Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Sci Rep ; 12(1): 15755, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36130991

RESUMEN

COVID-19 has impacted millions of patients across the world. Molecular testing occurring now identifies the presence of the virus at the sampling site: nasopharynx, nares, or oral cavity. RNA sequencing has the potential to establish both the presence of the virus and define the host's response in COVID-19. Single center, prospective study of patients with COVID-19 admitted to the intensive care unit where deep RNA sequencing (> 100 million reads) of peripheral blood with computational biology analysis was done. All patients had positive SARS-CoV-2 PCR. Clinical data was prospectively collected. We enrolled fifteen patients at a single hospital. Patients were critically ill with a mortality of 47% and 67% were on a ventilator. All the patients had the SARS-CoV-2 RNA identified in the blood in addition to RNA from other viruses, bacteria, and archaea. The expression of many immune modulating genes, including PD-L1 and PD-L2, were significantly different in patients who died from COVID-19. Some proteins were influenced by alternative transcription and splicing events, as seen in HLA-C, HLA-E, NRP1 and NRP2. Entropy calculated from alternative RNA splicing and transcription start/end predicted mortality in these patients. Current upper respiratory tract testing for COVID-19 only determines if the virus is present. Deep RNA sequencing with appropriate computational biology may provide important prognostic information and point to therapeutic foci to be precisely targeted in future studies.


Asunto(s)
COVID-19 , Antígeno B7-H1/genética , Prueba de COVID-19 , Antígenos HLA-C/genética , Humanos , Unidades de Cuidados Intensivos , Estudios Prospectivos , ARN Viral/genética , SARS-CoV-2/genética , Análisis de Secuencia de ARN
2.
Immun Ageing ; 19(1): 27, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650631

RESUMEN

BACKGROUND: Influenza causes a serious infection in older individuals who are at the highest risk for mortality from this virus. Changes in the immune system with age are well known. This study used transcriptomic analysis to evaluate how aging specifically affects the functional host response to influenza in the lung. Adult (12-16 weeks) and aged (72-76 weeks) mice were infected with influenza and lungs were processed for RNA analysis. RESULTS: Older mice demonstrated a delayed anti-viral response on the level of transcription compared to adults, similar to the immunologic responses measured in prior work. The transcriptional differences, however, were evident days before observable differences in the protein responses described previously. The transcriptome response to influenza in aged mice was dominated by immunoglobulin genes and B cell markers compared to adult animals, suggesting immune dysregulation. Despite these differences, both groups of mice had highly similar transcriptional responses involving non-immune genes one day after inoculation and T cell genes during resolution. CONCLUSIONS: These results define a delayed and dysregulated immune response in the lungs of aged mice infected with influenza. The findings implicate B cells and immunoglobulins as markers or mechanisms of immune aging. In addition to discovering new therapeutic targets, the findings underscore the value of transcription studies and network analysis to characterize complex biological processes, and serve as a model to analyze the susceptibility of the elderly to infectious agents.

4.
Front Mol Biosci ; 9: 1080964, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36589229

RESUMEN

Variants of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) continue to cause disease and impair the effectiveness of treatments. The therapeutic potential of convergent neutralizing antibodies (NAbs) from fully recovered patients has been explored in several early stages of novel drugs. Here, we identified initially elicited NAbs (Ig Heavy, Ig lambda, Ig kappa) in response to COVID-19 infection in patients admitted to the intensive care unit at a single center with deep RNA sequencing (>100 million reads) of peripheral blood as a diagnostic tool for predicting the severity of the disease and as a means to pinpoint specific compensatory NAb treatments. Clinical data were prospectively collected at multiple time points during ICU admission, and amino acid sequences for the NAb CDR3 segments were identified. Patients who survived severe COVID-19 had significantly more of a Class 3 antibody (C135) to SARS-CoV-2 compared to non-survivors (15059.4 vs. 1412.7, p = 0.016). In addition to highlighting the utility of RNA sequencing in revealing unique NAb profiles in COVID-19 patients with different outcomes, we provided a physical basis for our findings via atomistic modeling combined with molecular dynamics simulations. We established the interactions of the Class 3 NAb C135 with the SARS-CoV-2 spike protein, proposing a mechanistic basis for inhibition via multiple conformations that can effectively prevent ACE2 from binding to the spike protein, despite C135 not directly blocking the ACE2 binding motif. Overall, we demonstrate that deep RNA sequencing combined with structural modeling offers the new potential to identify and understand novel therapeutic(s) NAbs in individuals lacking certain immune responses due to their poor endogenous production. Our results suggest a possible window of opportunity for administration of such NAbs when their full sequence becomes available. A method involving rapid deep RNA sequencing of patients infected with SARS-CoV-2 or its variants at the earliest infection time could help to develop personalized treatments using the identified specific NAbs.

5.
medRxiv ; 2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33469603

RESUMEN

PURPOSE: COVID-19 has impacted millions of patients across the world. Molecular testing occurring now identifies the presence of the virus at the sampling site: nasopharynx, nares, or oral cavity. RNA sequencing has the potential to establish both the presence of the virus and define the host's response in COVID-19. METHODS: Single center, prospective study of patients with COVID-19 admitted to the intensive care unit where deep RNA sequencing (>100 million reads) of peripheral blood with computational biology analysis was done. All patients had positive SARS-CoV-2 PCR. Clinical data was prospectively collected. RESULTS: We enrolled fifteen patients at a single hospital. Patients were critically ill with a mortality of 47% and 67% were on a ventilator. All the patients had the SARS-CoV-2 RNA identified in the blood in addition to RNA from other viruses, bacteria, and archaea. The expression of many immune modulating genes, including PD-L1 and PD-L2, were significantly different in patients who died from COVID-19. Some proteins were influenced by alternative transcription and splicing events, as seen in HLA-C, HLA-E, NRP1 and NRP2. Entropy calculated from alternative RNA splicing and transcription start/end predicted mortality in these patients. CONCLUSIONS: Current upper respiratory tract testing for COVID-19 only determines if the virus is present. Deep RNA sequencing with appropriate computational biology may provide important prognostic information and point to therapeutic foci to be precisely targeted in future studies. TAKE HOME MESSAGE: Deep RNA sequencing provides a novel diagnostic tool for critically ill patients. Among ICU patients with COVID-19, RNA sequencings can identify gene expression, pathogens (including SARS-CoV-2), and can predict mortality. TWEET: Deep RNA sequencing is a novel technology that can assist in the care of critically ill COVID-19 patients & can be applied to other disease.

6.
Transpl Infect Dis ; 22(5): e13328, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32416005

RESUMEN

Ureaplasma species (spp.) are common colonizers of the urogenital tract but may cause systemic infection in immunocompromised patients. They release significant amounts of ammonia via urea hydrolysis and have been recently implicated in the pathogenesis of hyperammonemia syndrome after organ transplantation. We describe a unique case of hyperammonemia syndrome after kidney transplant caused by U urealyticum infection, and the first, to our knowledge, case of a fluoroquinolone-resistant Ureaplasma strain causing hyperammonemia syndrome. A 17-year-old female developed intermittent fevers, rising creatinine, sterile pyuria and debilitating polyarthritis approximately 1 year after kidney transplant. Serum ammonia level was elevated, and urine PCR was positive for U urealyticum. Near the end of treatment with levofloxacin, she had rebound hyperammonemia, which preceded clinical relapse of polyarthritis and encephalopathy. Blood and urine PCR and synovial fluid culture were positive for U urealyticum. Susceptibility testing showed fluoroquinolone resistance, but she responded well to azithromycin and doxycycline. The frequency of Ureaplasma spp. infection in immunocompromised patients is probably underestimated due to diagnostic challenges. Ammonia levels were helpful biomarkers of response to antimicrobial therapy in our case. Susceptibility testing of clinical isolates should be pursued. In serious Ureaplasma spp. infections, particularly in immunocompromised patients, two empiric antibiotics may be indicated given the potential for antimicrobial resistance.


Asunto(s)
Hiperamonemia , Trasplante de Riñón , Adolescente , Antibacterianos/uso terapéutico , Femenino , Fluoroquinolonas , Humanos , Hiperamonemia/tratamiento farmacológico , Ureaplasma , Ureaplasma urealyticum
7.
Virulence ; 10(1): 643-656, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31314675

RESUMEN

Pathogens enhance their survival during infections by manipulating host defenses. Francisella tularensis evades innate immune responses, which we have found to be dependent on an understudied gene ybeX (FTL_0883/FTT_0615c). To understand the function of YbeX, we sought protein interactors in F. tularensis subsp. holarctica live vaccine strain (LVS). An unstudied Francisella protein co-immunoprecipitated with recombinant YbeX, which is a predicted glycosyltransferase with a DXD-motif. There are up to four genomic copies of this gene with identical sequence in strains of F. tularensis pathogenic to humans, despite ongoing genome decay. Disruption mutations were generated by intron insertion into all three copies of this glycosyltransferase domain containing gene in LVS, gdcA1-3. The resulting strains stimulated more cytokines from macrophages in vitro than wild-type LVS and were attenuated in two in vivo infection models. GdcA was released from LVS during culture and was sufficient to block NF-κB activation when expressed in eukaryotic cells. When co-expressed in zebrafish, GdcA and YbeX were synergistically lethal to embryo development. Glycosyltransferases with DXD-motifs are found in a variety of pathogens including NleB, an Escherichia coli type-III secretion system effector that inhibits NF-κB by antagonizing death receptor signaling. To our knowledge, GdcA is the first DXD-motif glycosyltransferase that inhibits NF-κB in immune cells. Together, these findings suggest DXD-motif glycosyltransferases may be a conserved virulence mechanism used by pathogenic bacteria to remodel host defenses.


Asunto(s)
Proteínas Bacterianas/inmunología , Francisella tularensis/enzimología , Glicosiltransferasas/inmunología , Interacciones Huésped-Patógeno , Animales , Proteínas Bacterianas/genética , Citocinas , Femenino , Francisella tularensis/genética , Glicosiltransferasas/genética , Humanos , Inmunidad Innata , Células Jurkat , Macrófagos/microbiología , Ratones Endogámicos C57BL , Mariposas Nocturnas , Mutación , Tularemia/inmunología , Tularemia/microbiología , Virulencia , Pez Cebra
8.
Clin Infect Dis ; 69(1): 1-11, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-30715222

RESUMEN

BACKGROUND: Infections caused by antibiotic-resistant bacteria, including carbapenem-resistant Enterobacteriaceae, have increased in frequency, resulting in significant patient morbidity and mortality. The Infectious Diseases Society of America continues to propose legislative, regulatory, and funding solutions to address this escalating crisis. This report updates the status of development and approval of systemic antibiotics in the United States as of late 2018. METHODS: We performed a review of the published literature and on-line clinical trials registry at www.clinicaltrials.gov to identify new systemically acting orally and/or intravenously administered antibiotic drug candidates in the development pipeline, as well as agents approved by the US Food and Drug Administration since 2012. RESULTS: Since our 2013 pipeline status report, the number of new antibiotics annually approved for marketing in the United States has reversed its previous decline, likely influenced by new financial incentives and increased regulatory flexibility. Although our survey demonstrates progress in development of new antibacterial drugs that target infections caused by resistant bacterial pathogens, the majority of recently approved agents have been modifications of existing chemical classes of antibiotics, rather than new chemical classes. Furthermore, larger pharmaceutical companies continue to abandon the field, and smaller companies face financial difficulties as a consequence. CONCLUSIONS: Unfortunately, if 20 × '20 is achieved due to efforts embarked upon in decades past, it could mark the apex of antibiotic drug development for years to come. Without increased regulatory, governmental, industry, and scientific support and collaboration, durable solutions to the clinical, regulatory, and economic problems posed by bacterial multidrug resistance will not be found.


Asunto(s)
Antibacterianos/uso terapéutico , Aprobación de Drogas/estadística & datos numéricos , United States Food and Drug Administration , Aprobación de Drogas/organización & administración , Descubrimiento de Drogas , Farmacorresistencia Bacteriana Múltiple , Sociedades Médicas , Estados Unidos
9.
IDCases ; 12: 161-164, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942780

RESUMEN

Acute otitis media affects 700 million people each year with children being disproportionately affected relative to adults. Group A streptococcus is a pathogen implicated in a broad array of human pathology. It is, however, a rare cause of acute otitis media and neuroinvasive disease in older adults with only 2-3 cases occurring per year in the United States. We describe two such cases from a single institution in Rhode Island in 2017. The clinical presentation, neuroimaging and management are reviewed. The mechanism of intracranial spread may have involved dehiscence of the bony tegmen of the roof of the middle ear cavity.

10.
Artículo en Inglés | MEDLINE | ID: mdl-28652232

RESUMEN

Francisella tularensis is a highly infectious Gram-negative intracellular pathogen that causes tularemia. Because of its potential as a bioterrorism agent, there is a need for new therapeutic agents. We therefore developed a whole-animal Caenorhabditis elegans-F. tularensis pathosystem for high-throughput screening to identify and characterize potential therapeutic compounds. We found that the C. elegans p38 mitogen-activate protein (MAP) kinase cascade is involved in the immune response to F. tularensis, and we developed a robust F. tularensis-mediated C. elegans killing assay with a Z' factor consistently of >0.5, which was then utilized to screen a library of FDA-approved compounds that included 1,760 small molecules. In addition to clinically used antibiotics, five FDA-approved drugs were also identified as potential hits, including the anti-inflammatory drug diflunisal that showed anti-F. tularensis activity in vitro Moreover, the nonsteroidal anti-inflammatory drug (NSAID) diflunisal, at 4× MIC, blocked the replication of an F. tularensis live vaccine strain (LVS) in primary human macrophages and nonphagocytic cells. Diflunisal was nontoxic to human erythrocytes and HepG2 human liver cells at concentrations of ≥32 µg/ml. Finally, diflunisal exhibited synergetic activity with the antibiotic ciprofloxacin in both a checkerboard assay and a macrophage infection assay. In conclusion, the liquid C. elegans-F. tularensis LVS assay described here allows screening for anti-F. tularensis compounds and suggests that diflunisal could potentially be repurposed for the management of tularemia.


Asunto(s)
Antibacterianos/farmacología , Antiinflamatorios/farmacología , Caenorhabditis elegans/efectos de los fármacos , Francisella tularensis/efectos de los fármacos , Animales , Vacunas Bacterianas/inmunología , Caenorhabditis elegans/inmunología , Línea Celular Tumoral , Ciprofloxacina/farmacología , Eritrocitos/microbiología , Francisella tularensis/inmunología , Células Hep G2 , Humanos , Hígado/microbiología , Macrófagos/microbiología , Vacunas Atenuadas/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Clin Infect Dis ; 62(12): 1552-1555, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27045122

RESUMEN

We investigated the duration of Ebola virus (EBOV) RNA and infectious EBOV in semen specimens of 5 Ebola virus disease (EVD) survivors. EBOV RNA and infectious EBOV was detected by real-time RT-PCR and virus culture out to 290 days and 70 days, respectively, after EVD onset.


Asunto(s)
Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/virología , Semen/virología , Adulto , Estudios de Cohortes , Ebolavirus/patogenicidad , Humanos , Masculino , Sobrevivientes
12.
Nucleic Acids Res ; 43(3): e20, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25428368

RESUMEN

Identifying conserved and divergent response patterns in gene networks is becoming increasingly important. A common approach is integrating expression information with gene association networks in order to find groups of connected genes that are activated or repressed. In many cases, researchers are also interested in comparisons across species (or conditions). Finding an active sub-network is a hard problem and applying it across species requires further considerations (e.g. orthology information, expression data and networks from different sources). To address these challenges we devised ModuleBlast, which uses both expression and network topology to search for highly relevant sub-networks. We have applied ModuleBlast to expression and interaction data from mouse, macaque and human to study immune response and aging. The immune response analysis identified several relevant modules, consistent with recent findings on apoptosis and NFκB activation following infection. Temporal analysis of these data revealed cascades of modules that are dynamically activated within and across species. We have experimentally validated some of the novel hypotheses resulting from the analysis of the ModuleBlast results leading to new insights into the mechanisms used by a key mammalian aging protein.


Asunto(s)
Redes Reguladoras de Genes , Envejecimiento/genética , Animales , Apoptosis , Humanos , Macaca , Ratones , Especificidad de la Especie
13.
Ann Am Thorac Soc ; 11 Suppl 4: S201-10, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25148426

RESUMEN

Pulmonary diseases and infections are among the top contributors to human morbidity and mortality worldwide, and despite the successful history of vaccines and antimicrobial therapeutics, infectious disease still presents a significant threat to human health. Effective vaccines are frequently unavailable in developing countries, and successful vaccines have yet to be developed for major global maladies, such as tuberculosis. Furthermore, antibiotic resistance poses a growing threat to human health. The "Challenges and Future in Vaccines, Drug Development, and Immunomodulatory Therapy" session of the 2013 Pittsburgh International Lung Conference highlighted several recent and current studies related to treatment and prevention of antibiotic-resistant bacterial infections, highly pathogenic influenza, respiratory syncytial virus, and tuberculosis. Research presented here focused on novel antimicrobial therapies, new vaccines that are either in development or currently in clinical trials, and the potential for immunomodulatory therapies. These studies are making important contributions to the areas of microbiology, virology, and immunology related to pulmonary diseases and infections and are paving the way for improvements in the efficacy of vaccines and antimicrobials.


Asunto(s)
Diseño de Fármacos , Factores Inmunológicos/uso terapéutico , Inmunomodulación , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Vacunas/normas , Humanos
14.
BMC Microbiol ; 14: 84, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24708478

RESUMEN

BACKGROUND: The absence of rapid tests evaluating antibiotic susceptibility results in the empirical prescription of antibiotics. This can lead to treatment failures due to escalating antibiotic resistance, and also furthers the emergence of drug-resistant bacteria. This study reports a rapid optical method to detect ß-lactamase and thereby assess activity of ß-lactam antibiotics, which could provide an approach for targeted prescription of antibiotics. The methodology is centred on a fluorescence quenching based probe (ß-LEAF--ß-Lactamase Enzyme Activated Fluorophore) that mimics the structure of ß-lactam antibiotics. RESULTS: The ß-LEAF assay was performed for rapid determination of ß-lactamase production and activity of ß-lactam antibiotic (cefazolin) on a panel of Staphylococcus aureus ATCC strains and clinical isolates. Four of the clinical isolates were determined to be lactamase producers, with the capacity to inactivate cefazolin, out of the twenty-five isolates tested. These results were compared against gold standard methods, nitrocefin disk test for ß-lactamase detection and disk diffusion for antibiotic susceptibility, showing results to be largely consistent. Furthermore, in the sub-set of ß-lactamase producers, it was demonstrated and validated that multiple antibiotics (cefazolin, cefoxitin, cefepime) could be assessed simultaneously to predict the antibiotic that would be most active for a given bacterial isolate. CONCLUSIONS: The study establishes the rapid ß-LEAF assay for ß-lactamase detection and prediction of antibiotic activity using S. aureus clinical isolates. Although the focus in the current study is ß-lactamase-based resistance, the overall approach represents a broad diagnostic platform. In the long-term, these studies form the basis for the development of assays utilizing a broader variety of targets, pathogens and drugs.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/enzimología , beta-Lactamasas/análisis , beta-Lactamas/farmacología , Cefazolina/farmacología , Fluorometría/métodos , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación
15.
Microbes Infect ; 16(4): 300-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24374051

RESUMEN

Biosynthesis and acquisition of nutrients during infection are integral to pathogenesis. Members of a metabolic pathway, the glycine cleavage system, have been identified in virulence screens of the intracellular bacterium Francisella tularensis but their role in pathogenesis remains unknown. This system generates 5,10-methylenetetrahydrofolate, a precursor of amino acid and DNA synthesis, from glycine degradation. To characterize this pathway, deletion of the gcvT homolog, an essential member of this system, was performed in attenuated and virulent F. tularensis strains. Deletion mutants were auxotrophic for serine but behaved similar to wild-type strains with respect to host cell invasion, intracellular replication, and stimulation of TNF-α. Unexpectedly, the glycine cleavage system was required for the pathogenesis of virulent F. tularensis in a murine model. Deletion of the gcvT homolog delayed mortality and lowered bacterial burden, particularly in the liver and bloodstream. To reconcile differences between the cell culture model and animal model, minimal tissue culture media was employed to mimic the nutritionally limiting environment of the host. This reevaluation demonstrated that the glycine cleavage system contributes to the intracellular replication of virulent F. tularensis in serine limiting environments. Thus, the glycine cleavage system is the serine biosynthetic pathway of F. tularensis and contributes to pathogenesis in vivo.


Asunto(s)
Aminoácido Oxidorreductasas , Proteínas Portadoras , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidad , Complejos Multienzimáticos , Transferasas , Tularemia/microbiología , Tularemia/patología , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Francisella tularensis/genética , Eliminación de Gen , Ratones Endogámicos C57BL , Tetrahidrofolatos/metabolismo , Virulencia
16.
Artículo en Inglés | MEDLINE | ID: mdl-24367766

RESUMEN

The highly infectious and deadly pathogen, Francisella tularensis, is classified by the CDC as a Category A bioterrorism agent. Inhalation of a single bacterium results in an acute pneumonia with a 30-60% mortality rate without treatment. Due to the prevalence of antibiotic resistance, there is a strong need for new types of antibacterial drugs. Resazurin is commonly used to measure bacterial and eukaryotic cell viability through its reduction to the fluorescent product resorufin. When tested on various bacterial taxa at the recommended concentration of 44 µM, a potent bactericidal effect was observed against various Francisella and Neisseria species, including the human pathogens type A F. tularensis (Schu S4) and N. gonorrhoeae. As low as 4.4 µM resazurin was sufficient for a 10-fold reduction in F. tularensis growth. In broth culture, resazurin was reduced to resorufin by F. tularensis. Resorufin also suppressed the growth of F. tularensis suggesting that this compound is the biologically active form responsible for decreasing the viability of F. tularensis LVS bacteria. Replication of F. tularensis in primary human macrophages and non-phagocytic cells was abolished following treatment with 44 µM resazurin indicating this compound could be an effective therapy for tularemia in vivo.


Asunto(s)
Antiinfecciosos/farmacología , Francisella tularensis/efectos de los fármacos , Oxazinas/farmacología , Xantenos/farmacología , Antiinfecciosos/metabolismo , Biotransformación , Línea Celular , Recuento de Colonia Microbiana , Células Epiteliales/microbiología , Humanos , Macrófagos/microbiología , Viabilidad Microbiana/efectos de los fármacos , Oxazinas/metabolismo , Xantenos/metabolismo
17.
Am J Pathol ; 183(4): 1223-1232, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23920326

RESUMEN

Francisella tularensis is the causative agent of the debilitating febrile illness tularemia. The severe morbidity associated with F. tularensis infections is attributed to its ability to evade the host immune response. Innate immune activation is undetectable until more than 48 hours after infection. The ensuing inflammatory response is considered pathological, eliciting a septic-like state characterized by hypercytokinemia and cell death. To investigate potential pathological consequences of the innate immune response, mice deficient in a key innate immune signaling molecule, MyD88, were studied. MyD88 knockout (KO) mice were infected with the prototypical virulent F. tularensis strain, Schu S4. MyD88 KO mice succumbed to infection more rapidly than wild-type mice. The enhanced pathogenicity of Schu S4 in MyD88 KO mice was associated with greater bacterial burdens in lungs and distal organs, and the absence of IFN-γ in the lungs, spleens, and sera. Cellular infiltrates were not observed on histological evaluation of the lungs, livers, or spleens of MyD88 KO mice, the first KO mouse described with this phenotype to our knowledge. Despite the absence of cellular infiltration, there was more cell death in the lungs of MyD88 KO mice. Thus, the host proinflammatory response is beneficial, and MyD88 signaling is required to limit bacterial burden and prolong survival during pulmonary infection by virulent F. tularensis.


Asunto(s)
Costo de Enfermedad , Francisella tularensis/patogenicidad , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Tularemia/patología , Animales , Muerte Celular , Citocinas/metabolismo , Francisella tularensis/crecimiento & desarrollo , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Enfermedades Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Análisis de Supervivencia , Tularemia/metabolismo , Tularemia/microbiología , Virulencia
18.
Microbes Infect ; 15(3): 201-11, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23211929

RESUMEN

Pneumonic tularemia is a potentially fatal disease caused by the Category A bioterrorism agent Francisella tularensis. Understanding the pulmonary immune response to this bacterium is necessary for developing effective vaccines and therapeutics. In this study, characterization of immune cell populations in the lungs of mice infected with the type A strain Schu S4 revealed a significant loss in natural killer (NK) cells over time. Since this decline in NK cells correlated with morbidity and mortality, we hypothesized these cells contribute to host defense against Schu S4 infection. Depletion of NK cells prior to Schu S4 challenge significantly reduced IFN-γ and granzyme B in the lung but had no effect on bacterial burden or disease progression. Conversely, increasing NK cell numbers with the anti-apoptotic cytokine IL-15 and soluble receptor IL-15Rα had no significant impact on Schu S4 growth in vivo. A modest decrease in median time to death, however, was observed in live vaccine strain (LVS)-vaccinated mice depleted of NK1.1+ cells and challenged with Schu S4. Therefore, NK cells do not appear to contribute to host defense against acute respiratory infection with type A F. tularensis in vivo, but they play a minor role in protection elicited by LVS vaccination.


Asunto(s)
Francisella tularensis/inmunología , Células Asesinas Naturales/inmunología , Tularemia/inmunología , Análisis de Varianza , Animales , Supervivencia Celular/inmunología , Femenino , Granzimas/inmunología , Granzimas/metabolismo , Interacciones Huésped-Patógeno , Interferón gamma/metabolismo , Interleucina-15/inmunología , Interleucina-15/metabolismo , Células Asesinas Naturales/microbiología , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Tularemia/microbiología
19.
Cytokine ; 60(1): 233-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22749533

RESUMEN

Mycobacterium tuberculosis (MTB) remains a leading infectious threat to human health. Macrophages are the cells targeted for infection by the bacterium as well as key effector cells for clearance of the pathogen. Interleukin (IL)-27 opposes macrophage-mediated control of MTB because supplying IL-12 and blocking the activity of IL-27 limits bacterial growth in primary human macrophages. The purpose of this study was to determine the immunological regulators of this macrophage mechanism to restrict MTB growth. Interferon (IFN)-γ, TNF-α, and IL-18 were all demonstrated to be important to the environment that limits bacterial growth when IL-12 is supplied and IL-27 is neutralized. We find IL-18 works in conjunction with IL-12 to achieve optimal IFN-γ production in this system. We also demonstrate novel interactions between these cytokines to influence the expression or responsiveness to one another. Quantitative assays show that IFN-γ enhances expression of the IL-18 receptor signaling chain, as well as TNF expression and secretion. In turn, TNF-α augments expression of the receptor for IFN-γ, the amount at the cell surface, and the extent of IFN-γ -induced signaling. We further define how the cytokine environment supports an enhanced state of classical macrophage activation. Collectively, these results describe novel immunological mechanisms that provide additional insights into the effects of IL-12 and IL-27 on macrophage regulation during MTB infection.


Asunto(s)
Interferón gamma/inmunología , Interleucina-18/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Arginasa/genética , Arginasa/inmunología , Arginasa/metabolismo , Células Cultivadas , Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Immunoblotting , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-12/farmacología , Interleucina-18/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Microscopía Confocal , Mycobacterium tuberculosis/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Interferón/metabolismo , Receptores de Interleucina-18/genética , Receptores de Interleucina-18/inmunología , Receptores de Interleucina-18/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Receptor de Interferón gamma
20.
PLoS One ; 7(2): e31172, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355343

RESUMEN

Vaccination is a proven strategy to mitigate morbidity and mortality of infectious diseases. The methodology of identifying and testing new vaccine candidates could be improved with rational design and in vitro testing prior to animal experimentation. The tularemia vaccine, Francisella tularensis live vaccine strain (LVS), does not elicit complete protection against lethal challenge with a virulent type A Francisella strain. One factor that may contribute to this poor performance is limited stimulation of antigen-presenting cells. In this study, we examined whether the interaction of genetically modified LVS strains with human antigen-presenting cells correlated with effectiveness as tularemia vaccine candidates. Human dendritic cells infected with wild-type LVS secrete low levels of proinflammatory cytokines, fail to upregulate costimulatory molecules, and activate human T cells poorly in vitro. One LVS mutant, strain 13B47, stimulated higher levels of proinflammatory cytokines from dendritic cells and macrophages and increased costimulatory molecule expression on dendritic cells compared to wild type. Additionally, 13B47-infected dendritic cells activated T cells more efficiently than LVS-infected cells. A deletion allele of the same gene in LVS displayed similar in vitro characteristics, but vaccination with this strain did not improve survival after challenge with a virulent Francisella strain. In vivo, this mutant was attenuated for growth and did not stimulate T cell responses in the lung comparable to wild type. Therefore, stimulation of antigen-presenting cells in vitro was improved by genetic modification of LVS, but did not correlate with efficacy against challenge in vivo within this model system.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vacunas Bacterianas/administración & dosificación , Francisella tularensis/inmunología , Inmunización , Tularemia/terapia , Vacunas Atenuadas/administración & dosificación , Animales , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Femenino , Citometría de Flujo , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , Tularemia/inmunología , Tularemia/mortalidad , Vacunación , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA