Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
JAMA Netw Open ; 7(2): e240383, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38407904

RESUMEN

Importance: Sepsis is a leading cause of pediatric mortality. Little attention has been paid to the association between viral DNA and mortality in children and adolescents with sepsis. Objective: To assess the association of the presence of viral DNA with sepsis-related mortality in a large multicenter study. Design, Setting, and Participants: This cohort study compares pediatric patients with and without plasma cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus 1 (HSV-1), human herpesvirus 6 (HHV-6), parvovirus B19 (B19V), BK polyomavirus (BKPyV), human adenovirus (HAdV), and torque teno virus (TTV) DNAemia detected by quantitative real-time polymerase chain reaction or plasma IgG antibodies to CMV, EBV, HSV-1, or HHV-6. A total of 401 patients younger than 18 years with severe sepsis were enrolled from 9 pediatric intensive care units (PICUs) in the Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network. Data were collected from 2015 to 2018. Samples were assayed from 2019 to 2022. Data were analyzed from 2022 to 2023. Main Outcomes and Measures: Death while in the PICU. Results: Among the 401 patients included in the analysis, the median age was 6 (IQR, 1-12) years, and 222 (55.4%) were male. One hundred fifty-four patients (38.4%) were previously healthy, 108 (26.9%) were immunocompromised, and 225 (56.1%) had documented infection(s) at enrollment. Forty-four patients (11.0%) died in the PICU. Viral DNAemia with at least 1 virus (excluding TTV) was detected in 191 patients (47.6%) overall, 63 of 108 patients (58.3%) who were immunocompromised, and 128 of 293 (43.7%) who were not immunocompromised at sepsis onset. After adjustment for age, Pediatric Risk of Mortality score, previously healthy status, and immunocompromised status at sepsis onset, CMV (adjusted odds ratio [AOR], 3.01 [95% CI, 1.36-6.45]; P = .007), HAdV (AOR, 3.50 [95% CI, 1.46-8.09]; P = .006), BKPyV (AOR. 3.02 [95% CI, 1.17-7.34]; P = .02), and HHV-6 (AOR, 2.62 [95% CI, 1.31-5.20]; P = .007) DNAemia were each associated with increased mortality. Two or more viruses were detected in 78 patients (19.5%), with mortality among 12 of 32 (37.5%) who were immunocompromised and 9 of 46 (19.6%) who were not immunocompromised at sepsis onset. Herpesvirus seropositivity was common (HSV-1, 82 of 246 [33.3%]; CMV, 107 of 254 [42.1%]; EBV, 152 of 251 [60.6%]; HHV-6, 253 if 257 [98.4%]). After additional adjustment for receipt of blood products in the PICU, EBV seropositivity was associated with increased mortality (AOR, 6.10 [95% CI, 1.00-118.61]; P = .049). Conclusions and Relevance: The findings of this cohort study suggest that DNAemia for CMV, HAdV, BKPyV, and HHV-6 and EBV seropositivity were independently associated with increased sepsis mortality. Further investigation of the underlying biology of these viral DNA infections in children with sepsis is warranted to determine whether they only reflect mortality risk or contribute to mortality.


Asunto(s)
Infecciones por Citomegalovirus , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 1 , Sepsis , Adolescente , Humanos , Masculino , Niño , Lactante , Preescolar , Femenino , ADN Viral , Estudios de Cohortes , Herpesvirus Humano 4 , Virus ADN
2.
Pediatr Res ; 95(2): 418-435, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37225779

RESUMEN

BACKGROUND: With the emergence of newborn congenital cytomegalovirus (cCMV) screening programs, more infants are being diagnosed and require long-term follow-up. The objective of the study was to summarize the literature to date on neurodevelopmental outcomes in children with cCMV with attention to study-specific definitions of disease severity (symptomatic vs. asymptomatic). METHODS: This systematic scoping review included studies of children with cCMV (≤18 years-old) measuring neurodevelopment in ≥1 domain: global, gross motor, fine motor, speech/language, and intellectual/cognitive. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. PubMed, PsychInfo, and Embase databases were searched. RESULTS: 33 studies met inclusion criteria. Global development most frequently measured (n = 21), followed by cognitive/intellectual (n = 16) and speech/language (n = 8). Most (31/33) studies differentiated children by cCMV severity (symptomatic vs. asymptomatic), definitions of which ranged broadly. 15/21 studies described global development categorically (e.g., normal vs. abnormal). Across studies and domains, children with cCMV generally had equivalent or lower scores (vs. controls or normed measures). CONCLUSIONS: Variation in definitions of cCMV severity and blunt categorical outcomes may limit the generalizability of findings. Future studies should utilize standardized definitions of disease severity and in-depth measurement and reporting of neurodevelopmental outcomes in children with cCMV. IMPACT: Neurodevelopmental delays are common among children with cCMV, although gaps in the literature to have made quantification of such delays challenging. Variation in definitions of asymptomatic and symptomatic cCMV as well as the use of categorical outcomes of neurodevelopment (e.g., normal vs. abnormal) limits the generalizability and clinical utility of findings.


Asunto(s)
Infecciones por Citomegalovirus , Pérdida Auditiva Sensorineural , Lactante , Recién Nacido , Niño , Humanos , Adolescente , Pérdida Auditiva Sensorineural/diagnóstico , Citomegalovirus , Infecciones por Citomegalovirus/complicaciones , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/congénito , Tamizaje Neonatal
3.
Transplantation ; 107(8): 1820-1827, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36959162

RESUMEN

BACKGROUND: Solid organ transplant recipients are at high risk for Staphylococcus aureus bacteremia, but the risks before and after transplantation require further research. METHODS: We performed a population-based retrospective self-controlled study using the State Inpatient Database from 10 states in the United States. Adult and pediatric patients who had solid organ transplantation from 2004 to 2018 were tracked longitudinally for 1 y before and after transplantation outside of the immediate peritransplant periods. The exposure of interest was solid organ transplantation, and the outcome of interest was hospitalization with S. aureus bacteremia. RESULTS: Of 75 549 patients, 581 (0.77%) and 239 (0.32%) were hospitalized with S. aureus bacteremia in the pretransplant and posttransplant periods, respectively ( P < 0.001). Overall, the odds of hospitalization with S. aureus bacteremia increased from 7 to 12 mo to 1 to 6 mo before transplantation (odds ratio, 1.24; 95% confidence interval, 1.05-1.46) and then decreased following transplantation (odds ratio, 0.35; 95% confidence interval, 0.28-0.45; 7-12 mo after transplantation). The decreased rate after transplantation was driven by the cases associated with central line-associated bloodstream infections and endocarditis among kidney and heart transplant recipients. Odds of hospitalization with S. aureus bacteremia did not change after liver transplantation, whereas they increased after lung transplantation. CONCLUSIONS: In addition to immunosuppression, the reversal of organ failure and associated requirements for organ support following transplantation may play an important role in the risk of S. aureus bacteremia in solid organ transplant recipients. These results can guide infection prevention approaches and future research on S. aureus infections in transplant patients.


Asunto(s)
Bacteriemia , Trasplante de Órganos , Infecciones Estafilocócicas , Adulto , Humanos , Niño , Estados Unidos/epidemiología , Staphylococcus aureus , Estudios Retrospectivos , Trasplante de Órganos/efectos adversos , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/epidemiología , Bacteriemia/diagnóstico , Bacteriemia/epidemiología , Factores de Riesgo
4.
Trends Mol Med ; 29(1): 4-19, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36336610

RESUMEN

The consequences of human adenovirus (HAdV) infections are generally mild. However, despite the perception that HAdVs are harmless, infections can cause severe disease in certain individuals, including newborns, the immunocompromised, and those with pre-existing conditions, including respiratory or cardiac disease. In addition, HAdV outbreaks remain relatively common events and the recent emergence of more pathogenic genomic variants of various genotypes has been well documented. Coupled with evidence of zoonotic transmission, interspecies recombination, and the lack of approved AdV antivirals or widely available vaccines, HAdVs remain a threat to public health. At the same time, the detailed understanding of AdV biology garnered over nearly 7 decades of study has made this group of viruses a molecular workhorse for vaccine and gene therapy applications.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Infecciones del Sistema Respiratorio , Recién Nacido , Humanos , Adenoviridae/genética , Infecciones por Adenovirus Humanos/epidemiología , Adenovirus Humanos/genética , Genómica , Genotipo , Filogenia
6.
J Virol ; 96(6): e0170621, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35045262

RESUMEN

Bone marrow transplantation (BMT) recipients are at risk for substantial morbidity and mortality from human adenovirus infections, often in the setting of reactivation of persistent virus. Human adenovirus persistence in mucosal lymphocytes has been described, but specific cellular reservoirs of persistence and effects of persistence on host responses to unrelated stimuli are not completely understood. We used mouse adenovirus type 1 (MAV-1) to characterize persistence of an adenovirus in its natural host and test the hypothesis that persistence increases complications of BMT. Following intranasal infection of C57BL/6J mice, MAV-1 DNA was detected in lung, mediastinal lymph nodes, and liver during acute infection at 7 days postinfection (dpi), and at lower levels at 28 dpi that remained stable through 150 dpi. Expression of early and late viral transcripts was detected in those organs at 7 dpi but not at later time points. MAV-1 persistence was not affected by deficiency of IFN-γ. We detected no evidence of MAV-1 reactivation in vivo following allogeneic BMT of persistently infected mice. Persistent infection did not substantially affect mortality, weight loss, or pulmonary inflammation following BMT. However, T cell infiltration and increased expression of pro-inflammatory cytokines consistent with graft-versus-host disease (GVHD) were more pronounced in livers of persistently infected BMT mice than in uninfected BMT mice. These results suggest that MAV-1 persists in multiple sites without detectable evidence of ongoing replication. Our results indicate that MAV-1 persistence alters host responses to an unrelated challenge, even in the absence of detectable reactivation. IMPORTANCE Long-term persistence in an infected host is an essential step in the life cycle of DNA viruses. Adenoviruses persist in their host following acute infection, but the nature of adenovirus persistence remains incompletely understood. Following intranasal infection of mice, we found that MAV-1 persists for a prolonged period in multiple organs, although we did not detect evidence of ongoing replication. Because BMT recipients are at risk for substantial morbidity and mortality from human adenovirus infections, often in the setting of reactivation of persistent virus in the recipient, we extended our findings using MAV-1 infection in a mouse model of BMT. MAV-1 persistence exacerbated GVHD-like inflammation following allogeneic BMT, even in the absence of virus reactivation. This novel finding suggests that adenovirus persistence has consequences, and it highlights the potential for a persistent adenovirus to influence host responses to unrelated challenges.


Asunto(s)
Infecciones por Adenoviridae , Adenoviridae , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Adenoviridae/inmunología , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/fisiopatología , Infecciones por Adenoviridae/virología , Infecciones por Adenovirus Humanos , Animales , Trasplante de Médula Ósea/efectos adversos , Enfermedad Injerto contra Huésped/complicaciones , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/virología , Inflamación , Ratones , Ratones Endogámicos C57BL
7.
Front Immunol ; 12: 762789, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868007

RESUMEN

Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O2) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated development of bronchopulmonary dysplasia (BPD) in preterm infants. Therefore, we aimed to determine age-dependent differences in immune responses of monocyte-derived MФ comparing cord blood samples derived from preterm (n=14) and term (n=19) infants as well as peripheral blood samples from healthy adults (n=17) after lipopolysaccharide (LPS) exposure. Compared to term and adult MФ, LPS-stimulated preterm MФ showed an enhanced and sustained pro-inflammatory immune response determined by transcriptome analysis, cytokine release inducing a RORC upregulation due to T cell polarization of neonatal T cells, and TLR4 surface expression. In addition, a double-hit model was developed to study pulmonary relevant exposure factors by priming MФ with hyperoxia (O2 = 65%) or hypoxia (O2 = 3%) followed by lipopolysaccharide (LPS, 100ng/ml). When primed by 65% O2, subsequent LPS stimulation in preterm MФ led to an exaggerated pro-inflammatory response (e.g. increased HLA-DR expression and cytokine release) compared to LPS stimulation alone. Both, exposure to 65% or 3% O2 together with subsequent LPS stimulation, resulted in an exaggerated pro-inflammatory response of preterm MФ determined by transcriptome analysis. Downregulation of two major transcriptional factors, early growth response gene (Egr)-2 and growth factor independence 1 (Gfi1), were identified to play a role in the exaggerated pro-inflammatory response of preterm MФ to LPS insult after priming with 65% or 3% O2. Preterm MФ responses to LPS and hyperoxia/hypoxia suggest their involvement in excessive inflammation due to age-dependent differences, potentially mediated by downregulation of Egr2 and Gfi1 in the developing lung.


Asunto(s)
Recien Nacido Prematuro/inmunología , Inflamación/etiología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Oxígeno/farmacología , Adulto , Displasia Broncopulmonar/etiología , Citocinas/biosíntesis , Femenino , Perfilación de la Expresión Génica , Edad Gestacional , Humanos , Recién Nacido , Macrófagos/inmunología , Masculino , Receptor Toll-Like 4/fisiología
8.
Comp Med ; 71(5): 342-358, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34535198

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic was caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus has challenged civilization and modern science in ways that few infectious diseases and natural disasters have previously, causing globally significant human morbidity and mortality and triggering economic downturns across financial markets that will be dealt with for generations. Despite this, the pandemic has also brought an opportunity for humanity to come together and participate in a shared scientific investigation. Clinically, SARS-CoV-2 is associated with lower mortality rates than other recently emerged coronaviruses, such as SARS-CoV and the Middle East respiratory syndrome coronavirus (MERS-CoV). However, SARS-CoV-2 exhibits efficient human-to-human spread, with transmission often occurring before symptom recognition; this feature averts containment strategies that had worked previ- ously for SARS-CoV and MERS-CoV. Severe COVID-19 disease is characterized by dysregulated inflammatory responses associated with pulmonary congestion and intravascular coagulopathy leading to pneumonia, vascular insults, and multiorgan disease. Approaches to treatment have combined supportive care with antivirals, such as remdesivir, with immunomodulatory medications, including corticosteroids and cytokine-blocking antibody therapies; these treatments have advanced rapidly through clinical trials. Innovative approaches to vaccine development have facilitated rapid advances in design, testing, and distribution. Much remains to be learned about SARS-CoV-2 and COVID-19, and further biomedical research is necessary, including comparative medicine studies in animal models. This overview of COVID-19 in humans will highlight important aspects of disease, relevant pathophysiology, underlying immunology, and therapeutics that have been developed to date.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Animales , Antivirales , COVID-19/mortalidad , COVID-19/transmisión , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio , Pandemias , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , SARS-CoV-2
10.
Antiviral Res ; 188: 105034, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33577808

RESUMEN

Human adenoviruses (HAdV) are ubiquitous human pathogens that cause a significant burden of respiratory, ocular, and gastrointestinal illnesses. Although HAdV infections are generally self-limiting, pediatric and immunocompromised individuals are at particular risk for developing severe disease. Currently, no approved antiviral therapies specific to HAdV exist. Recent outbreaks underscore the need for effective antiviral agents to treat life-threatening infections. In this review we will focus on recent developments in search of potential therapeutic agents for controlling HAdV infections, with a focus on those targeting post-entry stages of the virus replicative cycle.


Asunto(s)
Infecciones por Adenovirus Humanos/tratamiento farmacológico , Adenovirus Humanos/efectos de los fármacos , Antivirales/uso terapéutico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/genética , Adenovirus Humanos/fisiología , Antivirales/farmacología , Replicación del ADN/efectos de los fármacos , Reposicionamiento de Medicamentos , Quimioterapia Combinada , Epigénesis Genética/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Inmunoterapia Adoptiva , Linfocitos T/inmunología , Replicación Viral/efectos de los fármacos
11.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641484

RESUMEN

Human adenoviruses (HAdV) are ubiquitous within the human population and comprise a significant burden of respiratory illnesses worldwide. Pediatric and immunocompromised individuals are at particular risk for developing severe disease; however, no approved antiviral therapies specific to HAdV exist. Ivermectin is an FDA-approved broad-spectrum antiparasitic drug that also exhibits antiviral properties against a diverse range of viruses. Its proposed function is inhibiting the classical protein nuclear import pathway mediated by importin-α (Imp-α) and -ß1 (Imp-ß1). Many viruses, including HAdV, rely on this host pathway for transport of viral proteins across the nuclear envelope. In this study, we show that ivermectin inhibits HAdV-C5 early gene transcription, early and late protein expression, genome replication, and production of infectious viral progeny. Similarly, ivermectin inhibits genome replication of HAdV-B3, a clinically important pathogen responsible for numerous recent outbreaks. Mechanistically, we show that ivermectin disrupts binding of the viral E1A protein to Imp-α without affecting the interaction between Imp-α and Imp-ß1. Our results further extend ivermectin's broad antiviral activity and provide a mechanistic underpinning for its mode of action as an inhibitor of cellular Imp-α/ß1-mediated nuclear import.IMPORTANCE Human adenoviruses (HAdVs) represent a ubiquitous and clinically important pathogen without an effective antiviral treatment. HAdV infections typically cause mild symptoms; however, individuals such as children, those with underlying conditions, and those with compromised immune systems can develop severe disseminated disease. Our results demonstrate that ivermectin, an FDA-approved antiparasitic agent, is effective at inhibiting replication of several HAdV types in vitro This is in agreement with the growing body of literature suggesting ivermectin has broad antiviral activity. This study expands our mechanistic knowledge of ivermectin by showing that ivermectin targets the ability of importin-α (Imp-α) to recognize nuclear localization sequences, without effecting the Imp-α/ß1 interaction. These data also exemplify the applicability of targeting host factors upon which viruses rely as a viable antiviral strategy.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Adenovirus Humanos/efectos de los fármacos , Antiparasitarios/farmacología , Ivermectina/farmacología , Replicación Viral/efectos de los fármacos , alfa Carioferinas/genética , beta Carioferinas/genética , Células A549 , Transporte Activo de Núcleo Celular/genética , Adenovirus Humanos/genética , Adenovirus Humanos/metabolismo , Adenovirus Humanos/patogenicidad , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/virología , Citosol/efectos de los fármacos , Citosol/metabolismo , Citosol/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Transducción de Señal , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo , alfa Carioferinas/antagonistas & inhibidores , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo
12.
Virology ; 547: 12-19, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32560900

RESUMEN

CD8 T cells contribute to effective clearance of mouse adenovirus type 1 (MAV-1) and to virus-induced pulmonary inflammation. We characterized effects of a CD8 T cell effector, TNF, on MAV-1 pathogenesis. TNF inhibited MAV-1 replication in vitro. TNF deficiency or immunoneutralization had no effect on lung viral loads or viral gene expression in mice infected intranasally with MAV-1. Absence of TNF delayed virus-induced weight loss and reduced histological evidence of pulmonary inflammation, although concentrations of proinflammatory cytokines and chemokines in bronchoalveolar lavage fluid (BALF) were not significantly affected. BALF concentrations of IL-10 were greater in TNF-deficient mice compared to controls. Our data indicate that TNF is not essential for control of viral replication in vivo, but virus-induced TNF contributes to some aspects of immunopathology and disease. Redundant CD8 T cell effectors and other aspects of immune function are sufficient for antiviral and pro-inflammatory responses to acute MAV-1 respiratory infection.


Asunto(s)
Infecciones por Adenoviridae/virología , Mastadenovirus/fisiología , Neumonía/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Replicación Viral , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Pulmón/inmunología , Pulmón/virología , Masculino , Mastadenovirus/genética , Ratones , Ratones Endogámicos C57BL , Neumonía/genética , Neumonía/virología , Factor de Necrosis Tumoral alfa/genética
13.
Radiol Imaging Cancer ; 2(6): e200058, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33778750

RESUMEN

Patients with cancer have been negatively impacted during the coronavirus disease 2019 (COVID-19) pandemic, as many of these individuals may be immunosuppressed and of older age. Additionally, cancer follow-up or imaging appointments have been delayed in many clinics around the world. Postponement of routine screening exams will result in delays in new cancer diagnoses. Clinics are continuing to monitor and adapt their appointment schedules based on local outbreaks of COVID-19. Studies on COVID-19 in patients with cancer are limited, but consistently indicate that this population is at risk for more severe COVID-19 illness. Data from recent studies also suggest that pediatric patients with cancer have a lower risk of severe COVID-19 illness compared to adults. Certain features of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection detected by lung, brain, and gastrointestinal imaging may confound radiologists' interpretation of cancer diagnosis, staging, and treatment response. Lastly, as clinics begin to re-open for routine appointments, protocols have been put in place to reduce SARS-CoV-2 exposure to patients during their visits. This review details different perspectives on the impact of the COVID-19 pandemic on patients with cancer and on cancer imaging. Keywords: Abdomen/GI, Cardiac, Infection, Nervous-Peripheral.


Asunto(s)
COVID-19/complicaciones , Diagnóstico por Imagen/métodos , Neoplasias/complicaciones , Neoplasias/diagnóstico por imagen , Atención al Paciente/métodos , Humanos , Pandemias , SARS-CoV-2
14.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092582

RESUMEN

Acute respiratory infection with mouse adenovirus type 1 (MAV-1) induces activity of the immunoproteasome, an inducible form of the proteasome that shapes CD8 T cell responses by enhancing peptide presentation by major histocompatibility complex (MHC) class I. We used mice deficient in all three immunoproteasome subunits (triple-knockout [TKO] mice) to determine whether immunoproteasome activity is essential for control of MAV-1 replication or inflammatory responses to acute infection. Complete immunoproteasome deficiency in adult TKO mice had no effect on MAV-1 replication, virus-induced lung inflammation, or adaptive immunity compared to C57BL/6 (B6) controls. In contrast, immunoproteasome deficiency in neonatal TKO mice was associated with decreased survival and decreased lung gamma interferon (IFN-γ) expression compared to B6 controls, although without substantial effects on viral replication, histological evidence of inflammation, or expression of the proinflammatory cytokines tumor necrosis factor alpha and interleukin-1ß in lungs or other organs. T cell recruitment and IFN-γ production was similar in lungs of infected B6 and TKO mice. In lungs of uninfected B6 mice, we detected low levels of immunoproteasome subunit mRNA and protein that increased with age. Immunoproteasome subunit expression was lower in lungs of adult IFN-γ-deficient mice compared to B6 controls. Together, these results demonstrate developmental regulation of the immunoproteasome that is associated with age-dependent differences in MAV-1 pathogenesis.IMPORTANCE MAV-1 infection is a useful model to study the pathogenesis of an adenovirus in its natural host. Host factors that control MAV-1 replication and contribute to inflammation and disease are not fully understood. The immunoproteasome is an inducible component of the ubiquitin proteasome system that shapes the repertoire of peptides presented by MHC class I to CD8 T cells, influences other aspects of T cell survival and activation, and promotes production of proinflammatory cytokines. We found that immunoproteasome activity is dispensable in adult mice. However, immunoproteasome deficiency in neonatal mice increased mortality and impaired IFN-γ responses in the lungs. Baseline immunoproteasome subunit expression in lungs of uninfected mice increased with age. Our findings suggest the existence of developmental regulation of the immunoproteasome, like other aspects of host immune function, and indicate that immunoproteasome activity is a critical protective factor early in life.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/patología , Factores de Edad , Linfocitos T CD8-positivos/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/patología , Animales , Modelos Animales de Enfermedad , Mastadenovirus/crecimiento & desarrollo , Mastadenovirus/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/deficiencia , Análisis de Supervivencia , Replicación Viral
15.
Virology ; 521: 129-137, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29908447

RESUMEN

CD8 T cells play a key role in clearance of mouse adenovirus type 1 (MAV-1) from the lung and contribute to virus-induced airway inflammation. We tested the hypothesis that interactions between Fas ligand (FasL) and Fas mediate the antiviral and proinflammatory effects of CD8 T cells. FasL and Fas expression were increased in the lungs of C57BL/6 (B6) mice during MAV-1 respiratory infection. Viral replication and weight loss were similar in B6 and Fas-deficient (lpr) mice. Histological evidence of pulmonary inflammation was similar in B6 and lpr mice, but lung mRNA levels and airway proinflammatory cytokine concentrations were lower in MAV-1-infected lpr mice compared to infected B6 mice. Virus-induced apoptosis in lungs was not affected by Fas deficiency. Our results suggest that the proinflammatory effects of CD8 T cells during MAV-1 infection are mediated in part by Fas activation and are distinct from CD8 T cell antiviral functions.


Asunto(s)
Infecciones por Adenoviridae/fisiopatología , Linfocitos T CD8-positivos/inmunología , Inflamación/fisiopatología , Infecciones del Sistema Respiratorio/fisiopatología , Receptor fas/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Proteína Ligando Fas/metabolismo , Perfilación de la Expresión Génica , Histocitoquímica , Inmunohistoquímica , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Carga Viral , Receptor fas/deficiencia
16.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L78-L86, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29565180

RESUMEN

Leptin is a pleiotropic hormone produced by white adipose tissue that regulates appetite and many physiological functions, including the immune response to infection. Genetic leptin deficiency in humans and mice impairs host defenses against respiratory tract infections. Since leptin deficiency is associated with obesity and other metabolic abnormalities, we generated mice that lack the leptin receptor (LepRb) in cells of the myeloid linage (LysM-LepRb-KO) to evaluate its impact in lean metabolically normal mice in a murine model of pneumococcal pneumonia. We observed higher lung and spleen bacterial burdens in LysM-LepRb-KO mice following an intratracheal challenge with Streptococcus pneumoniae. Although numbers of leukocytes recovered from bronchoalveolar lavage fluid did not differ between groups, we did observe higher levels of pulmonary IL-13 and TNFα in LysM-LepRb-KO mice 48 h post infection. Phagocytosis and killing of ingested S. pneumoniae were also impaired in alveolar macrophages (AMs) from LysM-LepRb-KO mice in vitro and were associated with reduced LTB4 and enhanced PGE2 synthesis in vitro. Pretreatment of AMs with LTB4 and the cyclooxygenase inhibitor, indomethacin, restored phagocytosis but not bacterial killing in vitro. These results confirm our previous observations in leptin-deficient ( ob/ob) and fasted mice and demonstrate that decreased leptin action, as opposed to metabolic irregularities associated with obesity or starvation, is responsible for the defective host defense against pneumococcal pneumonia. They also provide novel targets for therapeutic intervention in humans with bacterial pneumonia.


Asunto(s)
Pulmón/inmunología , Macrófagos/inmunología , Fagocitosis , Neumonía Neumocócica/inmunología , Receptores de Leptina/inmunología , Streptococcus pneumoniae/inmunología , Animales , Interleucina-13/genética , Interleucina-13/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Neumonía Neumocócica/genética , Neumonía Neumocócica/patología , Receptores de Leptina/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
17.
Virus Res ; 244: 90-98, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29141203

RESUMEN

Human adenovirus (HAdV) type 36 seropositivity has been linked to obesity in humans. That link is supported by a small number of studies using HAdV-36 infection of animals that are not natural hosts for HAdVs. In this study, we infected mice with mouse adenovirus type 1 (MAV-1), a mouse pathogen, to determine whether MAV-1 infected adipose tissue and was associated with adipose tissue inflammation and obesity. We detected MAV-1 in adipose tissue during acute MAV-1 infection, but we did not detect virus-induced increases in adipose tissue cytokine expression or histological evidence of adipose tissue inflammation during acute infection. MAV-1 did not persist in adipose tissue at later times, and we did not detect long-term adipose inflammation, increased adipose tissue mass, or body weight in infected mice. Our data indicate that MAV-1 is not associated with obesity in infected mice.


Asunto(s)
Infecciones por Adenoviridae/virología , Tejido Adiposo/virología , ADN Viral/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno , Mastadenovirus/genética , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Tejido Adiposo/inmunología , Animales , Peso Corporal , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/inmunología , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , ADN Viral/inmunología , Femenino , Fibroblastos/virología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Mastadenovirus/crecimiento & desarrollo , Mastadenovirus/metabolismo , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Replicación Viral
18.
Virology ; 507: 64-74, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28410483

RESUMEN

CD8 T cells are key components of the immune response to viruses, but their roles in the pathogenesis of adenovirus respiratory infection have not been characterized. We used mouse adenovirus type 1 (MAV-1) to define CD8 T cell contributions to the pathogenesis of adenovirus respiratory infection. CD8 T cell deficiency in ß2m-/- mice had no effect on peak viral replication in lungs, but clearance of virus was delayed in ß2m-/- mice. Virus-induced weight loss and increases in bronchoalveolar lavage fluid total protein, IFN-γ, TNF-α, IL-10, CCL2, and CCL5 concentrations were less in ß2m-/- mice than in controls. CD8 T cell depletion had similar effects on virus clearance, weight loss, and inflammation. Deficiency of IFN-γ or perforin had no effect on viral replication or inflammation, but perforin-deficient mice were partially protected from weight loss. CD8 T cells promote MAV-1-induced pulmonary inflammation via a mechanism that is independent of direct antiviral effects.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Mastadenovirus/fisiología , Enfermedades de los Roedores/inmunología , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/virología , Animales , Femenino , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Pulmón/virología , Masculino , Mastadenovirus/genética , Mastadenovirus/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Perforina/genética , Perforina/inmunología , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología , Replicación Viral
19.
Virology ; 498: 57-68, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27560373

RESUMEN

The immunoproteasome is an inducible host mechanism that aids in the clearance of damaged proteins. The immunoproteasome also influences immune function by enhancing peptide presentation by MHC class I and promotes inflammation via IκB degradation and activation of NF-κB. We used mouse adenovirus type 1 (MAV-1) to characterize the role of the immunoproteasome in adenovirus pathogenesis. Following intranasal infection of mice, immunoproteasome activity in the heart and lung was significantly increased in an IFN-γ-dependent manner. Absence of the ß5i immunoproteasome subunit and pharmacological inhibition of ß5i activity had minimal effects on viral replication, virus-induced cellular inflammation, or induction of cytokine expression. Likewise, the establishment of protective immunity following primary infection was not significantly altered by ß5i deficiency. Thus, although immunoproteasome activity is robustly induced during acute infection with MAV-1, our data suggest that other mechanisms are capable of compensating for immunoproteasome activity to maintain antiviral immunity and appropriate inflammatory responses.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/metabolismo , Adenoviridae/fisiología , Inmunomodulación , Interferones/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Infecciones por Adenoviridae/mortalidad , Infecciones por Adenoviridae/virología , Animales , Inmunomodulación/genética , Inmunomodulación/inmunología , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Interferones/genética , Ratones , Ratones Noqueados , Miocarditis/genética , Miocarditis/inmunología , Miocarditis/metabolismo , Miocarditis/virología , Complejo de la Endopetidasa Proteasomal/genética
20.
Sci Rep ; 6: 26836, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27241677

RESUMEN

Postpartum women are at increased risk of developing puerperal sepsis caused by group A Streptococcus (GAS). Specific GAS serotypes, including M1 and M28, are more commonly associated with puerperal sepsis. However, the mechanisms of GAS genital tract infection are not well understood. We utilized a murine genital tract carriage model to demonstrate that M1 and M28 GAS colonization triggers TNF-α, IL-1ß, and IL-17A production in the female genital tract. GAS-induced IL-17A significantly influences streptococcal carriage and alters local inflammatory responses in two genetically distinct inbred strains of mice. An absence of IL-17A or the IL-1 receptor was associated with reduced neutrophil recruitment to the site of infection; and clearance of GAS was significantly attenuated in IL-17A(-/-) mice and Rag1(-/-) mice (that lack mature lymphocytes) but not in mice deficient for the IL-1 receptor. Together, these findings support a role for IL-17A in contributing to the control of streptococcal mucosal colonization and provide new insight into the inflammatory mediators regulating host-pathogen interactions in the female genital tract.


Asunto(s)
Inflamación/inmunología , Interleucina-17/inmunología , Infecciones del Sistema Genital/inmunología , Infecciones del Sistema Genital/microbiología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Femenino , Inflamación/metabolismo , Inflamación/microbiología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Infecciones del Sistema Genital/metabolismo , Vagina/inmunología , Vagina/metabolismo , Vagina/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...