Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Nature ; 585(7825): 414-419, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32641828

RESUMO

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.


Assuntos
Ubiquitinação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Zika virus/metabolismo , Zika virus/patogenicidade , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Encéfalo/metabolismo , Linhagem Celular , Culicidae/citologia , Culicidae/virologia , Endossomos/metabolismo , Feminino , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Humanos , Masculino , Fusão de Membrana , Camundongos , Especificidade de Órgãos , Poliubiquitina/imunologia , Poliubiquitina/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Tropismo Viral , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia , Replicação Viral , Zika virus/química , Zika virus/genética , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia
2.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34855915

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Vírus da Encefalite Transmitidos por Carrapatos/patogenicidade , Encefalite Transmitida por Carrapatos/virologia , Febres Hemorrágicas Virais/virologia , Macaca nemestrina , Animais , Chlorocebus aethiops , Citocinas/sangue , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/patologia , Feminino , Células HEK293 , Febres Hemorrágicas Virais/imunologia , Febres Hemorrágicas Virais/patologia , Humanos , Linfonodos/virologia , Células Vero , Viremia
3.
Infect Immun ; 86(5)2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29463617

RESUMO

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.


Assuntos
Infecções por Chlamydia/imunologia , Chlamydia trachomatis/genética , Chlamydia trachomatis/imunologia , Macaca/imunologia , Plasmídeos/imunologia , Infecções do Sistema Genital/imunologia , Fatores de Virulência/imunologia , Animais , Feminino , Humanos , Camundongos , Plasmídeos/genética , Fatores de Virulência/genética
4.
Infect Immun ; 85(7)2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28461392

RESUMO

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.


Assuntos
Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/patologia , Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia muridarum/imunologia , Chlamydia trachomatis/crescimento & desenvolvimento , Histerectomia , Vagina/microbiologia , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/sangue , Infecções por Chlamydia/imunologia , Feminino , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
5.
J Immunol ; 192(10): 4648-54, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24711617

RESUMO

Trachoma, caused by the obligate intracellular organism Chlamydia trachomatis, is the world's leading cause of preventable blindness for which a vaccine is needed. We have previously shown that a plasmid-deficient live-attenuated trachoma vaccine delivered ocularly to macaques elicited either solid or partial protective immunity against a virulent ocular challenge. Solidly protected macaques shared the same MHC class II alleles implicating CD4(+) T cells in superior protective immunity. Understandably, we sought to define T cell immune correlates in these animals to potentially improve vaccine efficacy. In this study, following a 2-y resting period, these macaques were boosted i.m. with the live-attenuated trachoma vaccine and their peripheral T cell anamnestic responses studied. Both solidly and partially protected macaques exhibited a CD4(+) and CD8(+) T cell anamnestic response following booster immunization. CD8(+) but not CD4(+) T cells from solidly protected macaques proliferated against soluble chlamydial Ag. We observed a more rapid T cell inflammatory cytokine response in tears of solidly protected animals following ocular rechallenge. Most notably, depletion of CD8(+) T cells in solidly protected macaques completely abrogated protective immunity. Collectively, our findings support the conclusion that CD8(+) T cells play an important but unexpected role in live-attenuated trachoma vaccine-mediated protective immunity.


Assuntos
Vacinas Bacterianas/farmacologia , Linfócitos T CD8-Positivos/imunologia , Chlamydia trachomatis/imunologia , Tracoma/prevenção & controle , Animais , Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Proliferação de Células/efeitos dos fármacos , Feminino , Macaca nemestrina , Masculino , Tracoma/imunologia , Tracoma/patologia , Vacinas Atenuadas/farmacologia
6.
Infect Immun ; 83(2): 534-43, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25404022

RESUMO

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.


Assuntos
Infecções por Chlamydia/patologia , Chlamydia trachomatis/genética , Plasmídeos/genética , Tracoma/patologia , Fatores de Virulência/genética , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/imunologia , Chlamydia trachomatis/patogenicidade , Citocinas/biossíntese , Citocinas/genética , Células Epiteliais , Perfilação da Expressão Gênica , Glicogênio/metabolismo , Células HeLa , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Interleucinas/biossíntese , Interleucinas/genética , Tracoma/imunologia , Tracoma/microbiologia
7.
J Bacteriol ; 196(16): 2989-3001, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24914180

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Compostos de Benzilideno/farmacologia , Chlamydia muridarum/efeitos dos fármacos , Chlamydia muridarum/crescimento & desenvolvimento , Análise Mutacional de DNA , Farmacorresistência Bacteriana , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA
8.
Infect Immun ; 82(7): 2756-62, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24733093

RESUMO

Chlamydia trachomatis is an obligate intracellular mucosotropic pathogen of significant medical importance. It is the etiological agent of blinding trachoma and bacterial sexually transmitted diseases, infections that afflict hundreds of millions of people globally. The C. trachomatis polymorphic membrane protein D (PmpD) is a highly conserved autotransporter and the target of broadly cross-reactive neutralizing antibodies; however, its role in host-pathogen interactions is unknown. Here we employed a targeted reverse genetics approach to generate a pmpD null mutant that was used to define the role of PmpD in the pathogenesis of chlamydial infection. We show that pmpD is not an essential chlamydial gene and the pmpD null mutant has no detectable deficiency in cultured murine cells or in a murine mucosal infection model. Notably, however, the pmpD null mutant was significantly attenuated for macaque eyes and cultured human cells. A reduction in pmpD null infection of human endocervical cells was associated with a deficiency in chlamydial attachment to cells. Collectively, our results show that PmpD is a chlamydial virulence factor that functions in early host-cell interactions. This study is the first of its kind using reverse genetics to evaluate the contribution of a C. trachomatis gene to disease pathogenesis.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Feminino , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Macaca fascicularis , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Mutação
9.
Pathogens ; 13(2)2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38392905

RESUMO

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.

10.
Infect Immun ; 81(3): 636-44, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319558

RESUMO

Chlamydia trachomatis causes chronic inflammatory diseases of the eye and genital tract and has global medical importance. The chlamydial plasmid plays an important role in the pathophysiology of these diseases, as plasmid-deficient organisms are highly attenuated. The cryptic plasmid carries noncoding RNAs and eight conserved open reading frames (ORFs). To understand plasmid gene function, we generated plasmid shuttle vectors with deletions in each of the eight ORFs. The individual deletion mutants were used to transform chlamydiae and the transformants were characterized phenotypically and at the transcriptional level. We show that pgp1, -2, -6, and -8 are essential for plasmid maintenance, while the other ORFs can be deleted and the plasmid stably maintained. We further show that a pgp4 knockout mutant exhibits an in vitro phenotype similar to its isogenic plasmidless strain, in terms of abnormal inclusion morphology and lack of glycogen accumulation. Microarray and qRT-PCR analysis revealed that Pgp4 is a transcriptional regulator of plasmid-encoded pgp3 and multiple chromosomal genes, including the glycogen synthase gene glgA, that are likely important in chlamydial virulence. Our findings have major implications for understanding the plasmid's role in chlamydial pathogenesis at the molecular level.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Plasmídeos/metabolismo , Transcrição Gênica/fisiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Cromossomos Bacterianos , Deleção de Genes , Camundongos , Plasmídeos/genética , Análise Serial de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Virulência
11.
Nat Commun ; 14(1): 4481, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37491352

RESUMO

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.


Assuntos
COVID-19 , Interferon Tipo I , Humanos , Camundongos , Animais , Citocinas , SARS-CoV-2 , Camundongos Transgênicos , Inflamação/genética , Modelos Animais de Doenças , Pulmão
12.
J Infect Dis ; 203(8): 1120-8, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21321103

RESUMO

Chlamydia muridarum and Chlamydia trachomatis mouse models of genital infection have been used to study chlamydial immunity and vaccine development. To assess the protective role of CD4(+) T cells in resolving C. trachomatis and C. muridarum genital tract infections, we used the female mouse model and evaluated infection in the presence and absence of CD4(+) T cells. In contrast to C. muridarum infection, C. trachomatis infection was unaltered in the absence of CD4(+) T cells. Mice infected with C. trachomatis developed protective immunity to re-challenge, but unlike C. muridarum infection, optimum resistance required multiple infectious challenges, despite the generation of adaptive serum and local chlamydial specific immune responses. Thus, understanding the chlamydial pathogenic and host immunologic factors that result in a diminished protective role for CD4(+) T cells in C. trachomatis murine infection might lead to new insights important to human immunity and vaccine development.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Infecções por Chlamydia/imunologia , Chlamydia trachomatis , Depleção Linfocítica , Imunidade Adaptativa , Animais , Chlamydia trachomatis/classificação , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C3H , Fatores de Tempo
13.
Life Sci Alliance ; 5(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35039442

RESUMO

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.


Assuntos
Envelhecimento/imunologia , COVID-19/imunologia , COVID-19/veterinária , SARS-CoV-2/imunologia , Doença Aguda , Animais , Formação de Anticorpos/imunologia , Líquido da Lavagem Broncoalveolar , COVID-19/complicações , COVID-19/genética , Citocinas/sangue , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genômica , Imunidade Celular/genética , Imunomodulação , Inflamação/complicações , Inflamação/patologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Tecido Linfoide/patologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Modelos Biológicos , Análise de Célula Única , Linfócitos T/imunologia , Transcrição Gênica
14.
Cell Rep ; 37(4): 109888, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34706234

RESUMO

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.


Assuntos
Proteínas com Domínio LIM/metabolismo , Mitofagia , Proteínas Quinases/metabolismo , Transdução de Sinais , Infecção por Zika virus/metabolismo , Zika virus/metabolismo , eIF-2 Quinase/metabolismo , Células A549 , Animais , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Knockout , Proteínas Quinases/genética , Células Vero , Zika virus/genética , Infecção por Zika virus/genética , eIF-2 Quinase/genética
15.
Sci Transl Med ; 13(578)2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33431511

RESUMO

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.


Assuntos
COVID-19/epidemiologia , COVID-19/virologia , Pulmão/virologia , SARS-CoV-2/fisiologia , Análise de Sequência de RNA , Análise de Célula Única , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Animais , Líquido da Lavagem Broncoalveolar/virologia , COVID-19/genética , Chlorocebus aethiops , DNA Viral/genética , Feminino , Genoma Viral/genética , Inflamação/patologia , Pulmão/patologia , Linfonodos/patologia , Macrófagos/patologia , Macrófagos/virologia , Masculino , Mediastino/patologia , Transcrição Gênica , Carga Viral , Replicação Viral
16.
Nat Commun ; 12(1): 5454, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526512

RESUMO

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.


Assuntos
Infecções por Chlamydia/imunologia , Chlamydia/imunologia , Inflamação/imunologia , Células Mieloides/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neutrófilos/imunologia , Receptores Purinérgicos P2X7/imunologia , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Chlamydia/fisiologia , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/microbiologia , Modelos Animais de Doenças , Feminino , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Células Mieloides/microbiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo
17.
Infect Immun ; 78(9): 3660-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20547745

RESUMO

Chlamydia trachomatis is a human pathogen of global importance. An obstacle to studying the pathophysiology of human chlamydial disease is the lack of a suitable murine model of C. trachomatis infection. Mice are less susceptible to infection with human isolates due in part to innate mouse-specific host defense mechanisms to which human strains are sensitive. Another possible factor that influences the susceptibility of mice to infection is that human isolates are commonly cultivated in vitro prior to infection of mice; therefore, virulence genes could be lost as a consequence of negative selective pressure. We tested this hypothesis by infecting innate immunity-deficient C3H/HeJ female mice intravaginally with a human serovar D urogenital isolate that had undergone multiple in vitro passages. We observed early and late infection clearance phenotypes. Strains of each phenotype were isolated and then used to reinfect naïve mice. Following infection, the late-clearance strain was significantly more virulent. It caused unvarying infections of much longer durations with greater infectious burdens that naturally ascended to the upper genital tract, causing salpingitis. Despite contrasting in vivo virulence characteristics, the strains exhibited no differences in the results of in vitro infectivity assays or sensitivities to gamma interferon. Genome sequencing of the strains revealed mutations that localized to a single gene (CT135), implicating it as a critical virulence factor. Mutations in CT135 were not unique to serovar D but were also found in multiple oculogenital reference strains. Our findings provide new information about the pathogenomics of chlamydial infection and insights for improving murine models of infection using human strains.


Assuntos
Infecções por Chlamydia/etiologia , Chlamydia trachomatis/patogenicidade , Mutação da Fase de Leitura , Doenças dos Genitais Femininos/etiologia , Fatores de Virulência/genética , Animais , Sequência de Bases , Infecções por Chlamydia/patologia , Chlamydia trachomatis/genética , Feminino , Doenças dos Genitais Femininos/patologia , Camundongos , Camundongos Endogâmicos C3H , Dados de Sequência Molecular , Fenótipo , Polimorfismo Genético
18.
bioRxiv ; 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32839775

RESUMO

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.

19.
Infect Immun ; 77(1): 508-16, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19001072

RESUMO

Chlamydia trachomatis is a globally important obligate intracellular bacterial pathogen that is a leading cause of sexually transmitted disease and blinding trachoma. Effective control of these diseases will likely require a preventative vaccine. C. trachomatis polymorphic membrane protein D (PmpD) is an attractive vaccine candidate as it is conserved among C. trachomatis strains and is a target of broadly cross-reactive neutralizing antibodies. We show here that immunoaffinity-purified native PmpD exists as an oligomer with a distinct 23-nm flower-like structure. Two-dimensional blue native-sodium dodecyl sulfate-polyacrylamide gel electrophoresis analyses showed that the oligomers were composed of full-length PmpD (p155) and two proteolytically processed fragments, the p73 passenger domain (PD) and the p82 translocator domain. We also show that PmpD undergoes an infection-dependent proteolytic processing step late in the growth cycle that yields a soluble extended PD (p111) that was processed into a p73 PD and a novel p30 fragment. Interestingly, soluble PmpD peptides possess putative eukaryote-interacting functional motifs, implying potential secondary functions within or distal to infected cells. Collectively, our findings show that PmpD exists as two distinct forms, a surface-associated oligomer exhibiting a higher-order flower-like structure and a soluble form restricted to infected cells. We hypothesize that PmpD is a multifunctional virulence factor important in chlamydial pathogenesis and could represent novel vaccine or drug targets for the control of human chlamydial infections.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Chlamydia trachomatis/química , Chlamydia trachomatis/metabolismo , Multimerização Proteica , Proteínas da Membrana Bacteriana Externa/ultraestrutura , Eletroforese em Gel de Poliacrilamida/métodos , Células Epiteliais/química , Células Epiteliais/microbiologia , Células HeLa , Humanos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Peptídeo Hidrolases/química , Peptídeo Hidrolases/metabolismo
20.
Cell Rep ; 27(11): 3269-3283.e6, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189110

RESUMO

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.


Assuntos
Infecções por Flavivirus/metabolismo , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Fatores de Restrição Antivirais , Gatos , Chlorocebus aethiops , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Flavivirus/patogenicidade , Flavivirus/fisiologia , Infecções por Flavivirus/virologia , Células HEK293 , Humanos , Ligação Proteica , Proteólise , Especificidade por Substrato , Ubiquitinação , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA