Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nature ; 585(7825): 414-419, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32641828

RESUMEN

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.


Asunto(s)
Ubiquitinación , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Virus Zika/metabolismo , Virus Zika/patogenicidad , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Encéfalo/metabolismo , Línea Celular , Culicidae/citología , Culicidae/virología , Endosomas/metabolismo , Femenino , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Masculino , Fusión de Membrana , Ratones , Especificidad de Órganos , Poliubiquitina/inmunología , Poliubiquitina/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Tropismo Viral , Viremia/inmunología , Viremia/prevención & control , Viremia/virología , Replicación Viral , Virus Zika/química , Virus Zika/genética , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/virología
2.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34855915

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Encefalitis Transmitida por Garrapatas/virología , Fiebres Hemorrágicas Virales/virología , Macaca nemestrina , Animales , Chlorocebus aethiops , Citocinas/sangre , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/patología , Femenino , Células HEK293 , Fiebres Hemorrágicas Virales/inmunología , Fiebres Hemorrágicas Virales/patología , Humanos , Ganglios Linfáticos/virología , Células Vero , Viremia
3.
Infect Immun ; 86(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29463617

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/genética , Chlamydia trachomatis/inmunología , Macaca/inmunología , Plásmidos/inmunología , Infecciones del Sistema Genital/inmunología , Factores de Virulencia/inmunología , Animales , Femenino , Humanos , Ratones , Plásmidos/genética , Factores de Virulencia/genética
4.
Infect Immun ; 85(7)2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28461392

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/crecimiento & desarrollo , Chlamydia muridarum/inmunología , Chlamydia trachomatis/crecimiento & desarrollo , Histerectomía , Vagina/microbiología , Animales , Anticuerpos Antibacterianos/análisis , Anticuerpos Antibacterianos/sangre , Infecciones por Chlamydia/inmunología , Femenino , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Linfocitos T/inmunología
5.
J Immunol ; 192(10): 4648-54, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24711617

RESUMEN

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.


Asunto(s)
Vacunas Bacterianas/farmacología , Linfocitos T CD8-positivos/inmunología , Chlamydia trachomatis/inmunología , Tracoma/prevención & control , Animales , Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Proliferación Celular/efectos de los fármacos , Femenino , Macaca nemestrina , Masculino , Tracoma/inmunología , Tracoma/patología , Vacunas Atenuadas/farmacología
6.
Infect Immun ; 83(2): 534-43, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25404022

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/patología , Chlamydia trachomatis/genética , Plásmidos/genética , Tracoma/patología , Factores de Virulencia/genética , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/inmunología , Chlamydia trachomatis/patogenicidad , Citocinas/biosíntesis , Citocinas/genética , Células Epiteliales , Perfilación de la Expresión Génica , Glucógeno/metabolismo , Células HeLa , Humanos , Inflamación/inmunología , Inflamación/microbiología , Interleucinas/biosíntesis , Interleucinas/genética , Tracoma/inmunología , Tracoma/microbiología
7.
J Infect Dis ; 210(8): 1239-47, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24755433

RESUMEN

BACKGROUND: Sexually transmitted infections (STIs) are associated with an increased risk of human immunodeficiency virus (HIV) infection, but their biological effect on HIV susceptibility is not fully understood. METHODS: Female pig-tailed macaques inoculated with Chlamydia trachomatis and Trichomonas vaginalis (n = 9) or medium (controls; n = 7) were repeatedly challenged intravaginally with SHIVSF162p3. Virus levels were evaluated by real-time polymerase chain reaction, plasma and genital cytokine levels by Luminex assays, and STI clinical signs by colposcopy. RESULTS: Simian/HIV (SHIV) susceptibility was enhanced in STI-positive macaques (P = .04, by the log-rank test; relative risk, 2.5 [95% confidence interval, 1.1-5.6]). All STI-positive macaques were SHIV infected, whereas 3 controls (43%) remained uninfected. Moreover, relative to STI-negative animals, SHIV infections occurred earlier in the menstrual cycle in STI-positive macaques (P = .01, by the Wilcoxon test). Levels of inflammatory cytokines (interferon γ, interleukin 6, and granulocyte colony-stimulating factor [G-CSF]) were higher in STI-positive macaques during STI inoculation and SHIV exposure periods (P ≤ .05, by the Wilcoxon test). CONCLUSIONS: C. trachomatis and T. vaginalis infection increase the susceptibility to SHIV, likely because of prolonged genital tract inflammation. These novel data demonstrate a biological link between these nonulcerative STIs and the risk of SHIV infection, supporting epidemiological associations of HIV and STIs. This study establishes a macaque model for studies of high-risk HIV transmission and prevention.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis , Coinfección/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Vaginitis por Trichomonas/complicaciones , Trichomonas vaginalis , Animales , Cuello del Útero/microbiología , Cuello del Útero/parasitología , Cuello del Útero/patología , Colposcopía , Femenino , Macaca nemestrina , Factores de Riesgo , Enfermedades de Transmisión Sexual/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
8.
J Bacteriol ; 196(16): 2989-3001, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24914180

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Bencilideno/farmacología , Chlamydia muridarum/efectos de los fármacos , Chlamydia muridarum/crecimiento & desarrollo , Análisis Mutacional de ADN , Farmacorresistencia Bacteriana , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
9.
Infect Immun ; 82(7): 2756-62, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24733093

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Línea Celular , Femenino , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Macaca fascicularis , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C3H , Mutación
10.
J Med Primatol ; 43(3): 135-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24460742

RESUMEN

BACKGROUND: Rectal sexually transmitted infections (STIs) may increase HIV susceptibility in men who have sex with men (MSM), and Chlamydia trachomatis is prevalent among HIV-positive MSM. To study STIs and HIV infection in MSM, we first evaluated whether cynomolgus macaques can sustain both C. trachomatis and SHIVSF162p3 infections. METHODS: Four SHIVSF162p3 -positive male cynomolgus macaques were used (n = 3 rectally inoculated with 10(6) IFU; n = 1 control). Systemic and rectal SHIV RNA levels and cytokines were measured by real-time PCR and Luminex assays, respectively. RESULTS: Macaques were successfully Chlamydia infected. Rectal SHIV shedding (P = 0.02 χ(2) ) and levels of G-CSF, IL-1ra, IL-6, IL-8, IFN-γ, and TNF-α (P ≤ 0.01, Mann-Whitney) in rectal secretions increased following infection. CONCLUSIONS: These pilot data successfully demonstrate rectal C. trachomatis-SHIV coinfection in cynomolgus macaques and suggest the feasibility of a rectal C. trachomatis model for SHIV susceptibility and biomedical prevention studies in the context of rectal STIs.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Modelos Animales de Enfermedad , Infecciones por VIH/complicaciones , Enfermedades de Transmisión Sexual/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Animales , Infecciones por Chlamydia/sangre , Infecciones por Chlamydia/patología , Chlamydia trachomatis , Coinfección , Citocinas/sangre , Citocinas/orina , Infecciones por VIH/sangre , Infecciones por VIH/virología , Macaca fascicularis , Masculino , Proyectos Piloto , ARN Viral/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Recto/microbiología , Recto/patología , Enfermedades de Transmisión Sexual/sangre , Enfermedades de Transmisión Sexual/microbiología , Enfermedades de Transmisión Sexual/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Virus de la Inmunodeficiencia de los Simios , Esparcimiento de Virus
11.
Pathogens ; 13(2)2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38392905

RESUMEN

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.

12.
Infect Immun ; 81(3): 636-44, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319558

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Chlamydia trachomatis/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Plásmidos/metabolismo , Transcripción Genética/fisiología , Animales , Proteínas Bacterianas/genética , Línea Celular , Chlamydia trachomatis/citología , Chlamydia trachomatis/genética , Cromosomas Bacterianos , Eliminación de Gen , Ratones , Plásmidos/genética , Análisis por Matrices de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virulencia
13.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35233576

RESUMEN

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.

14.
Nat Commun ; 14(1): 4481, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37491352

RESUMEN

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.


Asunto(s)
COVID-19 , Interferón Tipo I , Humanos , Ratones , Animales , Citocinas , SARS-CoV-2 , Ratones Transgénicos , Inflamación/genética , Modelos Animales de Enfermedad , Pulmón
15.
J Infect Dis ; 203(8): 1120-8, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21321103

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Infecciones por Chlamydia/inmunología , Chlamydia trachomatis , Depleción Linfocítica , Inmunidad Adaptativa , Animales , Chlamydia trachomatis/clasificación , Femenino , Humanos , Ratones , Ratones Endogámicos C3H , Factores de Tiempo
16.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35039442

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , COVID-19/inmunología , COVID-19/veterinaria , SARS-CoV-2/inmunología , Enfermedad Aguda , Animales , Formación de Anticuerpos/inmunología , Líquido del Lavado Bronquioalveolar , COVID-19/complicaciones , COVID-19/genética , Citocinas/sangre , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica , Inmunidad Celular/genética , Inmunomodulación , Inflamación/complicaciones , Inflamación/patología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Tejido Linfoide/patología , Macaca mulatta/inmunología , Macaca mulatta/virología , Modelos Biológicos , Análisis de la Célula Individual , Linfocitos T/inmunología , Transcripción Genética
17.
J Med Primatol ; 40(4): 214-23, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21781129

RESUMEN

BACKGROUND: Sexually transmitted infections (STIs) are associated with an increased risk of HIV infection. To model the interaction between STIs and HIV infection, we evaluated the capacity of the pigtail macaque model to sustain triple infection with Trichomonas vaginalis, Chlamydia trachomatis, and SHIV(SF162P3). METHODS: Seven SHIV(SF162P3) -infected pigtail macaques were inoculated with T. vaginalis only (n = 2), C. trachomatis only (n = 1), both T. vaginalis and C. trachomatis (n = 2), or control media (no STI; n = 2). Infections were confirmed by culture and/or nucleic acid testing. Genital mucosa was visualized by colposcopy. RESULTS: Characteristic gynecologic signs were observed for both STIs, but not in control animals. Manifestations were most prominent at days 7-10 post-infection. STIs persisted between 4 and 6 weeks and were cleared with antibiotics. CONCLUSIONS: These pilot studies demonstrate the first successful STI-SHIV triple infection of pigtail macaques, with clinical presentation of genital STI symptoms similar to those observed in humans.


Asunto(s)
Infecciones por Chlamydia/patología , Modelos Animales de Enfermedad , Infecciones por VIH/complicaciones , Enfermedades de Transmisión Sexual/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Vaginitis por Trichomonas/patología , Animales , Cuello del Útero/microbiología , Cuello del Útero/parasitología , Cuello del Útero/patología , Infecciones por Chlamydia/sangre , Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis , Colposcopía , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/virología , Macaca nemestrina , Proyectos Piloto , Enfermedades de Transmisión Sexual/sangre , Enfermedades de Transmisión Sexual/microbiología , Enfermedades de Transmisión Sexual/parasitología , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Virus de la Inmunodeficiencia de los Simios , Vaginitis por Trichomonas/sangre , Vaginitis por Trichomonas/complicaciones , Trichomonas vaginalis , Enfermedades del Cuello del Útero/sangre , Enfermedades del Cuello del Útero/complicaciones , Enfermedades del Cuello del Útero/microbiología , Enfermedades del Cuello del Útero/parasitología , Vagina/microbiología , Vagina/parasitología , Vagina/patología
18.
Sci Transl Med ; 13(578)2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33431511

RESUMEN

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.


Asunto(s)
COVID-19/epidemiología , COVID-19/virología , Pulmón/virología , SARS-CoV-2/fisiología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Animales , Líquido del Lavado Bronquioalveolar/virología , COVID-19/genética , Chlorocebus aethiops , ADN Viral/genética , Femenino , Genoma Viral/genética , Inflamación/patología , Pulmón/patología , Ganglios Linfáticos/patología , Macrófagos/patología , Macrófagos/virología , Masculino , Mediastino/patología , Transcripción Genética , Carga Viral , Replicación Viral
19.
Cell Rep ; 37(4): 109888, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34706234

RESUMEN

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.


Asunto(s)
Proteínas con Dominio LIM/metabolismo , Mitofagia , Proteínas Quinasas/metabolismo , Transducción de Señal , Infección por el Virus Zika/metabolismo , Virus Zika/metabolismo , eIF-2 Quinasa/metabolismo , Células A549 , Animales , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Proteínas con Dominio LIM/genética , Ratones , Ratones Noqueados , Proteínas Quinasas/genética , Células Vero , Virus Zika/genética , Infección por el Virus Zika/genética , eIF-2 Quinasa/genética
20.
Nat Commun ; 12(1): 5454, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526512

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neutrófilos/inmunología , Receptores Purinérgicos P2X7/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Chlamydia/fisiología , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/microbiología , Modelos Animales de Enfermedad , Femenino , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/microbiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiología , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA