Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Pathogens ; 13(2)2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38392905

ABSTRACT

Single-cell RNA sequencing has soared in popularity in recent years. The ability to deeply profile the states of individual cells during the course of disease or infection has helped to expand our knowledge of coordinated responses. However, significant challenges arise when performing this analysis in high containment settings such as biosafety level 3 (BSL-3), BSL-3+ and BSL-4. Working in containment is necessary for many important pathogens, such as Ebola virus, Marburg virus, Lassa virus, Nipah and Hendra viruses. Since standard operating procedures (SOPs) for inactivation are extensive and may compromise sample integrity, we tested whether the removal of single-cell sequencing libraries from containment laboratories using existing inactivation protocols for nucleic acid extraction (Trizol, RLT buffer, or AVL buffer) was feasible. We have demonstrated that the inactivation does not affect sample quality and can work with existing methods for inactivation.

2.
Nat Commun ; 14(1): 4481, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37491352

ABSTRACT

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.


Subject(s)
COVID-19 , Interferon Type I , Humans , Mice , Animals , Cytokines , SARS-CoV-2 , Mice, Transgenic , Inflammation/genetics , Disease Models, Animal , Lung
3.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-35233576

ABSTRACT

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18- hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.

4.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35039442

ABSTRACT

Advanced age is a key predictor of severe COVID-19. To gain insight into this relationship, we used the rhesus macaque model of SARS-CoV-2 infection. Eight older and eight younger macaques were inoculated with SARS-CoV-2. Animals were evaluated using viral RNA quantification, clinical observations, thoracic radiographs, single-cell transcriptomics, multiparameter flow cytometry, multiplex immunohistochemistry, cytokine detection, and lipidomics analysis at predefined time points in various tissues. Differences in clinical signs, pulmonary infiltrates, and virus replication were limited. Transcriptional signatures of inflammation-associated genes in bronchoalveolar lavage fluid at 3 dpi revealed efficient mounting of innate immune defenses in both cohorts. However, age-specific divergence of immune responses emerged during the post-acute phase. Older animals exhibited sustained local inflammatory innate responses, whereas local effector T-cell responses were induced earlier in the younger animals. Circulating lipid mediator and cytokine levels highlighted increased repair-associated signals in the younger animals, and persistent pro-inflammatory responses in the older animals. In summary, despite similar disease outcomes, multi-omics profiling suggests that age may delay or impair antiviral cellular immune responses and delay efficient return to immune homeostasis.


Subject(s)
Aging/immunology , COVID-19/immunology , COVID-19/veterinary , SARS-CoV-2/immunology , Acute Disease , Animals , Antibody Formation/immunology , Bronchoalveolar Lavage Fluid , COVID-19/complications , COVID-19/genetics , Cytokines/blood , Gene Expression Regulation , Gene Regulatory Networks , Genomics , Immunity, Cellular/genetics , Immunomodulation , Inflammation/complications , Inflammation/pathology , Lung/immunology , Lung/pathology , Lung/virology , Lymphoid Tissue/pathology , Macaca mulatta/immunology , Macaca mulatta/virology , Models, Biological , Single-Cell Analysis , T-Lymphocytes/immunology , Transcription, Genetic
5.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Article in English | MEDLINE | ID: mdl-34855915

ABSTRACT

Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.


Subject(s)
Disease Models, Animal , Encephalitis Viruses, Tick-Borne/pathogenicity , Encephalitis, Tick-Borne/virology , Hemorrhagic Fevers, Viral/virology , Macaca nemestrina , Animals , Chlorocebus aethiops , Cytokines/blood , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/pathology , Female , HEK293 Cells , Hemorrhagic Fevers, Viral/immunology , Hemorrhagic Fevers, Viral/pathology , Humans , Lymph Nodes/virology , Vero Cells , Viremia
6.
Cell Rep ; 37(4): 109888, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706234

ABSTRACT

Dysregulated inflammation dominated by chemokine expression is a key feature of disease following infection with the globally important human pathogens Zika virus (ZIKV) and dengue virus, but a mechanistic understanding of how pro-inflammatory responses are initiated is lacking. Mitophagy is a quality-control mechanism that regulates innate immune signaling and cytokine production through selective degradation of damaged mitochondria. Here, we demonstrate that ZIKV nonstructural protein 5 (NS5) antagonizes mitophagy by binding to the host protein Ajuba and preventing its translocation to depolarized mitochondria where it is required for PINK1 activation and downstream signaling. Consequent mitophagy suppression amplifies the production of pro-inflammatory chemokines through protein kinase R (PKR) sensing of mitochondrial RNA. In Ajuba-/- mice, ZIKV induces early expression of pro-inflammatory chemokines associated with significantly enhanced dissemination to tissues. This work identifies Ajuba as a critical regulator of mitophagy and demonstrates a role for mitophagy in limiting systemic inflammation following infection by globally important human viruses.


Subject(s)
LIM Domain Proteins/metabolism , Mitophagy , Protein Kinases/metabolism , Signal Transduction , Zika Virus Infection/metabolism , Zika Virus/metabolism , eIF-2 Kinase/metabolism , A549 Cells , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , LIM Domain Proteins/genetics , Mice , Mice, Knockout , Protein Kinases/genetics , Vero Cells , Zika Virus/genetics , Zika Virus Infection/genetics , eIF-2 Kinase/genetics
7.
Nat Commun ; 12(1): 5454, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526512

ABSTRACT

Chlamydia trachomatis infection causes severe inflammatory disease resulting in blindness and infertility. The pathophysiology of these diseases remains elusive but myeloid cell-associated inflammation has been implicated. Here we show NLRP3 inflammasome activation is essential for driving a macrophage-associated endometritis resulting in infertility by using a female mouse genital tract chlamydial infection model. We find the chlamydial parasitophorous vacuole protein CT135 triggers NLRP3 inflammasome activation via TLR2/MyD88 signaling as a pathogenic strategy to evade neutrophil host defense. Paradoxically, a consequence of CT135 mediated neutrophil killing results in a submucosal macrophage-associated endometritis driven by ATP/P2X7R induced NLRP3 inflammasome activation. Importantly, macrophage-associated immunopathology occurs independent of macrophage infection. We show chlamydial infection of neutrophils and epithelial cells produce elevated levels of extracellular ATP. We propose this source of ATP serves as a DAMP to activate submucosal macrophage NLRP3 inflammasome that drive damaging immunopathology. These findings offer a paradigm of sterile inflammation in infectious disease pathogenesis.


Subject(s)
Chlamydia Infections/immunology , Chlamydia/immunology , Inflammation/immunology , Myeloid Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neutrophils/immunology , Receptors, Purinergic P2X7/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Chlamydia/physiology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Disease Models, Animal , Female , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Immune Evasion/immunology , Inflammation/metabolism , Inflammation/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism
8.
Sci Transl Med ; 13(578)2021 01 27.
Article in English | MEDLINE | ID: mdl-33431511

ABSTRACT

Detailed knowledge about the dynamics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is important for uncovering the viral and host factors that contribute to coronavirus disease 2019 (COVID-19) pathogenesis. Old-World nonhuman primates recapitulate mild to moderate cases of COVID-19, thereby serving as important pathogenesis models. We compared African green monkeys inoculated with infectious SARS-CoV-2 or irradiated, inactivated virus to study the dynamics of virus replication throughout the respiratory tract. Genomic RNA from the animals inoculated with the irradiated virus was found to be highly stable, whereas subgenomic RNA, an indicator of viral replication, was found to degrade quickly. We combined this information with single-cell RNA sequencing of cells isolated from the lung and lung-draining mediastinal lymph nodes and developed new analysis methods for unbiased targeting of important cells in the host response to SARS-CoV-2 infection. Through detection of reads to the viral genome, we were able to determine that replication of the virus in the lungs appeared to occur mainly in pneumocytes, whereas macrophages drove the inflammatory response. Monocyte-derived macrophages recruited to the lungs, rather than tissue-resident alveolar macrophages, were most likely to be responsible for phagocytosis of infected cells and cellular debris early in infection, with their roles switching during clearance of infection. Together, our dataset provides a detailed view of the dynamics of virus replication and host responses over the course of mild COVID-19 and serves as a valuable resource to identify therapeutic targets.


Subject(s)
COVID-19/epidemiology , COVID-19/virology , Lung/virology , SARS-CoV-2/physiology , Sequence Analysis, RNA , Single-Cell Analysis , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Animals , Bronchoalveolar Lavage Fluid/virology , COVID-19/genetics , Chlorocebus aethiops , DNA, Viral/genetics , Female , Genome, Viral/genetics , Inflammation/pathology , Lung/pathology , Lymph Nodes/pathology , Macrophages/pathology , Macrophages/virology , Male , Mediastinum/pathology , Transcription, Genetic , Viral Load , Virus Replication
9.
bioRxiv ; 2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32839775

ABSTRACT

Detailed knowledge about the dynamics of SARS-CoV-2 infection is important for unraveling the viral and host factors that contribute to COVID-19 pathogenesis. Old-World nonhuman primates recapitulate mild-moderate COVID-19 cases, thereby serving as important pathogenesis models. We compared African green monkeys inoculated with SARS-CoV-2 or inactivated virus to study the dynamics of virus replication throughout the respiratory tract. RNA sequencing of single cells from the lungs and mediastinal lymph nodes allowed a high-resolution analysis of virus replication and host responses over time. Viral replication was mainly localized to the lower respiratory tract, with evidence of replication in the pneumocytes. Macrophages were found to play a role in initiating a pro-inflammatory state in the lungs, while also interacting with infected pneumocytes. Our dataset provides a detailed view of changes in host and virus replication dynamics over the course of mild COVID-19 and serves as a valuable resource to identify therapeutic targets.

10.
Nature ; 585(7825): 414-419, 2020 09.
Article in English | MEDLINE | ID: mdl-32641828

ABSTRACT

Zika virus (ZIKV) belongs to the family Flaviviridae, and is related to other viruses that cause human diseases. Unlike other flaviviruses, ZIKV infection can cause congenital neurological disorders and replicates efficiently in reproductive tissues1-3. Here we show that the envelope protein (E) of ZIKV is polyubiquitinated by the E3 ubiquitin ligase TRIM7 through Lys63 (K63)-linked polyubiquitination. Accordingly, ZIKV replicates less efficiently in the brain and reproductive tissues of Trim7-/- mice. Ubiquitinated E is present on infectious virions of ZIKV when they are released from specific cell types, and enhances virus attachment and entry into cells. Specifically, K63-linked polyubiquitin chains directly interact with the TIM1 (also known as HAVCR1) receptor of host cells, which enhances virus entry in cells as well as in brain tissue in vivo. Recombinant ZIKV mutants that lack ubiquitination are attenuated in human cells and in wild-type mice, but not in live mosquitoes. Monoclonal antibodies against K63-linked polyubiquitin specifically neutralize ZIKV and reduce viraemia in mice. Our results demonstrate that the ubiquitination of ZIKV E is an important determinant of virus entry, tropism and pathogenesis.


Subject(s)
Ubiquitination , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization , Zika Virus/metabolism , Zika Virus/pathogenicity , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Brain/metabolism , Cell Line , Culicidae/cytology , Culicidae/virology , Endosomes/metabolism , Female , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Male , Membrane Fusion , Mice , Organ Specificity , Polyubiquitin/immunology , Polyubiquitin/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Tropism , Viremia/immunology , Viremia/prevention & control , Viremia/virology , Virus Replication , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
11.
Cell Rep ; 27(11): 3269-3283.e6, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189110

ABSTRACT

Tripartite motif-containing protein 5α (TRIM5α) is a cellular antiviral restriction factor that prevents early events in retrovirus replication. The activity of TRIM5α is thought to be limited to retroviruses as a result of highly specific interactions with capsid lattices. In contrast to this current understanding, we show that both human and rhesus macaque TRIM5α suppress replication of specific flaviviruses. Multiple viruses in the tick-borne encephalitis complex are sensitive to TRIM5α-dependent restriction, but mosquito-borne flaviviruses, including yellow fever, dengue, and Zika viruses, are resistant. TRIM5α suppresses replication by binding to the viral protease NS2B/3 to promote its K48-linked ubiquitination and proteasomal degradation. Importantly, TRIM5α contributes to the antiviral function of IFN-I against sensitive flaviviruses in human cells. Thus, TRIM5α possesses remarkable plasticity in the recognition of diverse virus families, with the potential to influence human susceptibility to emerging flaviviruses of global concern.


Subject(s)
Flavivirus Infections/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Antiviral Restriction Factors , Cats , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Flavivirus/pathogenicity , Flavivirus/physiology , Flavivirus Infections/virology , HEK293 Cells , Humans , Protein Binding , Proteolysis , Substrate Specificity , Ubiquitination , Vero Cells
12.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29463617

ABSTRACT

The Chlamydia trachomatis plasmid and inclusion membrane protein CT135 are virulence factors in the pathogenesis of murine female genital tract infection. To determine if these virulence factors play a similar role in female nonhuman primates, we infected pig-tailed macaques with the same C. trachomatis strains shown to be important in the murine model. Wild-type C. trachomatis and its isogenic mutant strain deficient in both plasmid and CT135 were used to infect macaques. Macaques were given primary and repeated cervicovaginal challenges with the wild-type and mutant strains. The infection rate, infection duration, and antibody response were similar among macaques infected with both strains. Unexpectedly, colposcopy, laparoscopy, and histologic analysis revealed no substantial genital tract pathology following either primary or repeated cervicovaginal challenges. Cytokine analysis of cervicovaginal secretions from both challenged groups revealed low concentrations of interleukin 1ß (IL-1ß) and elevated levels of the interleukin 1 receptor agonist (IL-1RA). We propose that an imbalance of IL-1ß and IL-1RA in macaques is the reason for the mild inflammatory responses observed in infected urogenital tissues. Thus, understanding the pathobiology of chlamydial infection requires a better understanding of host epigenetic and chlamydial genetic factors. Our findings also have implications for understanding the high frequency of asymptomatic infections in humans.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Macaca/immunology , Plasmids/immunology , Reproductive Tract Infections/immunology , Virulence Factors/immunology , Animals , Female , Humans , Mice , Plasmids/genetics , Virulence Factors/genetics
13.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28461392

ABSTRACT

We studied infection and immunity of hysterectomized mice infected with Chlamydia muridarum and Chlamydia trachomatis to determine if there were differences between these species in their ability to infect vaginal squamous epithelial cells in vivo independently of proximal upper genital tract tissues. We found that C. muridarum readily colonized and infected vaginal squamous epithelial cells, whereas C. trachomatis did not. Primary infection of the vaginal epithelium with C. muridarum produced infections of a duration longer than that reported for normal mice. Infection resulted in an inflammatory response in the vagina characterized by neutrophils and infiltrating submucosal plasma cells consisting primarily of T cells. Despite the delayed clearance, rechallenged C. muridarum-infected mice were highly immune. Mice vaginally infected with C. muridarum produced serum and vaginal wash antibodies and an antigen-specific gamma interferon-dominated Th1-biased T cell response. By comparison, mice vaginally infected with C. trachomatis exhibited transient low-burden infections, produced no detectable tissue inflammatory response, and failed to seroconvert. We discuss how these marked differences in the biology of vaginal infection between these otherwise genetically similar species are possibly linked to pathogen-specific virulence genes and how they may influence pathology and immunity in the upper genital tract.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/growth & development , Chlamydia muridarum/immunology , Chlamydia trachomatis/growth & development , Hysterectomy , Vagina/microbiology , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Chlamydia Infections/immunology , Female , Interferon-gamma/metabolism , Mice, Inbred C57BL , T-Lymphocytes/immunology
14.
Cell Rep ; 19(7): 1406-1417, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514660

ABSTRACT

Chlamydia trachomatis is a human pathogen associated with significant morbidity worldwide. As obligate intracellular parasites, chlamydiae must survive within eukaryotic cells for sufficient time to complete their developmental cycle. To promote host cell survival, chlamydiae express poorly understood anti-apoptotic factors. Using recently developed genetic tools, we show that three inclusion membrane proteins (Incs) out of eleven examined are required for inclusion membrane stability and avoidance of host cell death pathways. In the absence of specific Incs, premature inclusion lysis results in recognition by autophagolysosomes, activation of intrinsic apoptosis, and premature termination of the chlamydial developmental cycle. Inhibition of autophagy or knockdown of STING prevented host cell death and activation of intrinsic apoptosis. Significantly, these findings emphasize the importance of Incs in the establishment of a replicative compartment that sequesters the pathogen from host surveillance systems.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Host-Pathogen Interactions , Inclusion Bodies/metabolism , Membrane Proteins/metabolism , Autophagosomes/metabolism , Autophagy , Cell Death , Chlamydia trachomatis/growth & development , HeLa Cells , Humans , Mutation/genetics , Protein Biosynthesis , Solubility , Transport Vesicles/metabolism
15.
Pathog Dis ; 75(3)2017 04 01.
Article in English | MEDLINE | ID: mdl-28369275

ABSTRACT

Chlamydia trachomatis is an obligate intracellular pathogen characterized by a unique biphasic developmental cycle that alternates between infectious and non-infectious organisms. Chlamydial ChxR is a transcriptional activator that has been implicated in the regulation of the development cycle. We used a reverse genetics approach to generate three chxR null mutants. All three mutants grew normally in cultured mammalian cells. Whole genome sequencing identified SNPs in other genes; however, none of the mutated genes were common to all three ChxR null mutants arguing against a genetic compensatory mechanism that would explain the non-essential in vitro growth phenotype. Comparative proteomics identified five proteins, CT005, CT214, CT565, CT694 and CT695, that were significantly downregulated in all ChxR null mutants. This group includes established inclusion membrane and type III secreted proteins. ChxR transcriptional regulation of these genes was confirmed by qRT-PCR. Importantly, while ChxR null mutants exhibited no growth deficiencies in in vitro, they did show significant differences in in vivo growth using a mouse genital tract model. Collectively, our findings demonstrated that ChxR is a transcriptional activator that regulates the expression of virulence genes whose functions are restricted to in vivo infection.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Host-Pathogen Interactions , Transcription Factors/metabolism , Alleles , Animals , Bacterial Proteins/genetics , Cell Line , Chlamydia trachomatis/genetics , Chlamydia trachomatis/growth & development , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation , Genome, Bacterial , Humans , Mice , Mutation , Sequence Analysis, DNA , Transcription Factors/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
16.
Cell Rep ; 18(3): 816-829, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099857

ABSTRACT

The unprecedented 2013-2016 outbreak of Ebola virus (EBOV) resulted in over 11,300 human deaths. Host resistance to RNA viruses requires RIG-I-like receptor (RLR) signaling through the adaptor protein, mitochondrial antiviral signaling protein (MAVS), but the role of RLR-MAVS in orchestrating anti-EBOV responses in vivo is not known. Here we apply a systems approach to MAVS-/- mice infected with either wild-type or mouse-adapted EBOV. MAVS controlled EBOV replication through the expression of IFNα, regulation of inflammatory responses in the spleen, and prevention of cell death in the liver, with macrophages implicated as a major cell type influencing host resistance. A dominant role for RLR signaling in macrophages was confirmed following conditional MAVS deletion in LysM+ myeloid cells. These findings reveal tissue-specific MAVS-dependent transcriptional pathways associated with resistance to EBOV, and they demonstrate that EBOV adaptation to cause disease in mice involves changes in two distinct events, RLR-MAVS antagonism and suppression of RLR-independent IFN-I responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/pathology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Disease Models, Animal , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/mortality , Humans , Interferon Type I/metabolism , Kaplan-Meier Estimate , Liver/metabolism , Liver/pathology , Liver/virology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/virology , Signal Transduction , Spleen/metabolism , Spleen/pathology , Spleen/virology , Virus Replication
17.
mBio ; 6(6): e01648-15, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26556273

ABSTRACT

UNLABELLED: Chlamydia trachomatis is an obligate intracellular bacterium that is a globally important human pathogen. The chlamydial plasmid is an attenuating virulence factor, but the molecular basis for attenuation is not understood. Chlamydiae replicate within a membrane-bound vacuole termed an inclusion, where they undergo a biphasic developmental growth cycle and differentiate from noninfectious into infectious organisms. Late in the developmental cycle, the fragile chlamydia-laden inclusion retains its integrity by surrounding itself with scaffolds of host cytoskeletal proteins. The ability of chlamydiae to developmentally free themselves from this cytoskeleton network is a fundamental virulence trait of the pathogen. Here, we show that plasmidless chlamydiae are incapable of disrupting their cytoskeletal entrapment and remain intracellular as stable mature inclusions that support high numbers of infectious organisms. By using deletion mutants of the eight plasmid-carried genes (Δpgp1 to Δpgp8), we show that Pgp4, a transcriptional regulator of multiple chromosomal genes, is required for exit. Exit of chlamydiae is dependent on protein synthesis and is inhibited by the compound C1, an inhibitor of the type III secretion system (T3S). Exit of plasmid-free and Δpgp4 organisms, which failed to lyse infected cells, was rescued by latrunculin B, an inhibitor of actin polymerization. Our findings describe a genetic mechanism of chlamydial exit from host cells that is dependent on an unknown pgp4-regulated chromosomal T3S effector gene. IMPORTANCE: Chlamydia's obligate intracellular life style requires both entry into and exit from host cells. Virulence factors that function in exiting are unknown. The chlamydial inclusion is stabilized late in the infection cycle by F-actin. A prerequisite of chlamydial exit is its ability to disassemble actin from the inclusion. We show that chlamydial plasmid-free organisms, and also a plasmid gene protein 4 (pgp4) null mutant, do not disassociate actin from the inclusion and fail to exit cells. We further provide evidence that Pgp4-regulated exit is dependent on the chlamydial type III secretion system. This study is the first to define a genetic mechanism that functions in chlamydial lytic exit from host cells. The findings also have practical implications for understanding why plasmid-free chlamydiae are highly attenuated and have the ability to elicit robust protective immune responses.


Subject(s)
Chlamydia trachomatis/physiology , Epithelial Cells/microbiology , Host-Pathogen Interactions , Plasmids , Vacuoles/microbiology , Actins/metabolism , Chlamydia trachomatis/genetics , Chlamydia trachomatis/growth & development , HeLa Cells , Humans , Virulence , Virulence Factors/metabolism
18.
Pathog Dis ; 73(6): ftv043, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26109550

ABSTRACT

Chlamydia trachomatis is an important human pathogen causing both ocular and sexually transmitted disease. Recently, we identified CT135 as an important virulence determinant in a mouse infection model. Results from CEL 1 digestion assays and sequencing analyses indicated that CT135 was much more polymorphic in high in vitro passage reference serovars than it was in clinical strains that had undergone limited passaging. Herein, we used targeted next-generation sequencing of the CT134-135 locus, from reference strains and clinical isolates, enabling accurate discovery of single nucleotide polymorphisms and other population genetic variations. Our results indicate that CT134 is stable in all C. trachomatis serovars examined. In contrast, CT135 is highly polymorphic in high-passaged reference ocular and non-LGV genital serovars, with the majority of the mutations resulting in gene disruption. In low-passaged ocular clinical isolates, CT135 was frequently disrupted, whereas in genital clinical isolates CT135 was intact in almost all instances. When a serovar K isolate, with an intact CT134 and CT135, was subjected to serial passage in vitro CT134 remained invariable, while numerous gene interrupting mutations rapidly accumulated in CT135. Collectively, our data indicate that, for genital serovars, CT135 is under strong positive selection in vivo, and negative selection in vitro.


Subject(s)
Chlamydia trachomatis/genetics , Genetic Variation , Genomic Instability , Virulence Factors/genetics , Animals , Chlamydia Infections/microbiology , Chlamydia trachomatis/isolation & purification , Chlamydia trachomatis/pathogenicity , DNA, Bacterial/genetics , Gene Deletion , High-Throughput Nucleotide Sequencing , Humans , Mice , Mutation , Selection, Genetic , Sequence Analysis, DNA , Serial Passage
19.
Infect Immun ; 83(2): 534-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25404022

ABSTRACT

Chlamydia trachomatis is an obligate intracellular epitheliotropic bacterial pathogen of humans. Infection of the eye can result in trachoma, the leading cause of preventable blindness in the world. The pathophysiology of blinding trachoma is driven by multiple episodes of reinfection of conjunctival epithelial cells, producing an intense chronic inflammatory response resulting in submucosal tissue remodeling and scarring. Recent reports have shown that infection with trachoma organisms lacking the cryptic chlamydial plasmid is highly attenuated in macaque eyes, a relevant experimental model of human trachoma infection. To better understand the molecular basis of plasmid-mediated infection attenuation and the potential modulation of host immunity, we conducted transcriptional profiling of human epithelial cells infected with C. trachomatis plasmid-bearing (A2497) and plasmid-deficient (A2497P(-)) organisms. Infection of human epithelial cells with either strain increased the expression of host genes coding for proinflammatory (granulocyte-macrophage colony-stimulating factor [GM-CSF], macrophage colony-stimulating factor [MCSF], interleukin-6 [IL-6], IL-8, IL-1α, CXCL1, CXCL2, CXCL3, intercellular adhesion molecule 1 [ICAM1]), chemoattraction (CCL20, CCL5, CXCL10), immune suppression (PD-L1, NFKB1B, TNFAIP3, CGB), apoptosis (CASP9, FAS, IL-24), and cell growth and fibrosis (EGR1 and IL-20) proteins. Statistically significant increases in the levels of expression of many of these genes were found in A2497-infected cells compared to the levels of expression in A2497P(-)-infected cells. Our findings suggest that the chlamydial plasmid plays a focal role in the host cell inflammatory response to infection and immune avoidance. These results provide new insights into the role of the chlamydial plasmid as a chlamydial virulence factor and its contributions to trachoma pathogenesis.


Subject(s)
Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Plasmids/genetics , Trachoma/pathology , Virulence Factors/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/immunology , Chlamydia trachomatis/pathogenicity , Cytokines/biosynthesis , Cytokines/genetics , Epithelial Cells , Gene Expression Profiling , Glycogen/metabolism , HeLa Cells , Humans , Inflammation/immunology , Inflammation/microbiology , Interleukins/biosynthesis , Interleukins/genetics , Trachoma/immunology , Trachoma/microbiology
20.
J Bacteriol ; 196(16): 2989-3001, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24914180

ABSTRACT

Chlamydiae are widespread Gram-negative pathogens of humans and animals. Salicylidene acylhydrazides, developed as inhibitors of type III secretion system (T3SS) in Yersinia spp., have an inhibitory effect on chlamydial infection. However, these inhibitors also have the capacity to chelate iron, and it is possible that their antichlamydial effects are caused by iron starvation. Therefore, we have explored the modification of salicylidene acylhydrazides with the goal to uncouple the antichlamydial effect from iron starvation. We discovered that benzylidene acylhydrazides, which cannot chelate iron, inhibit chlamydial growth. Biochemical and genetic analyses suggest that the derivative compounds inhibit chlamydiae through a T3SS-independent mechanism. Four single nucleotide polymorphisms were identified in a Chlamydia muridarum variant resistant to benzylidene acylhydrazides, but it may be necessary to segregate the mutations to differentiate their roles in the resistance phenotype. Benzylidene acylhydrazides are well tolerated by host cells and probiotic vaginal Lactobacillus species and are therefore of potential therapeutic value.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzylidene Compounds/pharmacology , Chlamydia muridarum/drug effects , Chlamydia muridarum/growth & development , DNA Mutational Analysis , Drug Resistance, Bacterial , Molecular Sequence Data , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...