Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 224
Filtrar
1.
Front Immunol ; 15: 1364622, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38933273

RESUMEN

Introduction: Community-acquired pneumonia (CAP) is a global health concern, with 25% of cases attributed to Streptococcus pneumoniae (Spn). Viral infections like influenza A virus (IAV), respiratory syncytial virus (RSV), and human metapneumovirus (hMPV) increase the risk of Spn, leading to severe complications due to compromised host immunity. Methods: We evaluated the efficacy of an anti-PhtD monoclonal antibody (mAb) cocktail therapy (PhtD3 + 7) in improving survival rates in three viral/bacterial coinfection models: IAV/Spn, hMPV/Spn, and RSV/Spn. Results: The PhtD3 + 7 mAb cocktail outperformed antiviral mAbs, resulting in prolonged survival. In the IAV/Spn model, it reduced bacterial titers in blood and lungs by 2-4 logs. In the hMPV/Spn model, PhtD3 + 7 provided greater protection than the hMPV-neutralizing mAb MPV467, significantly reducing bacterial titers. In the RSV/Spn model, PhtD3 + 7 offered slightly better protection than the antiviral mAb D25, uniquely decreasing bacterial titers in blood and lungs. Discussion: Given the threat of antibiotic resistance, our findings highlight the potential of anti-PhtD mAb therapy as an effective option for treating viral and secondary pneumococcal coinfections.


Asunto(s)
Anticuerpos Monoclonales , Coinfección , Streptococcus pneumoniae , Sobreinfección , Animales , Humanos , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/inmunología , Streptococcus pneumoniae/inmunología , Ratones , Sobreinfección/inmunología , Sobreinfección/microbiología , Coinfección/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Metapneumovirus/inmunología , Virus de la Influenza A/inmunología , Modelos Animales de Enfermedad , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/prevención & control , Infecciones Neumocócicas/tratamiento farmacológico , Femenino , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/tratamiento farmacológico , Anticuerpos Antivirales/inmunología
2.
J Math Biol ; 89(1): 7, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38772937

RESUMEN

Malaria is a vector-borne disease that exacts a grave toll in the Global South. The epidemiology of Plasmodium vivax, the most geographically expansive agent of human malaria, is characterised by the accrual of a reservoir of dormant parasites known as hypnozoites. Relapses, arising from hypnozoite activation events, comprise the majority of the blood-stage infection burden, with implications for the acquisition of immunity and the distribution of superinfection. Here, we construct a novel model for the transmission of P. vivax that concurrently accounts for the accrual of the hypnozoite reservoir, (blood-stage) superinfection and the acquisition of immunity. We begin by using an infinite-server queueing network model to characterise the within-host dynamics as a function of mosquito-to-human transmission intensity, extending our previous model to capture a discretised immunity level. To model transmission-blocking and antidisease immunity, we allow for geometric decay in the respective probabilities of successful human-to-mosquito transmission and symptomatic blood-stage infection as a function of this immunity level. Under a hybrid approximation-whereby probabilistic within-host distributions are cast as expected population-level proportions-we couple host and vector dynamics to recover a deterministic compartmental model in line with Ross-Macdonald theory. We then perform a steady-state analysis for this compartmental model, informed by the (analytic) distributions derived at the within-host level. To characterise transient dynamics, we derive a reduced system of integrodifferential equations, likewise informed by our within-host queueing network, allowing us to recover population-level distributions for various quantities of epidemiological interest. In capturing the interplay between hypnozoite accrual, superinfection and acquired immunity-and providing, to the best of our knowledge, the most complete population-level distributions for a range of epidemiological values-our model provides insights into important, but poorly understood, epidemiological features of P. vivax.


Asunto(s)
Malaria Vivax , Conceptos Matemáticos , Mosquitos Vectores , Plasmodium vivax , Sobreinfección , Humanos , Plasmodium vivax/inmunología , Plasmodium vivax/fisiología , Sobreinfección/inmunología , Sobreinfección/transmisión , Sobreinfección/parasitología , Malaria Vivax/transmisión , Malaria Vivax/inmunología , Malaria Vivax/epidemiología , Malaria Vivax/parasitología , Animales , Mosquitos Vectores/parasitología , Mosquitos Vectores/inmunología , Reservorios de Enfermedades/parasitología , Modelos Biológicos , Simulación por Computador , Anopheles/parasitología , Anopheles/inmunología
3.
Virus Res ; 345: 199375, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38642618

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), has posed significant challenges to global health. While much attention has been directed towards understanding the primary mechanisms of SARS-CoV-2 infection, emerging evidence suggests co-infections or superinfections with other viruses may contribute to increased morbidity and mortality, particularly in severe cases of COVID-19. Among viruses that have been reported in patients with SARS-CoV-2, seropositivity for Human cytomegalovirus (HCMV) is associated with increased COVID-19 risk and hospitalization. HCMV is a ubiquitous beta-herpesvirus with a seroprevalence of 60-90 % worldwide and one of the leading causes of mortality in immunocompromised individuals. The primary sites of latency for HCMV include CD14+ monocytes and CD34+ hematopoietic cells. In this study, we sought to investigate SARS-CoV-2 infection of CD14+ monocytes latently infected with HCMV. We demonstrate that CD14+ cells are susceptible and permissive to SARS-CoV-2 infection and detect subgenomic transcripts indicative of replication. To further investigate the molecular changes triggered by SARS-CoV-2 infection in HCMV-latent CD14+ monocytes, we conducted RNA sequencing coupled with bioinformatic differential gene analysis. The results revealed significant differences in cytokine-cytokine receptor interactions and inflammatory pathways in cells superinfected with replication-competent SARS-CoV-2 compared to the heat-inactivated and mock controls. Notably, there was a significant upregulation in transcripts associated with pro-inflammatory response factors and a decrease in anti-inflammatory factors. Taken together, these findings provide a basis for the heightened inflammatory response, offering potential avenues for targeted therapeutic interventions among HCMV-infected severe cases of COVID-19. SUMMARY: COVID-19 patients infected with secondary viruses have been associated with a higher prevalence of severe symptoms. Individuals seropositive for human cytomegalovirus (HCMV) infection are at an increased risk for severe COVID-19 disease and hospitalization. HCMV reactivation has been reported in severe COVID-19 cases with respiratory failure and could be the result of co-infection with SARS-CoV-2 and HCMV. In a cell culture model of superinfection, HCMV has previously been shown to increase infection of SARS-CoV-2 of epithelial cells by upregulating the human angiotensin-converting enzyme-2 (ACE2) receptor. In this study, we utilize CD14+ monocytes, a major cell type that harbors latent HCMV, to investigate co-infection of SARS-CoV-2 and HCMV. This study is a first step toward understanding the mechanism that may facilitate increased COVID-19 disease severity in patients infected with SARS-CoV-2 and HCMV.


Asunto(s)
COVID-19 , Infecciones por Citomegalovirus , Citomegalovirus , Receptores de Lipopolisacáridos , Monocitos , SARS-CoV-2 , Sobreinfección , Humanos , Monocitos/virología , Monocitos/inmunología , Citomegalovirus/inmunología , Receptores de Lipopolisacáridos/metabolismo , SARS-CoV-2/inmunología , COVID-19/virología , COVID-19/inmunología , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/inmunología , Sobreinfección/virología , Sobreinfección/inmunología , Latencia del Virus , Inflamación , Coinfección/virología , Citocinas/metabolismo , Replicación Viral
4.
Am J Respir Cell Mol Biol ; 71(1): 110-120, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38574279

RESUMEN

Immune activation is essential for lung control of viral and bacterial infection, but an overwhelming inflammatory response often leads to the onset of acute respiratory distress syndrome. IL-10 plays a crucial role in regulating the balance between antimicrobial immunity and immunopathology. In the present study, we investigated the role of IL-10 in acute lung injury induced by influenza A virus and methicillin-resistant Staphylococcus aureus coinfection. This unique coinfection model resembles patients with acute pneumonia undergoing appropriate antibiotic therapies. Using global IL-10 and IL-10 receptor gene-deficient mice, as well as in vivo neutralizing antibodies, we show that IL-10 deficiency promotes IFN-γ-dominant cytokine responses and triggers acute animal death. Interestingly, this extreme susceptibility is fully preventable by IFN-γ neutralization during coinfection. Further studies using mice with Il10ra deletion in selective myeloid subsets reveal that IL-10 primarily acts on mononuclear phagocytes to prevent IFN-γ/TNF-α hyperproduction and acute mortality. Importantly, this antiinflammatory IL-10 signaling is independent of its inhibitory effect on antiviral and antibacterial defense. Collectively, our results demonstrate a key mechanism of IL-10 in preventing hypercytokinemia and acute respiratory distress syndrome pathogenesis by counteracting the IFN-γ response.


Asunto(s)
Lesión Pulmonar Aguda , Modelos Animales de Enfermedad , Interferón gamma , Interleucina-10 , Sobreinfección , Animales , Interleucina-10/metabolismo , Interleucina-10/inmunología , Lesión Pulmonar Aguda/virología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/microbiología , Interferón gamma/metabolismo , Sobreinfección/inmunología , Sobreinfección/virología , Ratones , Ratones Endogámicos C57BL , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Coinfección/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/virología , Infecciones Estafilocócicas/inmunología , Ratones Noqueados , Virus de la Influenza A/inmunología , Pulmón/virología , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo
5.
J Virol ; 97(2): e0142322, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36692289

RESUMEN

Concurrent infections with multiple pathogens are often described in cattle with respiratory illness. However, how the host-pathogen interactions influence the clinical outcome has been only partially explored in this species. Influenza D virus (IDV) was discovered in 2011. Since then, IDV has been detected worldwide in different hosts. A significant association between IDV and bacterial pathogens in sick cattle was shown in epidemiological studies, especially with Mycoplasma bovis. In an experimental challenge, IDV aggravated M. bovis-induced pneumonia. However, the mechanisms through which IDV drives an increased susceptibility to bacterial superinfections remain unknown. Here, we used the organotypic lung model precision-cut lung slices to study the interplay between IDV and M. bovis coinfection. Our results show that a primary IDV infection promotes M. bovis superinfection by increasing the bacterial replication and the ultrastructural damages in lung pneumocytes. In our model, IDV impaired the innate immune response triggered by M. bovis by decreasing the expression of several proinflammatory cytokines and chemokines that are important for immune cell recruitment and the bacterial clearance. Stimulations with agonists of cytosolic helicases and Toll-like receptors (TLRs) revealed that a primary activation of RIG-I/MDA5 desensitizes the TLR2 activation, similar to what was observed with IDV infection. The cross talk between these two pattern recognition receptors leads to a nonadditive response, which alters the TLR2-mediated cascade that controls the bacterial infection. These results highlight innate immune mechanisms that were not described for cattle so far and improve our understanding of the bovine host-microbe interactions and IDV pathogenesis. IMPORTANCE Since the spread of the respiratory influenza D virus (IDV) infection to the cattle population, the question about the impact of this virus on bovine respiratory disease (BRD) remains still unanswered. Animals affected by BRD are often coinfected with multiple pathogens, especially viruses and bacteria. In particular, viruses are suspected to enhance secondary bacterial superinfections. Here, we use an ex vivo model of lung tissue to study the effects of IDV infection on bacterial superinfections. Our results show that IDV increases the susceptibility to the respiratory pathogen Mycoplasma bovis. In particular, IDV seems to activate immune pathways that inhibit the innate immune response against the bacteria. This may allow M. bovis to increase its proliferation and to delay its clearance from lung tissue. These results suggest that IDV could have a negative impact on the respiratory pathology of cattle.


Asunto(s)
Enfermedades de los Bovinos , Interacciones Microbiota-Huesped , Infecciones por Mycoplasma , Infecciones por Orthomyxoviridae , Transducción de Señal , Thogotovirus , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/virología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Mycoplasma bovis/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Transducción de Señal/inmunología , Sobreinfección/inmunología , Sobreinfección/veterinaria , Receptor Toll-Like 2 , Interacciones Microbiota-Huesped/inmunología , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/virología
7.
PLoS One ; 16(10): e0255309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34618816

RESUMEN

BACKGROUND: Type III interferon, or interferon lambda (IFNλ) is a crucial antiviral cytokine induced by influenza infection. While IFNλ is important for anti-viral host defense, published data demonstrate that IFNλ is pathogenic during influenza/bacterial super-infection. It is known that polymorphisms in specific IFNλ genes affect influenza responses, but the effect of IFNλ subtypes on bacterial super-infection is unknown. METHODS: Using an established model of influenza, Staphylococcus aureus super-infection, we studied IFNλ3-/- and control mice to model a physiologically relevant reduction in IFNλ and to address its role in super-infection. RESULTS: Surprisingly, IFNλ3-/- mice did not have significantly lower total IFNλ than co-housed controls, and displayed no change in viral or bacterial clearance. Importantly, both control and IFNλ3-/- mice displayed a positive correlation between viral burden and total IFNλ in the bronchoalveolar lavage during influenza/bacterial super-infection, suggesting that higher influenza viral burden drives a similar total IFNλ response regardless of IFNλ3 gene integrity. Interestingly, total IFNλ levels positively correlated with bacterial burden, while viral burden and bronchoalveolar lavage cellularity did not. CONCLUSIONS: These data suggest IFNλ2 can compensate for IFNλ3 to mount an effective antiviral and defense, revealing a functional redundancy in these highly similar IFNλ subtypes. Further, the IFNλ response to influenza, as opposed to changes in cellular inflammation or viral load, significantly correlates with susceptibility to bacterial super-infection. Moreover, the IFNλ response is regulated and involves redundant subtypes, suggesting it is of high importance to pulmonary pathogen defense.


Asunto(s)
Interferones/análisis , Interferones/inmunología , Interleucinas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Línea Celular , Coinfección/inmunología , Coinfección/microbiología , Perros , Femenino , Interferones/genética , Interleucinas/genética , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología , Polimorfismo Genético/genética , Infecciones Estafilocócicas/prevención & control , Sobreinfección/inmunología , Sobreinfección/microbiología , Carga Viral/inmunología , Interferón lambda
8.
Elife ; 102021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34263727

RESUMEN

Stimulating broadly neutralizing antibodies (bnAbs) directly from germline remains a barrier for HIV vaccines. HIV superinfection elicits bnAbs more frequently than single infection, providing clues of how to elicit such responses. We used longitudinal antibody sequencing and structural studies to characterize bnAb development from a superinfection case. BnAb QA013.2 bound initial and superinfecting viral Env, despite its probable naive progenitor only recognizing the superinfecting strain, suggesting both viruses influenced this lineage. A 4.15 Å cryo-EM structure of QA013.2 bound to native-like trimer showed recognition of V3 signatures (N301/N332 and GDIR). QA013.2 relies less on CDRH3 and more on framework and CDRH1 for affinity and breadth compared to other V3/glycan-specific bnAbs. Antigenic profiling revealed that viral escape was achieved by changes in the structurally-defined epitope and by mutations in V1. These results highlight shared and novel properties of QA013.2 relative to other V3/glycan-specific bnAbs in the setting of sequential, diverse antigens.


Asunto(s)
Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/aislamiento & purificación , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , Polisacáridos/inmunología , Sobreinfección/inmunología , Anticuerpos ampliamente neutralizantes/química , Anticuerpos ampliamente neutralizantes/genética , Microscopía por Crioelectrón , Epítopos/genética , Epítopos/inmunología , Femenino , Células HEK293 , VIH-1 , Humanos , Modelos Moleculares , Mutación , Polisacáridos/química
9.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34244443

RESUMEN

Single-stranded DNA phages of the family Microviridae have fundamentally different evolutionary origins and dynamics than the more frequently studied double-stranded DNA phages. Despite their small size (around 5 kb), which imposes extreme constraints on genomic innovation, they have adapted to become prominent members of viromes in numerous ecosystems and hold a dominant position among viruses in the human gut. We show that multiple, divergent lineages in the family Microviridae have independently become capable of lysogenizing hosts and have convergently developed hypervariable regions in their DNA pilot protein, which is responsible for injecting the phage genome into the host. By creating microviruses with combinations of genomic segments from different phages and infecting Escherichia coli as a model system, we demonstrate that this hypervariable region confers the ability of temperate Microviridae to prevent DNA injection and infection by other microviruses. The DNA pilot protein is present in most microviruses, but has been recruited repeatedly into this additional role as microviruses altered their lifestyle by evolving the ability to integrate in bacterial genomes, which linked their survival to that of their hosts. Our results emphasize that competition between viruses is a considerable and often overlooked source of selective pressure, and by producing similar evolutionary outcomes in distinct lineages, it underlies the prevalence of hypervariable regions in the genomes of microviruses and perhaps beyond.


Asunto(s)
Microvirus/fisiología , Sobreinfección/virología , Proteínas Virales/química , ADN Viral/metabolismo , Escherichia coli/virología , Inmunidad , Filogenia , Profagos/fisiología , Sobreinfección/inmunología
10.
BMC Infect Dis ; 21(1): 537, 2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-34098877

RESUMEN

BACKGROUND: Invasive aspergillosis of the central nervous system is a rare but increasingly prevalent disease. We present the unusual case of an immunosuppressed patient suffering from unexpected superinfected invasive aspergillosis with cerebral, pulmonal, and adrenal manifestations, mimicking a metastasized bronchial carcinoma. This report reveals the importance of including aspergillosis in the differential diagnosis of a cerebral mass lesion in the light of unspecific clinical findings. CASE PRESENTATION: A 58-year-old immunocompromised female presented to our emergency department with a single tonic-clonic seizure. Imaging showed a ring enhancing cerebral mass with perifocal edema and evidence of two smaller additional hemorrhagic cerebral lesions. In the setting of a mass lesion in the lung, and additional nodular lesions in the left adrenal gland the diagnosis of a metastasized bronchus carcinoma was suspected and the cerebral mass resected. However, histology did not reveal any evidence for a neoplastic lesion but septate hyphae consistent with aspergillus instead and microbiological cultures confirmed concomitant staphylococcal infection. CONCLUSIONS: A high index of suspicion for aspergillus infection should be maintained in the setting of immunosuppression. Clinical and radiological findings are often unspecific and even misleading. Definite confirmation usually relies on tissue diagnosis with histochemical stains. Surgical resection is crucial for establishing the diagnosis and guiding therapy with targeted antifungal medications.


Asunto(s)
Aspergilosis/diagnóstico , Neoplasias Encefálicas/diagnóstico , Infecciones Fúngicas del Sistema Nervioso Central/diagnóstico , Sobreinfección/diagnóstico , Antifúngicos/uso terapéutico , Aspergilosis/tratamiento farmacológico , Aspergilosis/inmunología , Aspergilosis/patología , Aspergillus/aislamiento & purificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Infecciones Fúngicas del Sistema Nervioso Central/tratamiento farmacológico , Infecciones Fúngicas del Sistema Nervioso Central/inmunología , Infecciones Fúngicas del Sistema Nervioso Central/patología , Diagnóstico Diferencial , Femenino , Humanos , Huésped Inmunocomprometido , Persona de Mediana Edad , Staphylococcus/aislamiento & purificación , Sobreinfección/tratamiento farmacológico , Sobreinfección/inmunología , Sobreinfección/patología
11.
Front Immunol ; 12: 652923, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34163470

RESUMEN

Previously, we constructed a library of Ligilactobacillus salivarius strains from the intestine of wakame-fed pigs and reported a strain-dependent capacity to modulate IFN-ß expression in porcine intestinal epithelial (PIE) cells. In this work, we further characterized the immunomodulatory activities of L. salivarius strains from wakame-fed pigs by evaluating their ability to modulate TLR3- and TLR4-mediated innate immune responses in PIE cells. Two strains with a remarkable immunomodulatory potential were selected: L. salivarius FFIG35 and FFIG58. Both strains improved IFN-ß, IFN-λ and antiviral factors expression in PIE cells after TLR3 activation, which correlated with an enhanced resistance to rotavirus infection. Moreover, a model of enterotoxigenic E. coli (ETEC)/rotavirus superinfection in PIE cells was developed. Cells were more susceptible to rotavirus infection when the challenge occurred in conjunction with ETEC compared to the virus alone. However, L. salivarius FFIG35 and FFIG58 maintained their ability to enhance IFN-ß, IFN-λ and antiviral factors expression in PIE cells, and to reduce rotavirus replication in the context of superinfection. We also demonstrated that FFIG35 and FFIG58 strains regulated the immune response of PIE cells to rotavirus challenge or ETEC/rotavirus superinfection through the modulation of negative regulators of the TLR signaling pathway. In vivo studies performed in mice models confirmed the ability of L. salivarius FFIG58 to beneficially modulate the innate immune response and protect against ETEC infection. The results of this work contribute to the understanding of beneficial lactobacilli interactions with epithelial cells and allow us to hypothesize that the FFIG35 or FFIG58 strains could be used for the development of highly efficient functional feed to improve immune health status and reduce the severity of intestinal infections and superinfections in weaned piglets.


Asunto(s)
Infecciones por Escherichia coli/veterinaria , Ligilactobacillus salivarius/inmunología , Probióticos/administración & dosificación , Infecciones por Rotavirus/veterinaria , Sobreinfección/veterinaria , Porcinos/inmunología , Alimentación Animal/microbiología , Animales , Modelos Animales de Enfermedad , Escherichia coli Enterotoxigénica/inmunología , Escherichia coli Enterotoxigénica/patogenicidad , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Femenino , Inmunidad Innata , Mucosa Intestinal/microbiología , Ratones , Poli I-C/administración & dosificación , Poli I-C/inmunología , Rotavirus/inmunología , Rotavirus/patogenicidad , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/prevención & control , Infecciones por Rotavirus/virología , Sobreinfección/inmunología , Sobreinfección/microbiología , Sobreinfección/prevención & control , Porcinos/microbiología , Undaria/inmunología , Destete
12.
PLoS Pathog ; 17(3): e1009405, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33690728

RESUMEN

Bacterial co-infections represent a major clinical complication of influenza. Host-derived interferon (IFN) increases susceptibility to bacterial infections following influenza, but the relative roles of type-I versus type-II IFN remain poorly understood. We have used novel mouse models of co-infection in which colonizing pneumococci were inoculated into the upper respiratory tract; subsequent sublethal influenza virus infection caused the bacteria to enter the lungs and mediate lethal disease. Compared to wild-type mice or mice deficient in only one pathway, mice lacking both IFN pathways demonstrated the least amount of lung tissue damage and mortality following pneumococcal-influenza virus superinfection. Therapeutic neutralization of both type-I and type-II IFN pathways similarly provided optimal protection to co-infected wild-type mice. The most effective treatment regimen was staggered neutralization of the type-I IFN pathway early during co-infection combined with later neutralization of type-II IFN, which was consistent with the expression and reported activities of these IFNs during superinfection. These results are the first to directly compare the activities of type-I and type-II IFN during superinfection and provide new insights into potential host-directed targets for treatment of secondary bacterial infections during influenza.


Asunto(s)
Coinfección/inmunología , Interferones/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Neumocócica/inmunología , Sobreinfección/inmunología , Animales , Susceptibilidad a Enfermedades , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
13.
J Microbiol Immunol Infect ; 54(1): 105-108, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32943328

RESUMEN

Cases of co-infection and secondary infection emerging during the current Coronavirus Disease-19 (COVID-19) pandemic are a major public health concern. Such cases may result from immunodysregulation induced by the SARS-CoV-2 virus. Pandemic preparedness must include identification of disease natural history and common secondary infections to implement clinical solutions.


Asunto(s)
COVID-19/inmunología , COVID-19/microbiología , Coinfección/inmunología , Coinfección/virología , SARS-CoV-2/inmunología , COVID-19/epidemiología , COVID-19/virología , Coinfección/epidemiología , Humanos , Terapia de Inmunosupresión , Linfopenia/inmunología , Linfopenia/microbiología , Linfopenia/virología , Pandemias , Prevalencia , Salud Pública , Sobreinfección/inmunología , Sobreinfección/microbiología , Sobreinfección/virología
14.
PLoS Pathog ; 16(5): e1008393, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32433711

RESUMEN

Infection with (SAg)-producing bacteria may precede or follow infection with or vaccination against influenza A viruses (IAVs). However, how SAgs alter the breadth of IAV-specific CD8+ T cell (TCD8) responses is unknown. Moreover, whether recall responses mediating heterosubtypic immunity to IAVs are manipulated by SAgs remains unexplored. We employed wild-type (WT) and mutant bacterial SAgs, SAg-sufficient/deficient Staphylococcus aureus strains, and WT, mouse-adapted and reassortant IAV strains in multiple in vivo settings to address the above questions. Contrary to the popular view that SAgs delete or anergize T cells, systemic administration of staphylococcal enterotoxin B (SEB) or Mycoplasma arthritidis mitogen before intraperitoneal IAV immunization enlarged the clonal size of 'select' IAV-specific TCD8 and reshuffled the hierarchical pattern of primary TCD8 responses. This was mechanistically linked to the TCR Vß makeup of the impacted clones rather than their immunodominance status. Importantly, SAg-expanded TCD8 retained their IFN-γ production and cognate cytolytic capacities. The enhancing effect of SEB on immunodominant TCD8 was also evident in primary responses to vaccination with heat-inactivated and live attenuated IAV strains administered intramuscularly and intranasally, respectively. Interestingly, in prime-boost immunization settings, the outcome of SEB administration depended strictly upon the time point at which this SAg was introduced. Accordingly, SEB injection before priming raised CD127highKLRG1low memory precursor frequencies and augmented the anamnestic responses of SEB-binding TCD8. By comparison, introducing SEB before boosting diminished recall responses to IAV-derived epitopes drastically and indiscriminately. This was accompanied by lower Ki67 and higher Fas, LAG-3 and PD-1 levels consistent with a pro-apoptotic and/or exhausted phenotype. Therefore, SAgs can have contrasting impacts on anti-IAV immunity depending on the naïve/memory status and the TCR composition of exposed TCD8. Finally, local administration of SEB or infection with SEB-producing S. aureus enhanced pulmonary TCD8 responses to IAV. Our findings have clear implications for superinfections and prophylactic vaccination.


Asunto(s)
Memoria Inmunológica/inmunología , Virus de la Influenza A/inmunología , Superantígenos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Epítopos/inmunología , Femenino , Humanos , Memoria Inmunológica/fisiología , Virus de la Influenza A/metabolismo , Gripe Humana/inmunología , Gripe Humana/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Staphylococcus aureus/inmunología , Superantígenos/fisiología , Sobreinfección/inmunología , Vacunación
15.
J Leukoc Biol ; 108(3): 825-834, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32125045

RESUMEN

In the past few years, our understanding of immunological memory has evolved remarkably due to a growing body of new knowledge in innate immune memory and immunity. Immunological memory now encompasses both innate and adaptive immune memory. The hypo-reactive and hyper-reactive types of innate immune memory lead to a suppressed and enhanced innate immune protective outcome, respectively. The latter is also named trained innate immunity (TII). The emerging information on innate immune memory has not only shed new light on the mechanisms of host defense but is also revolutionizing our long-held view of vaccination and vaccine strategies. Our current review will examine recent progress and knowledge gaps in innate immune memory with a focus on tissue-resident Mϕs, particularly lung Mϕs, and their relationship to local antimicrobial innate immunity. We will also discuss the impact of innate immune memory and TII on our understanding of vaccine concept and strategies and the significance of respiratory mucosal route of vaccination against respiratory pathogens.


Asunto(s)
Inmunidad Innata/inmunología , Inmunogenicidad Vacunal/inmunología , Macrófagos/inmunología , Vacunas/inmunología , Inmunidad Adaptativa/inmunología , Administración por Inhalación , Administración a través de la Mucosa , Animales , Vacuna BCG/inmunología , Humanos , Memoria Inmunológica/inmunología , Gripe Humana/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Inmunológicos , Mucosa Respiratoria/inmunología , Sobreinfección/inmunología , Tuberculosis/inmunología , Vacunación/métodos , Vacunas/administración & dosificación
17.
JCI Insight ; 4(14)2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31341107

RESUMEN

Postinfluenza bacterial superinfections cause increased morbidity and mortality compared with singular infection with influenza during both pandemics and seasonal epidemics. Vaccines and current treatments provide limited benefit, a rationale to conduct studies utilizing alternative therapies. FY1 and an optimized version, MEDI8852, anti-influenza HA mAbs, have been shown to neutralize influenza virus during singular influenza infection. MEDI4893*, an anti-Staphylococcus aureus α-toxin mAb, has been shown to improve survival when administered prophylactically prior to S. aureus pneumonia. Our objective was to determine if mAbs can improve survival during postinfluenza bacterial pneumonia. We administered FY1 in a murine model of postinfluenza methicillin-resistant S. aureus (MRSA) pneumonia and observed improved survival rates when given early during the course of influenza infection. Our findings indicate decreased lung injury and increased uptake and binding of bacteria by macrophages in the mice that received FY1 earlier in the course of influenza infection, corresponding to decreased bacterial burden. We also observed improved survival when mice were treated with a combination of FY1 and MEDI4893* late during the course of postinfluenza MRSA pneumonia. In conclusion, both FY1 and MEDI4893* prolong survival when used in a murine model of postinfluenza MRSA pneumonia, suggesting pathogen-specific mAbs as a possible therapeutic in the context of bacterial superinfection.


Asunto(s)
Antibacterianos/uso terapéutico , Antivirales/uso terapéutico , Gripe Humana/tratamiento farmacológico , Neumonía Estafilocócica/tratamiento farmacológico , Sobreinfección/tratamiento farmacológico , Animales , Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antivirales/farmacología , Anticuerpos ampliamente neutralizantes/farmacología , Anticuerpos ampliamente neutralizantes/uso terapéutico , Modelos Animales de Enfermedad , Quimioterapia Combinada/métodos , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/inmunología , Gripe Humana/mortalidad , Gripe Humana/virología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/mortalidad , Sobreinfección/inmunología , Sobreinfección/microbiología , Sobreinfección/mortalidad , Análisis de Supervivencia , Resultado del Tratamiento
18.
mBio ; 10(3)2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31164468

RESUMEN

Temperate phages encode an immunity system to control lytic gene expression during lysogeny. This gene regulatory circuit consists of multiple interacting genetic elements, and although it is essential for controlling phage growth, it is subject to conflicting evolutionary pressures. During superinfection of a lysogen, the prophage's circuit interacts with the superinfecting phage's circuit and prevents lytic growth if the two circuits are closely related. The circuitry is advantageous since it provides the prophage with a defense mechanism, but the circuitry is also disadvantageous since it limits the phage's host range during superinfection. Evolutionarily related phages have divergent, orthogonal immunity systems that no longer interact and are heteroimmune, but we do not understand how immunity systems evolve new specificities. Here, we use a group of Cluster A mycobacteriophages that exhibit a spectrum of genetic diversity to examine how immunity system evolution impacts superinfection immunity. We show that phages with mesotypic (i.e., genetically related but distinct) immunity systems exhibit asymmetric and incomplete superinfection phenotypes. They form complex immunity networks instead of well-defined immunity groups, and mutations conferring escape (i.e., virulence) from homotypic or mesotypic immunity have various escape specificities. Thus, virulence and the evolution of new immune specificities are shaped by interactions with homotypic and mesotypic immunity systems.IMPORTANCE Many aspects regarding superinfection, immunity, virulence, and the evolution of immune specificities are poorly understood due to the lack of large collections of isolated and sequenced phages with a spectrum of genetic diversity. Using a genetically diverse collection of Cluster A phages, we show that the classical and relatively straightforward patterns of homoimmunity, heteroimmunity, and virulence result from interactions between homotypic and heterotypic phages at the extreme edges of an evolutionary continuum of immune specificities. Genetic interactions between mesotypic phages result in more complex mesoimmunity phenotypes and virulence profiles. These results highlight that the evolution of immune specificities can be shaped by homotypic and mesotypic interactions and may be more dynamic than previously considered.


Asunto(s)
Evolución Molecular , Micobacteriófagos/clasificación , Micobacteriófagos/inmunología , Sobreinfección/inmunología , Genoma Viral , Filogenia , Profagos/genética , Profagos/inmunología , Virulencia
20.
Front Immunol ; 10: 336, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30881357

RESUMEN

Despite the widespread application of vaccination programs and antiviral drug treatments, influenza viruses are still among the most harmful human pathogens. Indeed, influenza results in significant seasonal and pandemic morbidity and mortality. Furthermore, severe bacterial infections can occur in the aftermath of influenza virus infection, and contribute substantially to the excess morbidity and mortality associated with influenza. Here, we review the main features of influenza viruses and current knowledge about the mechanical and immune mechanisms that underlie post-influenza secondary bacterial infections. We present the emerging literature describing the role of "innate-like" unconventional T cells in post-influenza bacterial superinfection. Unconventional T cell populations span the border between the innate and adaptive arms of the immune system, and are prevalent in mucosal tissues (including the airways). They mainly comprise Natural Killer T cells, mucosal-associated invariant T cells and γδ T cells. We provide an overview of the principal functions that these cells play in pulmonary barrier functions and immunity, highlighting their unique ability to sense environmental factors and promote protection against respiratory bacterial infections. We focus on two major opportunistic pathogens involved in superinfections, namely Streptococcus pneumoniae and Staphylococcus aureus. We discuss mechanisms through which influenza viruses alter the antibacterial activity of unconventional T cells. Lastly, we discuss recent fundamental advances and possible therapeutic approaches in which unconventional T cells would be targeted to prevent post-influenza bacterial superinfections.


Asunto(s)
Infecciones Bacterianas/inmunología , Gripe Humana/inmunología , Sobreinfección/inmunología , Linfocitos T/inmunología , Animales , Infecciones Bacterianas/terapia , Humanos , Gripe Humana/terapia , Sobreinfección/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...