subcomplex that regulates cristae morphology.


Subject(s)
CARD Signaling Adaptor Proteins , CD8-Positive T-Lymphocytes , Mitochondrial Membranes , Mitochondrial Proteins , Animals , CARD Signaling Adaptor Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism
18.
Viruses ; 13(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33804519

ABSTRACT

Australian bat lyssavirus (ABLV) is a rhabdovirus that circulates in four species of pteropid bats (ABLVp) and the yellow-bellied sheath-tailed bat (ABLVs) in mainland Australia. In the three confirmed human cases of ABLV, rabies illness preceded fatality. As with rabies virus (RABV), post-exposure prophylaxis (PEP) for potential ABLV infections consists of wound cleansing, administration of the rabies vaccine and injection of rabies immunoglobulin (RIG) proximal to the wound. Despite the efficacy of PEP, the inaccessibility of human RIG (HRIG) in the developing world and the high immunogenicity of equine RIG (ERIG) has led to consideration of human monoclonal antibodies (hmAbs) as a passive immunization option that offers enhanced safety and specificity. Using a recombinant vesicular stomatitis virus (rVSV) expressing the glycoprotein (G) protein of ABLVs and phage display, we identified two hmAbs, A6 and F11, which completely neutralize ABLVs/ABLVp, and RABV at concentrations ranging from 0.39 and 6.25 µg/mL and 0.19 and 0.39 µg/mL respectively. A6 and F11 recognize overlapping epitopes in the lyssavirus G protein, effectively neutralizing phylogroup 1 lyssaviruses, while having little effect on phylogroup 2 and non-grouped diverse lyssaviruses. These results suggest that A6 and F11 could be effective therapeutic and diagnostic tools for phylogroup 1 lyssavirus infections.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Cross Reactions/immunology , Lyssavirus/classification , Lyssavirus/immunology , Phylogeny , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/blood , Antibodies, Viral/immunology , Australia , Bites and Stings , Cell Surface Display Techniques , Chiroptera/virology , Epitopes/immunology , HEK293 Cells , Horses , Humans , Lyssavirus/genetics , Neutralization Tests , Post-Exposure Prophylaxis , Rabies/prevention & control , Rabies Vaccines/immunology , Rabies virus/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/prevention & control , Rhabdoviridae Infections/therapy , Vesiculovirus/genetics
19.
J Immunol ; 181(9): 6244-54, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941215

ABSTRACT

The cytosolic proteins protein kinase Ctheta (PKCtheta), Bcl10, and Malt1 play critical roles in TCR signaling to the transcription factor NF-kappaB. Our data confirm that CD4(+) T cells from PKCtheta, Bcl10, and Malt1 knockout mice show severe impairment of proliferation in response to TCR stimulation. Unexpectedly, we find that knockout CD8(+) T cells proliferate to a similar extent as wild-type cells in response to strong TCR signals, although a survival defect prevents their accumulation. Both CD4(+) and CD8(+) knockout T cells express activation markers, including CD25, following TCR stimulation. Addition of exogenous IL-2 rescues survival of knockout CD4(+) and CD8(+) T cells, but fails to overcome the proliferation defect of CD4(+) T cells. CD4(+) T cells from knockout mice are extremely deficient in TCR-induced NF-kappaB activation, whereas NF-kappaB activation is only partially impaired in CD8(+) T cells. Overall, our results suggest that defects in TCR signaling through PKCtheta, Bcl10, and Malt1 predominantly impair NF-kappaB activation and downstream functional responses of CD4(+) T cells. In contrast, CD8(+) T cells maintain substantial NF-kappaB signaling, implying the existence of a significant TCR-regulated NF-kappaB activation pathway in CD8(+) T cells that is independent of PKCtheta, Bcl10, and Malt1.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Caspases/deficiency , Cell Proliferation , Down-Regulation/immunology , Isoenzymes/deficiency , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/deficiency , Protein Kinase C/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , B-Cell CLL-Lymphoma 10 Protein , CD4-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Caspases/genetics , Caspases/physiology , Cells, Cultured , Down-Regulation/genetics , Isoenzymes/genetics , Isoenzymes/physiology , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitomycin/immunology , Molecular Sequence Data , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Ovalbumin/immunology , Protein Kinase C/genetics , Protein Kinase C/physiology , Protein Kinase C-theta , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology , Signal Transduction/genetics , Signal Transduction/immunology
20.
J Virol Methods ; 281: 113882, 2020 07.
Article in English | MEDLINE | ID: mdl-32407866

ABSTRACT

Traditional mouse models of lyssavirus pathogenesis rely on euthanizing large groups of animals at various time points post-infection, processing infected tissues, and performing histological and molecular analyses to determine anatomical sites of infection. While powerful by some measures, this approach is limited by the inability to monitor disease progression in the same mice over time. In this study, we established a novel non-invasive mouse model of lyssavirus pathogenesis, which consists of longitudinal imaging of a luciferase-expressing Australian bat lyssavirus (ABLV) reporter virus. In vivo bioluminescence imaging (BLI) in mice revealed viral spread from a peripheral site of inoculation into the central nervous system (CNS), with kinetically and spatially distinct foci of replication in the footpad, spinal cord, and hindbrain. Detection of virus within the CNS was associated with onset of clinical disease. Quantification of virus-derived luminescent signal in the brain was found to be a reliable measure of viral replication, when compared to traditional molecular methods. Furthermore, we demonstrate that in vivo imaging of ABLV infection is not restricted to the use of albino strains of mice, but rather strong BLI signal output can be achieved by shaving the hair from the heads and spines of pigmented strains, such as C57BL/6. Overall, our data show that in vivo BLI can be used to rapidly and non-invasively identify sites of lyssavirus replication and to semi-quantitatively determine viral load without the need to sacrifice mice at multiple time points.


Subject(s)
Antibodies, Viral/blood , Disease Models, Animal , Lyssavirus/pathogenicity , Rhabdoviridae Infections/virology , Animals , Brain/virology , Cell Line , Female , HEK293 Cells , Humans , Longitudinal Studies , Luciferases/genetics , Luminescent Measurements , Lyssavirus/enzymology , Lyssavirus/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Imaging , Rhabdoviridae Infections/immunology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL