Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 128(5): 570-584, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33501852

RESUMEN

RATIONALE: Patients with and without cardiovascular diseases have been shown to be at risk of influenza-mediated cardiac complications. Recent clinical reports support the notion of a direct link between laboratory-confirmed influenza virus infections and adverse cardiac events. OBJECTIVE: Define the molecular mechanisms underlying influenza virus-induced cardiac pathogenesis after resolution of pulmonary infection and the role of necroptosis in this process. METHODS AND RESULTS: Hearts from wild-type and necroptosis-deficient (MLKL [mixed lineage kinase domain-like protein]-KO) mice were dissected 12 days after initial influenza A virus (IAV) infection when viral titers were undetectable in the lungs. Immunofluorescence microscopy and plaque assays showed presence of viable IAV particles in the myocardium without generation of interferon responses. Global proteome and phosphoproteome analyses using high-resolution accurate mass-based LC-MS/MS and label-free quantitation showed that the global proteome as well as the phosphoproteome profiles were significantly altered in IAV-infected mouse hearts in a strain-independent manner. Necroptosis-deficient mice had increased survival and reduced weight loss post-IAV infection, as well as increased antioxidant and mitochondrial function, indicating partial protection to IAV infection. These findings were confirmed in vitro by pretreatment of human and rat myocytes with antioxidants or necroptosis inhibitors, which blunted oxidative stress and mitochondrial damage after IAV infection. CONCLUSIONS: This study provides the first evidence that the cardiac proteome and phosphoproteome are significantly altered post-pulmonary influenza infection. Moreover, viral particles can persist in the heart after lung clearance, altering mitochondrial function and promoting cell death without active replication and interferon responses. Finally, our findings show inhibition of necroptosis or prevention of mitochondrial damage as possible therapeutic interventions to reduce cardiac damage during influenza infections. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Cardiopatías/metabolismo , Miocitos Cardíacos/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Proteoma/metabolismo , Animales , Línea Celular , Cardiopatías/etiología , Cardiopatías/virología , Humanos , Virus de la Influenza A/patogenicidad , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/virología , Necroptosis , Infecciones por Orthomyxoviridae/complicaciones , Estrés Oxidativo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Quinasas/genética , Proteoma/genética , Ratas
2.
Crit Care ; 27(1): 155, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37081485

RESUMEN

BACKGROUND: The mechanisms used by SARS-CoV-2 to induce major adverse cardiac events (MACE) are unknown. Thus, we aimed to determine if SARS-CoV-2 can induce necrotic cell death to promote MACE in patients with severe COVID-19. METHODS: This observational prospective cohort study includes experiments with hamsters and human samples from patients with severe COVID-19. Cytokines and serum biomarkers were analysed in human serum. Cardiac transcriptome analyses were performed in hamsters' hearts. RESULTS: From a cohort of 70 patients, MACE was documented in 26% (18/70). Those who developed MACE had higher Log copies/mL of SARS-CoV-2, troponin-I, and pro-BNP in serum. Also, the elevation of IP-10 and a major decrease in levels of IL-17ɑ, IL-6, and IL-1rɑ were observed. No differences were found in the ability of serum antibodies to neutralise viral spike proteins in pseudoviruses from variants of concern. In hamster models, we found a stark increase in viral titters in the hearts 4 days post-infection. The cardiac transcriptome evaluation resulted in the differential expression of ~ 9% of the total transcripts. Analysis of transcriptional changes in the effectors of necroptosis (mixed lineage kinase domain-like, MLKL) and pyroptosis (gasdermin D) showed necroptosis, but not pyroptosis, to be elevated. An active form of MLKL (phosphorylated MLKL, pMLKL) was elevated in hamster hearts and, most importantly, in the serum of MACE patients. CONCLUSION: SARS-CoV-2 identification in the systemic circulation is associated with MACE and necroptosis activity. The increased pMLKL and Troponin-I indicated the occurrence of necroptosis in the heart and suggested necroptosis effectors could serve as biomarkers and/or therapeutic targets. Trial registration Not applicable.


Asunto(s)
COVID-19 , Enfermedades Cardiovasculares , Humanos , Proteínas Quinasas , Necroptosis , Estudios Prospectivos , Troponina I , SARS-CoV-2 , Biomarcadores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores
3.
J Virol ; 95(20): e0101021, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34319784

RESUMEN

The host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is poorly understood due to a lack of an animal model that recapitulates severe human disease. Here, we report a Syrian hamster model that develops progressive lethal pulmonary disease that closely mimics severe coronavirus disease 2019 (COVID-19). We evaluated host responses using a multi-omic, multiorgan approach to define proteome, phosphoproteome, and transcriptome changes. These data revealed both type I and type II interferon-stimulated gene and protein expression along with a progressive increase in chemokines, monocytes, and neutrophil-associated molecules throughout the course of infection that peaked in the later time points correlating with a rapidly developing diffuse alveolar destruction and pneumonia that persisted in the absence of active viral infection. Extrapulmonary proteome and phosphoproteome remodeling was detected in the heart and kidneys following viral infection. Together, our results provide a kinetic overview of multiorgan host responses to severe SARS-CoV-2 infection in vivo. IMPORTANCE The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has created an urgent need to understand the pathogenesis of this infection. These efforts have been impaired by the lack of animal models that recapitulate severe coronavirus disease 2019 (COVID-19). Here, we report a hamster model that develops severe COVID-19-like disease following infection with human isolates of SARS-CoV-2. To better understand pathogenesis, we evaluated changes in gene transcription and protein expression over the course of infection to provide an integrated multiorgan kinetic analysis of the host response to infection. These data reveal a dynamic innate immune response to infection and corresponding immune pathologies consistent with severe human disease. Altogether, this model will be useful for understanding the pathogenesis of severe COVID-19 and for testing interventions.


Asunto(s)
COVID-19/inmunología , COVID-19/metabolismo , Inmunidad Innata , Proteoma , Transcriptoma , Animales , COVID-19/genética , COVID-19/virología , Modelos Animales de Enfermedad , Ontología de Genes , Corazón/virología , Riñón/metabolismo , Riñón/virología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Mesocricetus , Miocardio/metabolismo , Fosfoproteínas/metabolismo , Proteómica , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad , Carga Viral
4.
Nat Immunol ; 10(10): 1073-80, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19701189

RESUMEN

Pattern-recognition receptors (PRRs), including Toll-like receptors (TLRs) and RIG-like helicase (RLH) receptors, are involved in innate immune antiviral responses. Here we show that nucleotide-binding oligomerization domain 2 (Nod2) can also function as a cytoplasmic viral PRR by triggering activation of interferon-regulatory factor 3 (IRF3) and production of interferon-beta (IFN-beta). After recognition of a viral ssRNA genome, Nod2 used the adaptor protein MAVS to activate IRF3. Nod2-deficient mice failed to produce interferon efficiently and showed enhanced susceptibility to virus-induced pathogenesis. Thus, the function of Nod2 as a viral PRR highlights the important function of Nod2 in host antiviral defense mechanisms.


Asunto(s)
Inmunidad Innata , Proteína Adaptadora de Señalización NOD2/inmunología , ARN Viral/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Fenómenos del Sistema Inmunológico , Immunoblotting , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Factor 3 Regulador del Interferón/biosíntesis , Factor 3 Regulador del Interferón/inmunología , Interferón beta/biosíntesis , Interferón beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , ARN Interferente Pequeño , Receptores de Reconocimiento de Patrones/genética , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Immunol ; 203(9): 2520-2531, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562213

RESUMEN

The innate immune sensing of allergens or allergen-associated components regulate the development of type 2 inflammatory responses. However, the underlying molecular basis by which allergens or allergen-associated components are detected by innate immune receptors remains elusive. In this study, we report that the most common aeroallergen, house dust mite (HDM), harbors a dsRNA species (HDM-dsRNA) that can activate TLR3-mediated IFN responses and counteract the development of an uncontrolled type 2 immune response. We demonstrate that the mouse strains defective in the dsRNA-sensing pathways show aggravated type 2 inflammation defined by severe eosinophilia, elevated level of type 2 cytokines, and mucus overproduction in a model of allergic lung inflammation. The inability to sense HDM-dsRNA resulted in significant increases in airway hyperreactivity. We further show that the administration of the purified HDM-dsRNA at a low dose is sufficient to induce an immune response to prevent the onset of a severe type 2 lung inflammation. Collectively, these results unveil a new role for the HDM-dsRNA/TLR3-signaling axis in the modulation of a type 2 lung inflammation in mice.


Asunto(s)
Alérgenos/inmunología , Interferones/biosíntesis , Neumonía/etiología , Pyroglyphidae/inmunología , ARN Bicatenario/inmunología , Animales , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Hipersensibilidad Respiratoria/etiología , Transducción de Señal/fisiología , Receptor Toll-Like 3/fisiología
6.
Microb Pathog ; 129: 242-249, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30776411

RESUMEN

Environmental allergens elicit complex immune responses in the lungs that can promote the development of asthma or exacerbate preexisting asthma in susceptible individuals. House dust mites are one of the most common indoor allergens and are a significant driver of allergic disease. Respiratory infections are known factors in acute exacerbations of asthma but the impact of allergen on the pathogen is not well understood. We investigated the pathogenesis of influenza A infection following exposure to house dust mites. Mice exposed to house dust mites lose less weight following infection and had more transcription of interferon-lambda than controls. These data correlated with less transcription of the influenza polymerase acidic gene suggesting diminished viral replication in house dust mite exposed mice. Altogether, these data suggest that exposure to environmental allergens can influence the pathogenesis of influenza infection.


Asunto(s)
Alérgenos/administración & dosificación , Asma/complicaciones , Infecciones por Orthomyxoviridae/prevención & control , Pyroglyphidae/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interferones/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/patología
7.
Am J Respir Cell Mol Biol ; 58(2): 253-260, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28915064

RESUMEN

Mycoplasma pneumoniae infection has been linked to poor asthma outcomes. M. pneumoniae produces an ADP-ribosylating and vacuolating toxin called community-acquired respiratory distress syndrome (CARDS) toxin that has a major role in inflammation and airway dysfunction. The objective was to evaluate the immunopathological effects in primates exposed to M. pneumoniae or CARDS toxin. A total of 13 baboons were exposed to M. pneumoniae or CARDS toxin. At Days 7 and 14, BAL fluid was collected and analyzed for cell count, percent of each type of cell, CARDS toxin by PCR, CARDS toxin by antigen capture, eosinophilic cationic protein, and cytokine profiles. Serum IgM, IgG, and IgE responses to CARDS toxin were measured. All animals had a necropsy for analysis of the histopathological changes on lungs. No animal developed signs of infection. The serological responses to CARDS toxin were variable. At Day 14, four of seven animals exposed to M. pneumoniae and all four animals exposed to CARDS toxin developed histological "asthma-like" changes. T cell intracellular cytokine analysis revealed an increasing ratio of IL-4/IFN-γ over time. Both M. pneumoniae and CARDS toxin exposure resulted in similar histopathological pulmonary changes, suggesting that CARDS toxin plays a major role in the inflammatory response.


Asunto(s)
Asma/inmunología , Asma/patología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Pulmón/inmunología , Pulmón/patología , Mycoplasma pneumoniae/patogenicidad , Animales , Linfocitos T CD4-Positivos/inmunología , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Interleucina-13/inmunología , Interleucina-4/inmunología , Pulmón/microbiología , Ratones , Mycoplasma pneumoniae/inmunología , Papio
8.
Am J Respir Crit Care Med ; 196(5): 609-620, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28614669

RESUMEN

RATIONALE: Up to one-third of patients hospitalized with pneumococcal pneumonia experience major adverse cardiac events (MACE) during or after pneumonia. In mice, Streptococcus pneumoniae can invade the myocardium, induce cardiomyocyte death, and disrupt cardiac function following bacteremia, but it is unknown whether the same occurs in humans with severe pneumonia. OBJECTIVES: We sought to determine whether S. pneumoniae can (1) translocate the heart, (2) induce cardiomyocyte death, (3) cause MACE, and (4) induce cardiac scar formation after antibiotic treatment during severe pneumonia using a nonhuman primate (NHP) model. METHODS: We examined cardiac tissue from six adult NHPs with severe pneumococcal pneumonia and three uninfected control animals. Three animals were rescued with antibiotics (convalescent animals). Electrocardiographic, echocardiographic, and serum biomarkers of cardiac damage were measured (troponin T, N-terminal pro-brain natriuretic peptide, and heart-type fatty acid binding protein). Histological examination included hematoxylin and eosin staining, immunofluorescence, immunohistochemistry, picrosirius red staining, and transmission electron microscopy. Immunoblots were used to assess the underlying mechanisms. MEASUREMENTS AND MAIN RESULTS: Nonspecific ischemic alterations were detected by electrocardiography and echocardiography. Serum levels of troponin T and heart-type fatty acid binding protein were increased (P < 0.05) after pneumococcal infection in both acutely ill and convalescent NHPs. S. pneumoniae was detected in the myocardium of all NHPs with acute severe pneumonia. Necroptosis and apoptosis were detected in the myocardium of both acutely ill and convalescent NHPs. Evidence of cardiac scar formation was observed only in convalescent animals by transmission electron microscopy and picrosirius red staining. CONCLUSIONS: S. pneumoniae invades the myocardium and induces cardiac injury with necroptosis and apoptosis, followed by cardiac scarring after antibiotic therapy, in an NHP model of severe pneumonia.


Asunto(s)
Cardiotoxicidad/etiología , Miocardio/patología , Neumonía Neumocócica/complicaciones , Streptococcus pneumoniae/patogenicidad , Animales , Antibacterianos/uso terapéutico , Western Blotting , Cardiotoxicidad/sangre , Modelos Animales de Enfermedad , Ecocardiografía , Electrocardiografía , Proteínas de Unión a Ácidos Grasos/sangre , Femenino , Corazón/microbiología , Masculino , Papio , Neumonía Neumocócica/sangre , Neumonía Neumocócica/tratamiento farmacológico , Troponina T/sangre
9.
Ann Allergy Asthma Immunol ; 119(2): 146-152.e2, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28634021

RESUMEN

BACKGROUND: Acute infections with Mycoplasma pneumoniae (Mp) have been associated with worsening asthma in children. Mp can be present in the respiratory tract for extended periods; it is unknown whether the long-term persistence of Mp in the respiratory tract affects long-term asthma control. OBJECTIVE: To determine the effect of Mp on asthma control. METHODS: We enrolled 31 pediatric subjects 3 to 10 years of age with persistent asthma who completed up to 8 visits over a 24-month period. We detected Mp by antigen capture and polymerase chain reaction. Primary outcome measurements included symptom scores, quality of life, medication scores, oral corticosteroid use, health care usage, school absences, and exhaled breath condensate pH. RESULTS: Low levels of Mp community-acquired respiratory distress syndrome toxin were detected in 20 subjects (64.5%) at enrollment. Subjects with Mp positivity at a given visit had a .579 probability of remaining Mp positive at the subsequent visit, whereas those with Mp negativity had a .348 probability of becoming Mp positive at the following visit. The incidence of Mp overall was higher in the spring and summer months. Overall, we found no significant relation between the detection of Mp and worse outcome measurements at the same visit or at subsequent visits. CONCLUSION: The long-term persistence of Mp in the respiratory tract is common in children with asthma. However, the detection of Mp was not associated significantly with worse asthma symptoms, quality of life, health care usage, school absences, or exhaled breath condensate pH in this pediatric asthma cohort.


Asunto(s)
Asma/inmunología , Asma/microbiología , Estado de Salud , Mycoplasma pneumoniae/aislamiento & purificación , Calidad de Vida , Sistema Respiratorio/microbiología , Niño , Preescolar , Femenino , Humanos , Masculino , Mycoplasma pneumoniae/inmunología , Neumonía por Mycoplasma/inmunología , Neumonía por Mycoplasma/microbiología , Estudios Prospectivos , Estaciones del Año
10.
Infect Immun ; 84(5): 1457-69, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26930705

RESUMEN

Streptococcus pneumoniae (the pneumococcus) is capable of invading the heart. Herein we observed that pneumococcal invasion of the myocardium occurred soon after development of bacteremia and was continuous thereafter. Using immunofluorescence microscopy (IFM), we observed that S. pneumoniae replication within the heart preceded visual signs of tissue damage in cardiac tissue sections stained with hematoxylin and eosin. Different S. pneumoniae strains caused distinct cardiac pathologies: strain TIGR4, a serotype 4 isolate, caused discrete pneumococcus-filled microscopic lesions (microlesions), whereas strain D39, a serotype 2 isolate, was, in most instances, detectable only using IFM and was associated with foci of cardiomyocyte hydropic degeneration and immune cell infiltration. Both strains efficiently invaded the myocardium, but cardiac damage was entirely dependent on the pore-forming toxin pneumolysin only for D39. Early microlesions caused by TIGR4 and microlesions formed by a TIGR4 pneumolysin-deficient mutant were infiltrated with CD11b(+) and Ly6G-positive neutrophils and CD11b(+) and F4/80-positive (F4/80(+)) macrophages. We subsequently demonstrated that macrophages in TIGR4-infected hearts died as a result of pneumolysin-induced necroptosis. The effector of necroptosis, phosphorylated mixed-lineage kinase domain-like protein (MLKL), was detected in CD11b(+) and F4/80(+) cells associated with microlesions. Likewise, treatment of infected mice and THP-1 macrophages in vitro with the receptor-interacting protein 1 kinase (RIP1) inhibitor necrostatin-5 promoted the formation of purulent microlesions and blocked cell death, respectively. We conclude that pneumococci that have invaded the myocardium are an important cause of cardiac damage, pneumolysin contributes to cardiac damage in a bacterial strain-specific manner, and pneumolysin kills infiltrated macrophages via necroptosis, which alters the immune response.


Asunto(s)
Bacteriemia/patología , Muerte Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Miocarditis/patología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/toxicidad , Animales , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones Endogámicos BALB C , Microscopía Fluorescente , Proteínas Quinasas/análisis , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo
11.
J Immunol ; 193(3): 1373-82, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24965773

RESUMEN

How intestinal epithelial cells (IECs) recognize pathogens and activate inflammasomes at intestinal surfaces is poorly understood. We hypothesized that IECs use integrin receptors to recognize pathogens and initiate inflammation within the intestinal tract. We find that IECs infected with Yersinia enterocolitica, an enteric pathogen, use ß1 integrins as pathogen recognition receptors detecting the bacterial adhesin invasin (Inv). The Inv-integrin interaction provides the first signal for NLRP3 inflammasome activation with the type three secretion system translocon providing the second signal for inflammasome activation, resulting in release of IL-18. During infection, Yersinia employs two virulence factors, YopE and YopH, to counteract Inv-mediated integrin-dependent inflammasome activation. Furthermore, NLRP3 inflammasome activation in epithelial cells requires components of the focal adhesion complex signaling pathway, focal adhesion kinase, and rac1. The binding of Inv to ß1 integrins rapidly induces IL-18 mRNA expression, suggesting integrins provide a first signal for NLRP3 inflammasome activation. These data suggest integrins function as pathogen recognition receptors on IECs to rapidly induce inflammasome-derived IL-18-mediated responses.


Asunto(s)
Células Epiteliales/inmunología , Inflamasomas/inmunología , Inflamasomas/metabolismo , Integrina alfa5beta1/fisiología , Mucosa Intestinal/inmunología , Transducción de Señal/inmunología , Yersinia enterocolitica/inmunología , Adhesinas Bacterianas/metabolismo , Adhesinas Bacterianas/fisiología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/farmacología , Células CACO-2 , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Interleucina-18/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Proteína con Dominio Pirina 3 de la Familia NLR , Unión Proteica/inmunología , Proteínas Tirosina Fosfatasas/deficiencia , Proteínas Tirosina Fosfatasas/genética , Factores de Virulencia/fisiología , Yersinia enterocolitica/genética
12.
Infect Immun ; 83(2): 614-24, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25422267

RESUMEN

Serratia marcescens, a member of the carbapenem-resistant Enterobacteriaceae, is an important emerging pathogen that causes a wide variety of nosocomial infections, spreads rapidly within hospitals, and has a systemic mortality rate of ≤41%. Despite multiple clinical descriptions of S. marcescens nosocomial pneumonia, little is known regarding the mechanisms of bacterial pathogenesis and the host immune response. To address this gap, we developed an oropharyngeal aspiration model of lethal and sublethal S. marcescens pneumonia in BALB/c mice and extensively characterized the latter. Lethal challenge (>4.0 × 10(6) CFU) was characterized by fulminate hemorrhagic pneumonia with rapid loss of lung function and death. Mice challenged with a sublethal dose (<2.0 × 10(6) CFU) rapidly lost weight, had diminished lung compliance, experienced lung hemorrhage, and responded to the infection with extensive neutrophil infiltration and histopathological changes in tissue architecture. Neutrophil extracellular trap formation and the expression of inflammatory cytokines occurred early after infection. Mice depleted of neutrophils were exquisitely susceptible to an otherwise nonlethal inoculum, thereby demonstrating the requirement for neutrophils in host protection. Mutation of the genes encoding the cytolysin ShlA and its transporter ShlB resulted in attenuated S. marcescens strains that failed to cause profound weight loss, extended illness, hemorrhage, and prolonged lung pathology in mice. This study describes a model of S. marcescens pneumonia that mimics known clinical features of human illness, identifies neutrophils and the toxin ShlA as a key factors important for defense and infection, respectively, and provides a solid foundation for future studies of novel therapeutics for this important opportunistic pathogen.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Hemolisinas/genética , Neumonía/patología , Infecciones por Serratia/inmunología , Serratia marcescens/inmunología , Animales , Líquido del Lavado Bronquioalveolar/citología , Infección Hospitalaria , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Hemorragia/microbiología , Hemorragia/patología , Inflamación/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Neumonía/microbiología , Neumonía/mortalidad , Infecciones por Serratia/microbiología , Infecciones por Serratia/mortalidad , Serratia marcescens/patogenicidad
14.
Res Sq ; 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36711834

RESUMEN

Background The mechanisms used by SARS-CoV-2 to induce major adverse cardiac events (MACE) are unknown. Thus, we aimed to determine if SARS-CoV-2 can infect the heart to kill cardiomyocytes and induce MACE in patients with severe COVID-19. Methods This observational prospective cohort study includes experiments with hamsters and human samples from patients with severe COVID-19. Cytokines and serum biomarkers were analyzed in human serum. Cardiac transcriptome analyses were performed in hamsters' hearts. Results From a cohort of 70 patients, MACE was documented in 26% (18/70). Those who developed MACE had higher Log copies/mL of SARS-CoV-2, troponin-I, and pro-BNP in serum. Also, the elevation of IP-10 and a major decrease in levels of IL-17ɑ, IL-6, and IL-1rɑ were observed. No differences were found in the ability of serum antibodies to neutralize viral spike proteins in pseudoviruses from variants of concern. In hamster models, we found a stark increase in viral titers in the hearts 4 days post-infection. The cardiac transcriptome evaluation resulted in the differential expression of ~ 9% of the total transcripts. Analysis of transcriptional changes of the effectors of necroptosis (mixed lineage kinase domain-like, MLKL) and pyroptosis (gasdermin D) showed necroptosis, but not pyroptosis, to be elevated. Active form of MLKL (phosphorylated MLKL, pMLKL) was elevated in hamster hearts and, most importantly, in the serum of MACE patients. Conclusion SARS-CoV-2 can reach the heart during severe COVID-19 and induce necroptosis in the heart of patients with MACE. Thus, pMLKL could be used as a biomarker of cardiac damage and a therapeutic target. Trial registration: Not applicable.

15.
Am J Respir Cell Mol Biol ; 46(6): 815-22, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22281984

RESUMEN

Mycoplasma pneumoniae causes acute and chronic lung infections in humans, leading to a variety of pulmonary and extrapulmonary sequelae. Of the airway complications of M. pneumoniae infection, M. pneumoniae-associated exacerbation of asthma and pediatric wheezing are emerging as significant sources of human morbidity. However, M. pneumoniae products capable of promoting allergic inflammation are unknown. Recently, we reported that M. pneumoniae produces an ADP-ribosylating and vacuolating toxin termed the community-acquired respiratory distress syndrome (CARDS) toxin. Here we report that naive mice exposed to a single dose of recombinant CARDS (rCARDS) toxin respond with a robust inflammatory response consistent with allergic disease. rCARDS toxin induced 30-fold increased expression of the Th-2 cytokines IL-4 and IL-13 and 70- to 80-fold increased expression of the Th-2 chemokines CCL17 and CCL22, corresponding to a mixed cellular inflammatory response comprised of a robust eosinophilia, accumulation of T cells and B cells, and mucus metaplasia. The inflammatory responses correlate temporally with toxin-dependent increases in airway hyperreactivity characterized by increases in airway restriction and decreases in lung compliance. Furthermore, CARDS toxin-mediated changes in lung function and histopathology are dependent on CD4(+) T cells. Altogether, the data suggest that rCARDS toxin is capable of inducing allergic-type inflammation in naive animals and may represent a causal factor in M. pneumoniae-associated asthma.


Asunto(s)
Toxinas Bacterianas/toxicidad , Eosinófilos/citología , Pulmón/efectos de los fármacos , Linfocitos/citología , Mycoplasma pneumoniae/fisiología , Animales , Líquido del Lavado Bronquioalveolar , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Pulmón/citología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
16.
Infect Immun ; 80(1): 289-97, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22083707

RESUMEN

Yersinia enterocolitica is a food-borne pathogen that preferentially infects the Peyer's patches and mesenteric lymph nodes, causing an acute inflammatory reaction. Even though Y. enterocolitica induces a robust inflammatory response during infection, the bacterium has evolved a number of virulence factors to limit the extent of this response. We previously demonstrated that interleukin-1α (IL-1α) was critical for the induction of gut inflammation characteristic of Y. enterocolitica infection. More recently, the known actions of IL-1α are becoming more complex because IL-1α can function both as a proinflammatory cytokine and as a nuclear factor. In this study, we tested the ability of Y. enterocolitica to modulate intracellular IL-1α-dependent IL-8 production in epithelial cells. Nuclear translocation of pre-IL-1α protein and IL-1α-dependent secretion of IL-8 into the culture supernatant were increased during infection with a strain lacking the 70-kDa virulence plasmid compared to the case during infection with the wild type, suggesting that Yersinia outer proteins (Yops) might be involved in modulating intracellular IL-1α signaling. Infection of HeLa cells with a strain lacking the yopP gene resulted in increased nuclear translocation of pre-IL-1α and IL-1α-dependent secretion of IL-8 similar to what is observed with bacteria lacking the virulence plasmid. YopP is a protein acetylase that inhibits mitogen-activated protein kinase (MAP kinase)- and NF-κB-dependent signal transduction pathways. Nuclear translocation of pre-IL-1α and IL-1α-dependent secretion of IL-8 in response to Yersinia enterocolitica infection were dependent on extracellular signal-regulated kinase (ERK) and p38 MAP kinase signaling but independent of NF-κB. These data suggest that Y. enterocolitica inhibits intracellular pre-IL-1α signaling and subsequent proinflammatory responses through inhibition of MAP kinase pathways.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-8/biosíntesis , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Transducción de Señal , Yersinia enterocolitica/patogenicidad , Transporte Activo de Núcleo Celular , Proteínas Bacterianas/inmunología , Núcleo Celular/química , Citoplasma/química , Regulación hacia Abajo , Células Epiteliales/inmunología , Células HeLa , Humanos , Interleucina-8/antagonistas & inhibidores , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo , Yersinia enterocolitica/inmunología
17.
Cell Res ; 32(9): 831-842, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35906408

RESUMEN

SARS-CoV-2 variants with adaptive mutations have continued to emerge, causing fresh waves of infection even amongst vaccinated population. The development of broad-spectrum antivirals is thus urgently needed. We previously developed two hetero-bivalent nanobodies (Nbs), aRBD-2-5 and aRBD-2-7, with potent neutralization activity against the wild-type (WT) Wuhan isolated SARS-CoV-2, by fusing aRBD-2 with aRBD-5 and aRBD-7, respectively. Here, we resolved the crystal structures of these Nbs in complex with the receptor-binding domain (RBD) of the spike protein, and found that aRBD-2 contacts with highly-conserved RBD residues and retains binding to the RBD of the Alpha, Beta, Gamma, Delta, Delta plus, Kappa, Lambda, Omicron BA.1, and BA.2 variants. In contrast, aRBD-5 and aRBD-7 bind to less-conserved RBD epitopes non-overlapping with the epitope of aRBD-2, and do not show apparent binding to the RBD of some variants. However, when fused with aRBD-2, they effectively enhance the overall binding affinity. Consistently, aRBD-2-5-Fc and aRBD-2-7-Fc potently neutralized all of the tested authentic or pseudotyped viruses, including WT, Alpha, Beta, Gamma, Delta, and Omicron BA.1, BA.1.1 and BA.2. Furthermore, aRBD-2-5-Fc provided prophylactic protection against the WT and mouse-adapted SARS-CoV-2 in mice, and conferred protection against the Omicron BA.1 variant in hamsters prophylactically and therapeutically, indicating that aRBD-2-5-Fc could potentially benefit the prevention and treatment of COVID-19 caused by the emerging variants of concern. Our strategy provides new solutions in the development of broad-spectrum therapeutic antibodies for COVID-19.


Asunto(s)
COVID-19 , Anticuerpos de Dominio Único , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/uso terapéutico , Epítopos , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2 , Anticuerpos de Dominio Único/farmacología , Glicoproteína de la Espiga del Coronavirus/genética
18.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33400692

RESUMEN

2'3'-cGAMP is known as a nonclassical second messenger and small immune modulator that possesses potent antitumor and antiviral activities via inducing the stimulator of IFN genes-mediated (STING-mediated) signaling pathway. However, its function in regulating type 2 immune responses remains unknown. Therefore, we sought to determine a role of STING activation by 2'3'-cGAMP in type 2 inflammatory reactions in multiple mouse models of eosinophilic asthma. We discovered that 2'3'-cGAMP administration strongly attenuated type 2 lung immunopathology and airway hyperreactivity induced by IL-33 and a fungal allergen, Aspergillus flavus. Mechanistically, upon the respiratory delivery, 2'3'-cGAMP was mainly internalized by alveolar macrophages, in which it activated the STING/IFN regulatory factor 3/type I IFN signaling axis to induce the production of inhibitory factors containing IFN-α, which blocked the IL-33-mediated activation of group 2 innate lymphoid (ILC2) cells in vivo. We further demonstrated that 2'3'-cGAMP directly suppressed the proliferation and function of both human and mouse ILC2 cells in vitro. Taken together, our findings suggest that STING activation by 2'3'-cGAMP in alveolar macrophages and ILC2 cells can negatively regulate type 2 immune responses, implying that the respiratory delivery of 2'3'-cGAMP might be further developed as an alternative strategy for treating type 2 immunopathologic diseases such as eosinophilic asthma.


Asunto(s)
Asma/inmunología , Interleucina-33/metabolismo , Linfocitos/inmunología , Macrófagos Alveolares/inmunología , Proteínas de la Membrana/metabolismo , Alérgenos/administración & dosificación , Animales , Aspergillus flavus/inmunología , Asma/metabolismo , Asma/patología , Modelos Animales de Enfermedad , Eosinofilia/inmunología , Eosinofilia/metabolismo , Eosinofilia/patología , Femenino , Nucleótidos de Guanina/administración & dosificación , Nucleótidos de Guanina/inmunología , Nucleótidos de Guanina/metabolismo , Humanos , Inmunidad Innata , Técnicas In Vitro , Interleucina-33/administración & dosificación , Interleucina-33/genética , Linfocitos/patología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal
19.
Infect Immun ; 78(9): 3716-25, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20584975

RESUMEN

Infection of the gut by invasive bacterial pathogens leads to robust inflammatory responses that if left unchecked can lead to autoimmune disease and other sequelae. How the immune system controls inflammation and limits collateral damage to the host during acute bacterial infection is poorly understood. Here, we report that antibody-mediated neutralization of transforming growth factor beta (TGF-beta) prior to infection with the model enteric pathogen Yersinia enterocolitica reduces the mean time to death by 1 day (P=0.001), leads to rapid colonization of the liver and lung, and is associated with exacerbation of inflammatory histopathology. During Yersinia enterocolitica infection CD4+ cells are the source of de novo TGF-beta transcription in the Peyer's patches, mesenteric lymph nodes, and spleen. Correspondingly there is both antigen-specific and -independent expansion of CD4+ CD25+ Foxp3+ and TGF-beta+ T-regulatory cells (T-regs) after Yersinia infection that is reduced in ovalbumin T-cell receptor-restricted OT-II mice. Functional inactivation of CD25 by anti-CD25 treatment results in more rapid death, dissemination of the bacteria to the liver and lungs, and exacerbated inflammatory histopathology, similar to what is seen during TGF-beta neutralization. Altogether, these data suggest that TGF-beta produced by T-regs is important in restricting bacteria during the acute phase of invasive bacterial infection of the gut. These data expand the roles of T-regs to include tempering inflammation during acute infection in addition to the well-established roles of T-regs in chronic infection, control of immune homeostasis, and autoimmune disease.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-2/fisiología , Enfermedades Intestinales/inmunología , Factor de Crecimiento Transformador beta/fisiología , Yersiniosis/inmunología , Yersinia enterocolitica , Animales , Linfocitos T CD4-Positivos/fisiología , Femenino , Interleucina-17/biosíntesis , Hígado/patología , Pulmón/patología , Ganglios Linfáticos/microbiología , Ratones , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/microbiología , Bazo/microbiología , Linfocitos T Reguladores/fisiología , Yersiniosis/patología
20.
BMC Immunol ; 11: 29, 2010 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-20565713

RESUMEN

BACKGROUND: Yersinia pestis is the causative agent of pneumonic plague; recently, we and others reported that during the first 24-36 hours after pulmonary infection with Y. pestis pro-inflammatory cytokine expression is undetectable in lung tissues. RESULTS: Here, we report that, intranasal infection of mice with CO92 delta yopH mutant results in an early pro-inflammatory response in the lungs characterized by an increase in the pro-inflammatory cytokines Tumor Necrosis Factor-alpha and Interleukin one-beta 24 hours post-infection. CO92 delta yopH colonizes the lung but does not disseminate to the liver or spleen and is cleared from the host within 72 hours post-infection. This is different from what is observed in a wild-type CO92 infection, where pro-inflammatory cytokine expression and immune cell infiltration into the lungs is not detectable until 36-48 h post-infection. CO92 rapidly disseminates to the liver and spleen resulting in high bacterial burdens in these tissues ultimately cumulating in death 72-94 h post-infection. Mice deficient in TNF-alpha are more susceptible to CO92 delta yopH infection with 40% of the mice succumbing to infection. CONCLUSIONS: Altogether, our results suggest that YopH can inhibit an early pro-inflammatory response in the lungs of mice and that this is an important step in the pathogenesis of infection.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Citocinas/inmunología , Mediadores de Inflamación/inmunología , Peste/inmunología , Peste/microbiología , Proteínas Tirosina Fosfatasas/inmunología , Yersinia pestis/inmunología , Administración Intranasal , Animales , Anticuerpos/inmunología , Líquido del Lavado Bronquioalveolar/microbiología , Modelos Animales de Enfermedad , Femenino , Interleucina-1beta/biosíntesis , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Mutación/genética , Peste/patología , Proteínas Tirosina Fosfatasas/deficiencia , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/deficiencia , Virulencia/inmunología , Yersinia pestis/enzimología , Yersinia pestis/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA