Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 222
1.
J Math Biol ; 89(1): 7, 2024 May 21.
Article En | MEDLINE | ID: mdl-38772937

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.


Malaria, Vivax , Mathematical Concepts , Mosquito Vectors , Plasmodium vivax , Superinfection , Humans , Plasmodium vivax/immunology , Plasmodium vivax/physiology , Superinfection/immunology , Superinfection/transmission , Superinfection/parasitology , Malaria, Vivax/transmission , Malaria, Vivax/immunology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Animals , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Disease Reservoirs/parasitology , Models, Biological , Computer Simulation , Anopheles/parasitology , Anopheles/immunology
2.
Virus Res ; 345: 199375, 2024 Jul.
Article En | MEDLINE | ID: mdl-38642618

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.


COVID-19 , Cytomegalovirus Infections , Cytomegalovirus , Lipopolysaccharide Receptors , Monocytes , SARS-CoV-2 , Superinfection , Humans , Monocytes/virology , Monocytes/immunology , Cytomegalovirus/immunology , Lipopolysaccharide Receptors/metabolism , SARS-CoV-2/immunology , COVID-19/virology , COVID-19/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/immunology , Superinfection/virology , Superinfection/immunology , Virus Latency , Inflammation , Coinfection/virology , Cytokines/metabolism , Virus Replication
3.
J Virol ; 97(2): e0142322, 2023 02 28.
Article En | MEDLINE | ID: mdl-36692289

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.


Cattle Diseases , Host Microbial Interactions , Mycoplasma Infections , Orthomyxoviridae Infections , Signal Transduction , Thogotovirus , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/virology , Lung/immunology , Lung/microbiology , Lung/virology , Mycoplasma bovis/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Signal Transduction/immunology , Superinfection/immunology , Superinfection/veterinary , Toll-Like Receptor 2 , Host Microbial Interactions/immunology , Mycoplasma Infections/immunology , Mycoplasma Infections/virology
5.
PLoS One ; 16(10): e0255309, 2021.
Article En | MEDLINE | ID: mdl-34618816

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.


Interferons/analysis , Interferons/immunology , Interleukins/immunology , Orthomyxoviridae Infections/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Cell Line , Coinfection/immunology , Coinfection/microbiology , Dogs , Female , Interferons/genetics , Interleukins/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Polymorphism, Genetic/genetics , Staphylococcal Infections/prevention & control , Superinfection/immunology , Superinfection/microbiology , Viral Load/immunology , Interferon Lambda
6.
Elife ; 102021 07 15.
Article En | MEDLINE | ID: mdl-34263727

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.


Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/isolation & purification , HIV Antibodies/immunology , HIV Infections/immunology , Polysaccharides/immunology , Superinfection/immunology , Broadly Neutralizing Antibodies/chemistry , Broadly Neutralizing Antibodies/genetics , Cryoelectron Microscopy , Epitopes/genetics , Epitopes/immunology , Female , HEK293 Cells , HIV-1 , Humans , Models, Molecular , Mutation , Polysaccharides/chemistry
7.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article En | MEDLINE | ID: mdl-34244443

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.


Microvirus/physiology , Superinfection/virology , Viral Proteins/chemistry , DNA, Viral/metabolism , Escherichia coli/virology , Immunity , Phylogeny , Prophages/physiology , Superinfection/immunology
8.
BMC Infect Dis ; 21(1): 537, 2021 Jun 07.
Article En | MEDLINE | ID: mdl-34098877

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.


Aspergillosis/diagnosis , Brain Neoplasms/diagnosis , Central Nervous System Fungal Infections/diagnosis , Superinfection/diagnosis , Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillosis/immunology , Aspergillosis/pathology , Aspergillus/isolation & purification , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Central Nervous System Fungal Infections/drug therapy , Central Nervous System Fungal Infections/immunology , Central Nervous System Fungal Infections/pathology , Diagnosis, Differential , Female , Humans , Immunocompromised Host , Middle Aged , Staphylococcus/isolation & purification , Superinfection/drug therapy , Superinfection/immunology , Superinfection/pathology
9.
Front Immunol ; 12: 652923, 2021.
Article En | MEDLINE | ID: mdl-34163470

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.


Escherichia coli Infections/veterinary , Ligilactobacillus salivarius/immunology , Probiotics/administration & dosage , Rotavirus Infections/veterinary , Superinfection/veterinary , Swine/immunology , Animal Feed/microbiology , Animals , Disease Models, Animal , Enterotoxigenic Escherichia coli/immunology , Enterotoxigenic Escherichia coli/pathogenicity , Epithelial Cells/immunology , Epithelial Cells/microbiology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Female , Immunity, Innate , Intestinal Mucosa/microbiology , Mice , Poly I-C/administration & dosage , Poly I-C/immunology , Rotavirus/immunology , Rotavirus/pathogenicity , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Superinfection/immunology , Superinfection/microbiology , Superinfection/prevention & control , Swine/microbiology , Undaria/immunology , Weaning
10.
PLoS Pathog ; 17(3): e1009405, 2021 03.
Article En | MEDLINE | ID: mdl-33690728

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.


Coinfection/immunology , Interferons/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Superinfection/immunology , Animals , Disease Susceptibility , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology
11.
J Microbiol Immunol Infect ; 54(1): 105-108, 2021 Feb.
Article En | MEDLINE | ID: mdl-32943328

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.


COVID-19/immunology , COVID-19/microbiology , Coinfection/immunology , Coinfection/virology , SARS-CoV-2/immunology , COVID-19/epidemiology , COVID-19/virology , Coinfection/epidemiology , Humans , Immunosuppression Therapy , Lymphopenia/immunology , Lymphopenia/microbiology , Lymphopenia/virology , Pandemics , Prevalence , Public Health , Superinfection/immunology , Superinfection/microbiology , Superinfection/virology
12.
PLoS Pathog ; 16(5): e1008393, 2020 05.
Article En | MEDLINE | ID: mdl-32433711

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.


Immunologic Memory/immunology , Influenza A virus/immunology , Superantigens/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Female , Humans , Immunologic Memory/physiology , Influenza A virus/metabolism , Influenza, Human/immunology , Influenza, Human/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Staphylococcus aureus/immunology , Superantigens/physiology , Superinfection/immunology , Vaccination
13.
J Leukoc Biol ; 108(3): 825-834, 2020 09.
Article En | MEDLINE | ID: mdl-32125045

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.


Immunity, Innate/immunology , Immunogenicity, Vaccine/immunology , Macrophages/immunology , Vaccines/immunology , Adaptive Immunity/immunology , Administration, Inhalation , Administration, Mucosal , Animals , BCG Vaccine/immunology , Humans , Immunologic Memory/immunology , Influenza, Human/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Immunological , Respiratory Mucosa/immunology , Superinfection/immunology , Tuberculosis/immunology , Vaccination/methods , Vaccines/administration & dosage
15.
JCI Insight ; 4(14)2019 07 25.
Article En | MEDLINE | ID: mdl-31341107

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.


Anti-Bacterial Agents/therapeutic use , Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Pneumonia, Staphylococcal/drug therapy , Superinfection/drug therapy , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antiviral Agents/pharmacology , Broadly Neutralizing Antibodies/pharmacology , Broadly Neutralizing Antibodies/therapeutic use , Disease Models, Animal , Drug Therapy, Combination/methods , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/immunology , Influenza, Human/mortality , Influenza, Human/virology , Lung/immunology , Lung/microbiology , Lung/virology , Macrophages/drug effects , Macrophages/immunology , Male , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Neutrophils/drug effects , Neutrophils/immunology , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/mortality , Superinfection/immunology , Superinfection/microbiology , Superinfection/mortality , Survival Analysis , Treatment Outcome
16.
mBio ; 10(3)2019 06 04.
Article En | MEDLINE | ID: mdl-31164468

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.


Evolution, Molecular , Mycobacteriophages/classification , Mycobacteriophages/immunology , Superinfection/immunology , Genome, Viral , Phylogeny , Prophages/genetics , Prophages/immunology , Virulence
18.
Front Immunol ; 10: 336, 2019.
Article En | MEDLINE | ID: mdl-30881357

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.


Bacterial Infections/immunology , Influenza, Human/immunology , Superinfection/immunology , T-Lymphocytes/immunology , Animals , Bacterial Infections/therapy , Humans , Influenza, Human/therapy , Superinfection/therapy
19.
Infect Dis (Lond) ; 51(1): 56-61, 2019 Jan.
Article En | MEDLINE | ID: mdl-30317905

BACKGROUND: The role of the anti-HIV neutralizing antibody response in protecting against HIV superinfection, and changes in neutralizing antibody potency and breadth after HIV superinfection have not been fully elucidated. This study examined the rate of HIV superinfection in men who have sex with men (MSM) also diagnosed with syphilis in Denmark, and the anti-HIV neutralizing antibody response in men who became superinfected. MATERIALS AND METHODS: MSM enrolled in the Danish HIV cohort who acquired syphilis were examined longitudinally for HIV superinfection using a validated next-generation sequencing assay. HIV superinfection cases were matched 3:1 to controls, and neutralizing antibody responses before (cases/controls) and after (cases) HIV superinfection were determined using a 20-pseudovirus panel. RESULTS: Four cases of HIV superinfection were identified from 95 MSM screened for a rate of HIV superinfection of 1.56/100 pys (95% CI = 0.43-4.01). Prior to HIV superinfection neutralizing antibody responses were low in breadth and potency, and did not differ between cases and controls (p = 1.0). In cases, neutralizing antibody responses increased modestly after HIV superinfection. CONCLUSIONS: These data support the theory that the natural neutralizing antibody response to HIV infection may not be the controlling factor in protecting against a subsequent HIV challenge.


Antibodies, Neutralizing/blood , HIV Antibodies/blood , HIV Infections/immunology , HIV/immunology , Homosexuality, Male , Denmark , HIV/classification , HIV/genetics , HIV Infections/virology , High-Throughput Nucleotide Sequencing , Humans , Longitudinal Studies , Male , Prospective Studies , Superinfection/immunology , Superinfection/virology , Syphilis/complications
20.
AIDS ; 33(3): 399-410, 2019 03 01.
Article En | MEDLINE | ID: mdl-30531316

OBJECTIVE: The aim of this study was to investigate the impact of intersubtype HIV-1 superinfection on viremia, reservoir reseeding, viral evolution and disease progression in HIV controllers (HIC). DESIGN: A longitudinal analysis of two Brazilian HIC individuals (EEC09 and VC32) previously identified as dually infected with subtypes B and F1 viruses. METHODS: Changes in plasma viremia, total HIV-1 DNA levels, CD4+ T-cell counts and HIV-1 quasispecies composition were measured over time. HIV-1 env diversity in peripheral blood mononuclear cell (PBMC) and plasma samples was accessed by single genome amplification and next-generation sequencing approaches, respectively. Viral evolution was evaluated by estimating nucleotide diversity and divergence. RESULTS: Individual EEC09 was probably initially infected with a CCR5-tropic subtype B strain and sequentially superinfected with a CXCR4-tropic subtype B strain and with a subtype F1 variant. Individual VC32 was infected with a subtype B strain and superinfected with a subtype F1 variant. The intersubtype superinfection events lead to a moderate increase in viremia and extensive turnover of viral population in plasma but exhibited divergent impact on the size and composition of cell-associated HIV DNA population. Both individuals maintained virologic control (<2000 copies/ml) and presented no evidence of viral evolution or immunologic progression for at least 2 years after the intersubtype superinfection event. CONCLUSION: These data revealed that some HIC are able to repeatedly limit replication and evolution of superinfecting viral strains of a different subtype with no signs of disease progression.


HIV Infections/immunology , HIV Infections/virology , HIV Long-Term Survivors , HIV-1/growth & development , Superinfection/immunology , Superinfection/virology , Virus Replication , Adult , Brazil , CD4 Lymphocyte Count , Disease Progression , Disease Reservoirs/virology , Genetic Variation , Genotype , HIV-1/classification , HIV-1/genetics , HIV-1/immunology , Humans , Longitudinal Studies , Male , Middle Aged , Viral Load , Viremia/virology , env Gene Products, Human Immunodeficiency Virus/genetics
...