Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Virol ; : e0079424, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38940558

RESUMO

Coronavirus disease 2019 (COVID-19) has claimed millions of lives since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and lung disease appears the primary cause of death in COVID-19 patients. However, the underlying mechanisms of COVID-19 pathogenesis remain elusive, and there is no existing model where human disease can be faithfully recapitulated and conditions for the infection process can be experimentally controlled. Herein we report the establishment of an ex vivo human precision-cut lung slice (hPCLS) platform for studying SARS-CoV-2 pathogenicity and innate immune responses, and for evaluating the efficacy of antiviral drugs against SARS-CoV-2. We show that while SARS-CoV-2 continued to replicate during the course of infection of hPCLS, infectious virus production peaked within 2 days, and rapidly declined thereafter. Although most proinflammatory cytokines examined were induced by SARS-CoV-2 infection, the degree of induction and types of cytokines varied significantly among hPCLS from individual donors. Two cytokines in particular, IP-10 and IL-8, were highly and consistently induced, suggesting a role in the pathogenesis of COVID-19. Histopathological examination revealed focal cytopathic effects late in the infection. Transcriptomic and proteomic analyses identified molecular signatures and cellular pathways that are largely consistent with the progression of COVID-19 in patients. Furthermore, we show that homoharringtonine, a natural plant alkaloid derived from Cephalotoxus fortunei, not only inhibited virus replication but also production of pro-inflammatory cytokines, and thus ameliorated the histopathological changes caused by SARS-CoV-2 infection, demonstrating the usefulness of the hPCLS platform for evaluating antiviral drugs. IMPORTANCE: Here, established an ex vivo human precision-cut lung slice platform for assessing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, viral replication kinetics, innate immune response, disease progression, and antiviral drugs. Using this platform, we identified early induction of specific cytokines, especially IP-10 and IL-8, as potential predictors for severe coronavirus disease 2019 (COVID-19), and uncovered a hitherto unrecognized phenomenon that while infectious virus disappears at late times of infection, viral RNA persists and lung histopathology commences. This finding may have important clinical implications for both acute and post-acute sequelae of COVID-19. This platform recapitulates some of the characteristics of lung disease observed in severe COVID-19 patients and is therefore a useful platform for understanding mechanisms of SARS-CoV-2 pathogenesis and for evaluating the efficacy of antiviral drugs.

2.
Infect Immun ; 91(7): e0013123, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37338372

RESUMO

Inhalation of respiratory droplets infected with Yersinia pestis results in a rapidly progressing and lethal necrotic pneumonia called primary pneumonic plague. Disease manifests as biphasic, with an initial preinflammatory phase with rapid bacterial replication in the lungs absent readily detectable host immune responses. This is followed by the onset of a proinflammatory phase that sees the dramatic upregulation of proinflammatory cytokines and extensive neutrophil accumulation in the lungs. The plasminogen activator protease (Pla) is an essential virulence factor that is responsible for survival of Y. pestis in the lungs. Our lab recently showed that Pla functions as an adhesin that promotes binding to alveolar macrophages to facilitate translocation of effector proteins called Yops into the cytosol of target host cells via a type 3 secretion system (T3SS). Loss of Pla-mediated adherence disrupted the preinflammatory phase of disease and resulted in early neutrophil migration to the lungs. While it is established that Yersinia broadly suppresses host innate immune responses, it is not clear precisely which signals need to be inhibited to establish a preinflammatory stage of infection. Here, we show that early Pla-mediated suppression of Interleukin-17 (IL-17) expression in alveolar macrophages and pulmonary neutrophils limits neutrophil migration to the lungs and aids in establishing a preinflammatory phase of disease. In addition, IL-17 ultimately contributes to neutrophil migration to the airways that defines the later proinflammatory phase of infection. These results suggest that the pattern of IL-17 expression contributes to the progression of primary pneumonic plague.


Assuntos
Peste , Yersinia pestis , Animais , Camundongos , Interleucina-17/genética , Interleucina-17/metabolismo , Infiltração de Neutrófilos , Pulmão/microbiologia , Yersinia pestis/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
3.
Antimicrob Agents Chemother ; 66(1): e0127521, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34780267

RESUMO

Severe and late-stage pneumonias are often difficult to treat with antibiotics alone due to overwhelming host inflammatory responses mounted to clear infection. These host responses contribute to pulmonary damage leading to acute lung injury, acute respiratory distress syndrome, and death. In order to effectively treat severe and late-stage pneumonias, use of adjunctive therapies must be considered to reduce pulmonary damage when antimicrobial agents can be administered. Pneumonic plague, a severe pneumonia caused by inhalation of Yersinia pestis, is a fatal disease that causes death within 6 days without antibiotic intervention. Late-stage pneumonic plague is difficult to treat, as antibiotics must be delivered within 24 h after onset of symptoms to be effective. Here, we use a murine model of primary pneumonic plague to examine how host inflammatory responses impact antibiotic treatment of late-stage pneumonic plague. We developed a murine infection model demonstrating the poor outcomes associated with delayed delivery of antibiotics. We show that pretreatment of mice with intranasal fluticasone propionate increased the efficacy of delayed antibiotic delivery and enhanced murine survival. Mice receiving fluticasone propionate also showed decreased bacterial burden and reduced inflammatory pathology in the lungs. Further, we show that treatment and survival correlated with decreased levels of interleukin-6 (IL-6) and reduced neutrophil infiltration to the lungs. This work demonstrates how host inflammatory responses complicate treatment of late-stage pneumonic plague and suggests that targeting of host inflammatory responses may improve treatment of severe, late-stage pneumonia.


Assuntos
Peste , Yersinia pestis , Animais , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Fluticasona/uso terapêutico , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Peste/tratamento farmacológico , Peste/microbiologia
4.
Infect Immun ; 89(2)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33257531

RESUMO

Yersinia pestis is a highly virulent pathogen and the causative agent of bubonic, septicemic, and pneumonic plague. Primary pneumonic plague caused by inhalation of respiratory droplets contaminated with Y. pestis is nearly 100% lethal within 4 to 7 days without antibiotic intervention. Pneumonic plague progresses in two phases, beginning with extensive bacterial replication in the lung with minimal host responsiveness, followed by the abrupt onset of a lethal proinflammatory response. The precise mechanisms by which Y. pestis is able to colonize the lung and survive two very distinct disease phases remain largely unknown. To date, a few bacterial virulence factors, including the Ysc type 3 secretion system, are known to contribute to the pathogenesis of primary pneumonic plague. The bacterial GTPase BipA has been shown to regulate expression of virulence factors in a number of Gram-negative bacteria, including Pseudomonas aeruginosa, Escherichia coli, and Salmonella enterica serovar Typhi. However, the role of BipA in Y. pestis has yet to be investigated. Here, we show that BipA is a Y. pestis virulence factor that promotes defense against early neutrophil-mediated bacterial killing in the lung. This work identifies a novel Y. pestis virulence factor and highlights the importance of early bacterial/neutrophil interactions in the lung during primary pneumonic plague.


Assuntos
Proteínas de Bactérias/fisiologia , GTP Fosfo-Hidrolases/fisiologia , Peste/imunologia , Peste/fisiopatologia , Fatores de Virulência/fisiologia , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
5.
J Infect Dis ; 222(3): 407-416, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32128567

RESUMO

Early after inhalation, Yersinia pestis replicates to high numbers in the airways in the absence of disease symptoms or notable inflammatory responses to cause primary pneumonic plague. The plasminogen activator protease (Pla) is a critical Y. pestis virulence factor that is important for early bacterial growth in the lung via an unknown mechanism. In this article, we define a dual role for Pla in the initial stages of pulmonary infection. We show that Pla functions as an adhesin independent of its proteolytic function to suppress early neutrophil influx into the lungs, and that Pla enzymatic activity contributes to bacterial resistance to neutrophil-mediated bacterial killing. Our results suggest that the fate of Y. pestis infection of the lung is decided extremely early during infection and that Pla plays a dual role to tilt the balance in favor of the pathogen.


Assuntos
Interações Hospedeiro-Patógeno , Pulmão/microbiologia , Peste/etiologia , Ativadores de Plasminogênio/fisiologia , Yersinia pestis/metabolismo , Animais , Aderência Bacteriana , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Virulência , Yersinia pestis/patogenicidade
6.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31085709

RESUMO

Pneumonic plague is the deadliest form of disease caused by Yersinia pestis Key to the progression of infection is the activity of the plasminogen activator protease Pla. Deletion of Pla results in a decreased Y. pestis bacterial burden in the lung and failure to progress into the lethal proinflammatory phase of disease. While a number of putative functions have been attributed to Pla, its precise role in the pathogenesis of pneumonic plague is yet to be defined. Here, we show that Pla facilitates type 3 secretion into primary alveolar macrophages but not into the commonly used THP-1 cell line. We also establish human precision-cut lung slices as a platform for modeling early host/pathogen interactions during pneumonic plague and solidify the role of Pla in promoting optimal type 3 secretion using primary human tissue with relevant host cell heterogeneity. These results position Pla as a key player in the early host/pathogen interactions that define pneumonic plague and showcase the utility of human precision-cut lung slices as a platform to evaluate pulmonary infection by bacterial pathogens.


Assuntos
Interações Hospedeiro-Patógeno , Pulmão/microbiologia , Peste/etiologia , Ativadores de Plasminogênio/fisiologia , Yersinia pestis/metabolismo , Animais , Aderência Bacteriana , Linhagem Celular , Citocinas/metabolismo , Feminino , Humanos , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
7.
PLoS Pathog ; 11(3): e1004688, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25781467

RESUMO

Pneumonic plague is the most rapid and lethal form of Yersinia pestis infection. Increasing evidence suggests that Y. pestis employs multiple levels of innate immune evasion and/or suppression to produce an early "pre-inflammatory" phase of pulmonary infection, after which the disease is highly inflammatory in the lung and 100% fatal. In this study, we show that IL-1ß/IL-18 cytokine activation occurs early after bacteria enter the lung, and this activation eventually contributes to pulmonary inflammation and pathology during the later stages of infection. However, the inflammatory effects of IL-1ß/IL-1-receptor ligation are not observed during this first stage of pneumonic plague. We show that Y. pestis also activates the induction of IL-1 receptor antagonist (IL-1RA), and this activation likely contributes to the ability of Y. pestis to establish the initial pre-inflammatory phase of disease.


Assuntos
Interleucina-1beta/metabolismo , Peste/imunologia , Pneumonia/microbiologia , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Yersinia pestis/imunologia , Animais , Humanos , Camundongos , Pneumonia/patologia , Receptores de Interleucina-1/antagonistas & inibidores
8.
PLoS Pathog ; 9(10): e1003679, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098126

RESUMO

Inhalation of Yersinia pestis causes primary pneumonic plague, a highly lethal syndrome with mortality rates approaching 100%. Pneumonic plague progression is biphasic, with an initial pre-inflammatory phase facilitating bacterial growth in the absence of host inflammation, followed by a pro-inflammatory phase marked by extensive neutrophil influx, an inflammatory cytokine storm, and severe tissue destruction. Using a FRET-based probe to quantitate injection of effector proteins by the Y. pestis type III secretion system, we show that these bacteria target alveolar macrophages early during infection of mice, followed by a switch in host cell preference to neutrophils. We also demonstrate that neutrophil influx is unable to limit bacterial growth in the lung and is ultimately responsible for the severe inflammation during the lethal pro-inflammatory phase.


Assuntos
Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Peste/imunologia , Pneumonia Bacteriana/imunologia , Yersinia pestis/imunologia , Animais , Feminino , Inflamação/imunologia , Inflamação/patologia , Camundongos , Neutrófilos/patologia , Peste/patologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia
9.
Biomedicines ; 12(3)2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38540156

RESUMO

Recent epidemiological studies suggest that individuals with Down syndrome are more susceptible to SARS-CoV-2 infection and have higher rates of hospitalization and mortality than the general population. However, the main drivers behind these disparate health outcomes remain unknown. Herein, we performed experimental infections with SARS-CoV-2 in a well-established mouse model of Down syndrome. We observed similar SARS-CoV-2 replication kinetics and dissemination in the primary and secondary organs between mice with and without Down syndrome, suggesting that both groups have similar susceptibilities to SARS-CoV-2 infection. However, Down syndrome mice exhibited more severe disease as defined by clinical features including symptoms, weight loss, pulmonary function, and survival of mice. We found that increased disease severity in Down syndrome mice could not be attributed solely to increased infectivity or a more dramatic pro-inflammatory response to infection. Rather, results from RNA sequencing suggested that differences in the expression of genes from other physiological pathways, such as deficient oxidative phosphorylation, cardiopulmonary dysfunction, and deficient mucociliary clearance in the lungs may also contribute to heightened disease severity and mortality in Down syndrome mice following SARS-CoV-2 infection.

10.
bioRxiv ; 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37131640

RESUMO

COVID-19 has claimed millions of lives since the emergence of SARS-CoV-2, and lung disease appears the primary cause of the death in COVID-19 patients. However, the underlying mechanisms of COVID-19 pathogenesis remain elusive, and there is no existing model where the human disease can be faithfully recapitulated and conditions for the infection process can be experimentally controlled. Herein we report the establishment of an ex vivo human precision-cut lung slice (hPCLS) platform for studying SARS-CoV-2 pathogenicity and innate immune responses, and for evaluating the efficacy of antiviral drugs against SARS-CoV-2. We show that while SARS-CoV-2 continued to replicate during the course of infection of hPCLS, infectious virus production peaked within 2 days, and rapidly declined thereafter. Although most proinflammatory cytokines examined were induced by SARS-CoV-2 infection, the degree of induction and types of cytokines varied significantly among hPCLS from individual donors, reflecting the heterogeneity of human populations. In particular, two cytokines (IP-10 and IL-8) were highly and consistently induced, suggesting a role in the pathogenesis of COVID-19. Histopathological examination revealed focal cytopathic effects late in the infection. Transcriptomic and proteomic analyses identified molecular signatures and cellular pathways that are largely consistent with the progression of COVID-19 in patients. Furthermore, we show that homoharringtonine, a natural plant alkaloid derived from Cephalotoxus fortunei , not only inhibited virus replication but also production of pro-inflammatory cytokines, and ameliorated the histopathological changes of the lungs caused by SARS-CoV-2 infection, demonstrating the usefulness of the hPCLS platform for evaluating antiviral drugs. SIGNIFICANCE: Here we established an ex vivo human precision-cut lung slice platform for assessing SARS-CoV-2 infection, viral replication kinetics, innate immune response, disease progression, and antiviral drugs. Using this platform, we identified early induction of specific cytokines, especially IP-10 and IL-8, as potential predictors for severe COVID-19, and uncovered a hitherto unrecognized phenomenon that while infectious virus disappears at late times of infection, viral RNA persists and lung histopathology commences. This finding may have important clinical implications for both acute and post-acute sequelae of COVID-19. This platform recapitulates some of the characteristics of lung disease observed in severe COVID-19 patients and is therefore a useful platform for understanding mechanisms of SARS-CoV-2 pathogenesis and for evaluating the efficacy of antiviral drugs.

12.
bioRxiv ; 2021 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-33469587

RESUMO

SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) hospitalizations and deaths disportionally affect males and the elderly. Here we investigated the impact of male sex and age by infecting adult male, aged male, and adult female ferrets with SARS-CoV-2. Aged male ferrets had a decrease in temperature which was accompanied by prolonged viral replication with increased pathology in the upper respiratory tract after infection. Transcriptome analysis of the nasal turbinates and lungs indicated that female ferrets had significant increases in interferon response genes (OASL, MX1, ISG15, etc.) on day 2 post infection which was delayed in aged males. In addition, genes associated with taste and smell such as RTP1, CHGA, and CHGA1 at later time points were upregulated in males but not in females. These results provide insight into COVID-19 and suggests that older males may play a role in viral transmission due to decreased antiviral responses.

13.
Sci Rep ; 11(1): 14536, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34267262

RESUMO

SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) hospitalizations and deaths disportionally affect males and older ages. Here we investigated the impact of male sex and age comparing sex-matched or age-matched ferrets infected with SARS-CoV-2. Differences in temperature regulation was identified for male ferrets which was accompanied by prolonged viral replication in the upper respiratory tract after infection. Gene expression analysis of the nasal turbinates indicated that 1-year-old female ferrets had significant increases in interferon response genes post infection which were delayed in males. These results provide insight into COVID-19 and suggests that older males may play a role in viral transmission due to decreased antiviral responses.


Assuntos
COVID-19/virologia , Furões/virologia , Interferons/metabolismo , Fatores Etários , Animais , Anticorpos Antivirais , COVID-19/metabolismo , Modelos Animais de Doenças , Feminino , Furões/metabolismo , Interações entre Hospedeiro e Microrganismos , Interferons/genética , Masculino , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/fisiologia , Fatores Sexuais , Carga Viral , Replicação Viral
14.
Methods Mol Biol ; 2010: 17-28, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31177428

RESUMO

Pneumonic plague is a rapidly progressing and highly lethal pneumonia caused by pulmonary infection with Yersinia pestis. Disease is marked by the rapid replication of bacteria in the lungs in the absence of symptoms, followed by the abrupt onset of a highly lethal inflammatory response. A murine intranasal infection model has been key to characterizing the progression of disease. Mice are a natural Y. pestis host, and murine disease closely mirrors what is seen during human infection. Intranasal inoculation of mice with fully virulent Y. pestis strains allows for the detailed analysis of key bacterial and host factors that define disease progression. In this chapter I describe a method for intranasal inoculation of mice with Y. pestis, as well as techniques for processing lung tissue for analysis. These include protocols for isolating whole lungs and lavage fluid for measure of bacterial burden, transcriptomics, cytokine/chemokine expression, and flow cytometry. These techniques can be used to evaluate disease parameters of interest during typical infection, infection with bacterial mutants, or infection in the presence of pharmacological agents aimed at targeting specific host or bacterial factors. Combining a highly relevant murine infection model with these techniques provides a powerful platform for fully evaluating the progression of pneumonic plague.


Assuntos
Modelos Animais de Doenças , Pulmão/microbiologia , Peste/patologia , Pneumonia/patologia , Yersinia pestis/fisiologia , Animais , Líquido da Lavagem Broncoalveolar/microbiologia , Feminino , Citometria de Fluxo/métodos , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Peste/microbiologia , Pneumonia/microbiologia
15.
Artigo em Inglês | MEDLINE | ID: mdl-28507954

RESUMO

Pneumonia is a leading cause of death from infection in the United States and across the globe. During pulmonary infection, clear resolution of host inflammatory responses occurs in the absence of appreciable lung damage. Neutrophils are the first wave of leukocytes to arrive in the lung upon infection. After activation, neutrophils traffic from the vasculature via transendothelial migration through the lung interstitium and into the alveolar space. Successful pulmonary immunity requires neutrophil-mediated killing of invading pathogens by phagocytosis and release of a myriad of antimicrobial molecules, followed by resolution of inflammation, neutrophil apoptosis, and clearing of dead or dying neutrophils by macrophages. In addition to their antimicrobial role, it is becoming clear that neutrophils are also important modulators of innate and adaptive immune responses, primarily through the release of cytokines and recruitment of additional waves of neutrophils into the airways. Though typically essential to combating severe pneumonia, neutrophil influx into the airways is a double-edged sword: Overzealous neutrophil activation can cause severe tissue damage as a result of the release of toxic agents including proteases, cationic polypeptides, cytokines, and reactive oxygen species (ROS) aimed at killing invading microbes. In extreme cases, the damage caused by neutrophils and other innate immune mediators become the primary source of morbidity and mortality. Here, we review the complex role of neutrophils during severe pneumonia by highlighting specific molecules and processes that contribute to pulmonary immunity, but can also drive progression of severe disease. Depending on the identity of the infectious agent, enhancing or suppressing neutrophil-mediated responses may be key to effectively treating severe and typically lethal pneumonia.


Assuntos
Progressão da Doença , Neutrófilos/imunologia , Neutrófilos/microbiologia , Neutrófilos/fisiologia , Pneumonia/imunologia , Peptídeos Catiônicos Antimicrobianos/imunologia , Movimento Celular/imunologia , Citocinas/metabolismo , Humanos , Imunidade Inata , Inflamação/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Lesão Pulmonar/imunologia , Ativação de Neutrófilo/imunologia , Fagocitose , Infarto Pulmonar/imunologia , Infarto Pulmonar/microbiologia , Espécies Reativas de Oxigênio/imunologia , Serina Proteases/imunologia
16.
Trends Microbiol ; 24(3): 190-197, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26698952

RESUMO

Inhalation of the bacterium Yersinia pestis results in primary pneumonic plague. Pneumonic plague is the most severe manifestation of plague, with mortality rates approaching 100% in the absence of treatment. Its rapid disease progression, lethality, and ability to be transmitted via aerosol have compounded fears of the intentional release of Y. pestis as a biological weapon. Importantly, recent epidemics of plague have highlighted a significant role for pneumonic plague during outbreaks of Y. pestis infections. In this review we describe the characteristics of pneumonic plague, focusing on its disease progression and pathogenesis. The rapid time-course, severity, and difficulty of treating pneumonic plague highlight how differences in the route of disease transmission can enhance the lethality of an already deadly pathogen.


Assuntos
Peste/microbiologia , Peste/fisiopatologia , Yersinia pestis/patogenicidade , Animais , Armas Biológicas , Progressão da Doença , Humanos , Pulmão/microbiologia , Pulmão/patologia , Peste/terapia , Peste/transmissão , Virulência , Yersinia pestis/crescimento & desenvolvimento
17.
mBio ; 6(1): e02302-14, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25691593

RESUMO

UNLABELLED: Inhalation of Yersinia pestis results in primary pneumonic plague, a highly lethal and rapidly progressing necrotizing pneumonia. The disease begins with a period of extensive bacterial replication in the absence of disease symptoms, followed by the sudden onset of inflammatory responses that ultimately prove fatal. Very little is known about the bacterial and host factors that contribute to the rapid biphasic progression of pneumonic plague. In this work, we analyzed the in vivo transcription kinetics of 288 bacterial open reading frames previously shown by microarray analysis to be dynamically regulated in the lung. Using this approach combined with bacterial genetics, we were able to identify five Y. pestis genes that contribute to the development of pneumonic plague. Deletion of one of these genes, ybtX, did not alter bacterial survival but attenuated host inflammatory responses during late-stage disease. Deletion of ybtX in another lethal respiratory pathogen, Klebsiella pneumoniae, also resulted in diminished host inflammation during infection. Thus, our in vivo transcriptional screen has identified an important inflammatory mediator that is common to two Gram-negative bacterial pathogens that cause severe pneumonia. IMPORTANCE: Yersinia pestis is responsible for at least three major pandemics, most notably the Black Death of the Middle Ages. Due to its pandemic potential, ease of dissemination by aerosolization, and a history of its weaponization, Y. pestis is categorized by the Centers for Disease Control and Prevention as a tier 1 select agent most likely to be used as a biological weapon. To date, there is no licensed vaccine against Y. pestis. Importantly, an early "silent" phase followed by the rapid onset of nondescript influenza-like symptoms makes timely treatment of pneumonic plague difficult. A more detailed understanding of the bacterial and host factors that contribute to pathogenesis is essential to understanding the progression of pneumonic plague and developing or enhancing treatment options.


Assuntos
Perfilação da Expressão Gênica , Fatores de Virulência/biossíntese , Yersinia pestis/genética , Yersinia pestis/patogenicidade , Deleção de Genes , Interações Hospedeiro-Patógeno , Inflamação/imunologia , Inflamação/patologia , Peste/microbiologia , Peste/patologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/patologia , Transcrição Gênica , Fatores de Virulência/genética , Yersinia pestis/imunologia
18.
mBio ; 6(5): e01530-15, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26463167

RESUMO

UNLABELLED: During pneumonic plague, the bacterium Yersinia pestis elicits the development of inflammatory lung lesions that continue to expand throughout infection. This lesion development and persistence are poorly understood. Here, we examine spatially distinct regions of lung lesions using laser capture microdissection and transcriptome sequencing (RNA-seq) analysis to identify transcriptional differences between lesion microenvironments. We show that cellular pathways involved in leukocyte migration and apoptosis are downregulated in the center of lung lesions compared to the periphery. Probing for the bacterial factor(s) important for the alteration in neutrophil survival, we show both in vitro and in vivo that Y. pestis increases neutrophil survival in a manner that is dependent on the type III secretion system effector YopM. This research explores the complexity of spatially distinct host-microbe interactions and emphasizes the importance of cell relevance in assays in order to fully understand Y. pestis virulence. IMPORTANCE: Yersinia pestis is a high-priority pathogen and continues to cause outbreaks worldwide. The ability of Y. pestis to be transmitted via respiratory droplets and its history of weaponization has led to its classification as a select agent most likely to be used as a biological weapon. Unrestricted bacterial growth during the initial preinflammatory phase primes patients to be infectious once disease symptoms begin in the proinflammatory phase, and the rapid disease progression can lead to death before Y. pestis infection can be diagnosed and treated. Using in vivo analyses and focusing on relevant cell types during pneumonic plague infection, we can identify host pathways that may be manipulated to extend the treatment window for pneumonic plague patients.


Assuntos
Pulmão/patologia , Neutrófilos/imunologia , Peste/patologia , Yersinia pestis/imunologia , Animais , Apoptose , Proteínas da Membrana Bacteriana Externa/metabolismo , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Histocitoquímica , Humanos , Microdissecção e Captura a Laser , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neutrófilos/fisiologia
19.
Microbiol Mol Biol Rev ; 73(4): 684-711, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19946137

RESUMO

Francisella tularensis is a facultative intracellular gram-negative pathogen and the etiological agent of the zoonotic disease tularemia. Recent advances in the field of Francisella genetics have led to a rapid increase in both the generation and subsequent characterization of mutant strains exhibiting altered growth and/or virulence characteristics within various model systems of infection. In this review, we summarize the major properties of several Francisella species, including F. tularensis and F. novicida, and provide an up-to-date synopsis of the genes necessary for pathogenesis by these organisms and the determinants that are currently being targeted for vaccine development.


Assuntos
Vacinas Bacterianas/uso terapêutico , Francisella tularensis , Tularemia , Animais , Francisella tularensis/patogenicidade , Francisella tularensis/fisiologia , Genes Bacterianos , Interações Hospedeiro-Patógeno , Humanos , Tularemia/imunologia , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinação/tendências , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
20.
PLoS One ; 3(6): e2487, 2008 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-18575611

RESUMO

BACKGROUND: Francisella tularensis is a gram-negative coccobacillus that causes the febrile illness tularemia. Subspecies that are pathogenic for humans include those comprising the type A (subspecies tularensis) or type B (subspecies holarctica) biovars. An attenuated live vaccine strain (LVS) developed from a type B isolate has previously been used to vaccinate at-risk individuals, but offers limited protection against high dose (>1000 CFUs) challenge with type A strains delivered by the respiratory route. Due to differences between type A and type B F. tularensis strains at the genetic level, it has been speculated that utilization of an attenuated type A strain as a live vaccine might offer better protection against homologous respiratory challenge compared with LVS. Here, we report the construction and characterization of an unmarked Delta purMCD mutant in the highly virulent type A strain Schu S4. METHODOLOGY/PRINCIPAL FINDINGS: Growth of Schu S4 Delta purMCD was severely attenuated in primary human peripheral blood monocyte-derived macrophages and in the A549 human lung epithelial cell line. The Schu S4 Delta purMCD mutant was also highly attenuated in mice when delivered via either the intranasal or intradermal infection route. Mice vaccinated intranasally with Schu S4 Delta purMCD were well protected against high dose intradermal challenge with virulent type A or type B strains of F. tularensis. However, intranasal vaccination with Schu S4 Delta purMCD induced tissue damage in the lungs, and conferred only limited protection against high dose Schu S4 challenge delivered by the same route. The level of protection observed was similar to that conferred following vaccination with wild-type LVS or the analogous LVS Delta purMCD mutant. CONCLUSIONS/SIGNIFICANCE: Collectively, these results argue that development of the next generation live attenuated vaccine for Francisella should be based on use of the less pathogenic type B biovar rather than the more reactogenic type A biovar.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Purinas/imunologia , Administração Intranasal , Animais , Vacinas Bacterianas/administração & dosagem , Linhagem Celular , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Humanos , Pulmão/microbiologia , Macrófagos/microbiologia , Camundongos , Mutação , Virulência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa