Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.965
Filtrar
1.
Biomedica ; 44(3): 328-339, 2024 08 29.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-39241240

RESUMO

Introduction: Data on the prevalence of fungal coinfections/superinfections in patients with COVID-19 are limited. Objective: To describe the prevalence of fungal coinfections/superinfections in patients with COVID-19, as well as risk factors and demographic, clinical, and microbiological characteristics. Material and methods: We included patients with a confirmed COVID-19 diagnosis and a confirmed fungal infection hospitalized in the ICU from March 2020 to December 2021. We collected data on age, sex, comorbidities, hospital length of stay (days), laboratory (ferritin) and microbiological results, treatment for COVID-19, antifungal therapy, and outcomes obtained from the clinical records. Results: Only 11 out of 740 patients met the inclusion criteria. The coinfection rate was 0.3% and the superinfection was 1.2%. The most affected population was male adults. The coinfections/superinfections diagnosed were candiduria and candidemia, caused by Candida albicans, C. tropicalis, C. glabrata, C. lusitaniae, and Kluyveromyces marxianus (C. kefyr). In addition, tracheobronchitis due to Aspergillus fumigatus was found. The most used antifungals were fluconazole and caspofungin. The lethality in patients with fungal coinfections was 50% and superinfections, 22%. The length of hospital stay was 11-65 days. Eight patients required mechanical ventilation and six received corticosteroids. The main comorbidity was diabetes mellitus (81.8%). Conclusions: The rate of fungal coinfections/superinfections in COVID-19 patients was low, but the lethality found urges for routine fungal screening in patients with severe COVID-19 to timely detect fungal infections that may further compromise the patient's life.


Introducción: Los datos sobre la prevalencia de coinfecciones o sobreinfecciones fúngicas en pacientes con COVID-19 son limitados. Objetivo: Describir la prevalencia de coinfecciones o sobreinfecciones fúngicas en pacientes con COVID-19, así como los factores de riesgo y las características demográficas, clínicas y microbiológicas. Material y métodos: Se incluyeron pacientes con diagnóstico confirmado de COVID-19, hospitalizados en la unidad de cuidados intensivos y con infección fúngica confirmada entre marzo del 2020 y diciembre del 2021. Del expediente clínico se obtuvieron datos sobre edad, sexo, comorbilidades, días de estancia hospitalaria, resultados de laboratorio (ferritina) y microbiológicos, tratamiento contra COVID-19, terapia antifúngica y desenlace. Resultados: Once de 740 pacientes cumplieron con los criterios de inclusión. La tasa de coinfección fue del 0,3 % y la de sobreinfección fue del 1,2 %. La población más afectada fue la de hombres adultos. Las coinfecciones o sobreinfecciones diagnosticadas fueron candiduria y candidemia, causadas por Candida albicans, C. tropicalis, C. glabrata, C. lusitaniae y Kluyveromyces marxianus (C. kefyr). Además, se encontró una traqueobronquitis por Aspergillus fumigatus. Los antifúngicos más administrados fueron fluconazol y caspofungina. La letalidad en pacientes con coinfecciones fue del 50 % y con sobreinfecciones fúngicas, del 22 %. El tiempo de estancia intrahospitalaria fue de 11 a 65 días. Ocho de los pacientes requirieron asistencia respiratoria mecánica y seis recibieron corticoides. La principal comorbilidad fue diabetes mellitus (81,8 %). Conclusiones: La tasa de coinfecciones o sobreinfecciones por hongos en pacientes con COVID-19 fue baja, pero la letalidad de estas requiere, con urgencia, la realización de pruebas de rutina para detectar hongos en pacientes con COVID-19 grave para diagnosticar oportunamente infecciones fúngicas que puedan comprometer aún más la vida del paciente.


Assuntos
COVID-19 , Coinfecção , Superinfecção , Centros de Atenção Terciária , Humanos , Masculino , Coinfecção/epidemiologia , México/epidemiologia , COVID-19/epidemiologia , COVID-19/complicações , Superinfecção/epidemiologia , Feminino , Pessoa de Meia-Idade , Adulto , Idoso , Antifúngicos/uso terapêutico , Micoses/epidemiologia , Micoses/tratamento farmacológico , Micoses/diagnóstico , Prevalência , Fatores de Risco , Comorbidade , Tempo de Internação/estatística & dados numéricos , SARS-CoV-2 , Estudos Retrospectivos
2.
J Gen Virol ; 105(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39189607

RESUMO

West Nile virus (WNV) is the leading cause of mosquito-borne illness in the USA. There are currently no human vaccines or therapies available for WNV, and vector control is the primary strategy used to control WNV transmission. The WNV vector Culex tarsalis is also a competent host for the insect-specific virus (ISV) Eilat virus (EILV). ISVs such as EILV can interact with and cause superinfection exclusion (SIE) against human pathogenic viruses in their shared mosquito host, altering vector competence for these pathogenic viruses. The ability to cause SIE and their host restriction make ISVs a potentially safe tool to target mosquito-borne pathogenic viruses. In the present study, we tested whether EILV causes SIE against WNV in mosquito C6/36 cells and C. tarsalis mosquitoes. The titres of both WNV strains - WN02-1956 and NY99 - were suppressed by EILV in C6/36 cells as early as 48-72 h post-superinfection at both m.o.i. values tested in our study. The titres of WN02-1956 at both m.o.i. values remained suppressed in C6/36 cells, whereas those of NY99 showed some recovery towards the final timepoint. The mechanism of SIE remains unknown, but EILV was found to interfere with NY99 attachment in C6/36 cells, potentially contributing to the suppression of NY99 titres. However, EILV had no effect on the attachment of WN02-1956 or internalization of either WNV strain under superinfection conditions. In C. tarsalis, EILV did not affect the infection rate of either WNV strain at either timepoint. However, in mosquitoes, EILV enhanced NY99 infection titres at 3 days post-superinfection, but this effect disappeared at 7 days post-superinfection. In contrast, WN02-1956 infection titres were suppressed by EILV at 7 days post-superinfection. The dissemination and transmission of both WNV strains were not affected by superinfection with EILV at either timepoint. Overall, EILV caused SIE against both WNV strains in C6/36 cells; however, in C. tarsalis, SIE caused by EILV was strain specific potentially owing to differences in the rate of depletion of shared resources by the individual WNV strains.


Assuntos
Culex , Mosquitos Vetores , Superinfecção , Vírus do Nilo Ocidental , Animais , Culex/virologia , Vírus do Nilo Ocidental/fisiologia , Mosquitos Vetores/virologia , Superinfecção/virologia , Linhagem Celular , Febre do Nilo Ocidental/transmissão , Febre do Nilo Ocidental/virologia , Replicação Viral
3.
PLoS Pathog ; 20(8): e1012498, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39178311

RESUMO

Influenza infections result in a significant number of severe illnesses annually, many of which are complicated by secondary bacterial super-infection. Primary influenza infection has been shown to increase susceptibility to secondary methicillin-resistant Staphylococcus aureus (MRSA) infection by altering the host immune response, leading to significant immunopathology. Type III interferons (IFNs), or IFNλs, have gained traction as potential antiviral therapeutics due to their restriction of viral replication without damaging inflammation. The role of IFNλ in regulating epithelial biology in super-infection has recently been established; however, the impact of IFNλ on immune cells is less defined. In this study, we infected wild-type and IFNLR1-/- mice with influenza A/PR/8/34 followed by S. aureus USA300. We demonstrated that global IFNLR1-/- mice have enhanced bacterial clearance through increased uptake by phagocytes, which was shown to be cell-intrinsic specifically in myeloid cells in mixed bone marrow chimeras. We also showed that depletion of IFNLR1 on CX3CR1 expressing myeloid immune cells, but not neutrophils, was sufficient to significantly reduce bacterial burden compared to mice with intact IFNLR1. These findings provide insight into how IFNλ in an influenza-infected lung impedes bacterial clearance during super-infection and show a direct cell intrinsic role for IFNλ signaling on myeloid cells.


Assuntos
Camundongos Knockout , Infecções por Orthomyxoviridae , Fagócitos , Superinfecção , Animais , Camundongos , Fagócitos/imunologia , Infecções por Orthomyxoviridae/imunologia , Superinfecção/imunologia , Superinfecção/microbiologia , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/imunologia , Receptores de Interferon/metabolismo , Receptores de Interferon/genética , Interferon lambda , Interferons/metabolismo , Interferons/imunologia , Vírus da Influenza A/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Pulmão/imunologia , Pulmão/virologia , Pulmão/microbiologia , Interleucinas
4.
JBJS Case Connect ; 14(3)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-39208148

RESUMO

CASE: An 18-year-old immunocompetent man presented with symptomatic lumbar canal stenosis, discharging sinuses, and cold abscess for 3 years treated with multiple incision drainage procedures with cottonoid packing antibiotic therapy. Radiographic imaging showed a soft tissue mass in the canal causing bony destruction. Postoperative histopathological examination showed an Aspergillus fungal ball. Patient showed improvement at 6-month follow-up with medical management. CONCLUSION: As far as we know, this is the first case report showing an aspergilloma involving the vertebral column. Inadequate antibiotic treatment and blind introduction of a foreign body into sinus tracts can lead to fungal infections mimicking tuberculosis, causing disastrous outcomes. Fungal cultures are recommended routinely.


Assuntos
Aspergilose , Humanos , Masculino , Adolescente , Aspergilose/diagnóstico por imagem , Aspergilose/complicações , Superinfecção/microbiologia , Canal Medular/diagnóstico por imagem , Canal Medular/microbiologia , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia
6.
Viruses ; 16(7)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39066246

RESUMO

Neutrophils are critical immune cells in severe coronavirus disease 2019 (COVID-19). S100 calcium-binding protein A12 (S100A12) is highly expressed in neutrophils during acute inflammation. The aim of this study was to evaluate serum S100A12 levels as a diagnostic and prognostic tool in COVID-19. Serum samples of patients with moderate and severe COVID-19 were collected during 2020 to 2024. Enzyme-linked immunosorbent assay was used to measure serum S100A12 levels in 63 patients with moderate COVID-19, 60 patients with severe disease and 33 healthy controls. Serum S100A12 levels were elevated in moderate COVID-19 compared to controls and were even higher in severe cases. In moderate disease, serum S100A12 levels positively correlated with immune cell counts. While C-reactive protein and procalcitonin are established inflammation markers, they did not correlate with serum S100A12 levels in either patient cohort. Patients with severe COVID-19 and vancomycin-resistant enterococcus (VRE) infection had increased S100A12 levels. Elevated S100A12 levels were also observed in patients with herpes simplex reactivation. Fungal superinfections did not alter S100A12 levels. These data show that serum S100A12 increases in moderate and severe COVID-19 and is further elevated by VRE bloodstream infection and herpes simplex reactivation. Therefore, S100A12 may serve as a novel biomarker for severe COVID-19 and an early diagnostic indicator for bacterial and viral infections.


Assuntos
Biomarcadores , COVID-19 , Herpes Simples , Proteína S100A12 , SARS-CoV-2 , Humanos , COVID-19/diagnóstico , COVID-19/sangue , COVID-19/imunologia , Masculino , Feminino , Proteína S100A12/sangue , Pessoa de Meia-Idade , Biomarcadores/sangue , SARS-CoV-2/imunologia , Prognóstico , Idoso , Herpes Simples/diagnóstico , Herpes Simples/sangue , Adulto , Índice de Gravidade de Doença , Superinfecção/diagnóstico , Superinfecção/sangue , Farmacorresistência Bacteriana Múltipla , Neutrófilos/imunologia , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Enterococos Resistentes à Vancomicina
7.
ISME J ; 18(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-39052320

RESUMO

Biofilm is a group of heterogeneously structured and densely packed bacteria with limited access to nutrients and oxygen. These intrinsic features can allow a mono-species biofilm to diversify into polymorphic subpopulations, determining the overall community's adaptive capability to changing ecological niches. However, the specific biological functions underlying biofilm diversification and fitness adaptation are poorly demonstrated. Here, we launched and monitored the experimental evolution of Pseudomonas aeruginosa biofilms, finding that two divergent molecular trajectories were adopted for adaptation to higher competitive fitness in biofilm formation: one involved hijacking bacteriophage superinfection to aggressively inhibit kin competitors, whereas the other induced a subtle change in cyclic dimeric guanosine monophosphate signaling to gain a positional advantage via enhanced early biofilm adhesion. Bioinformatics analyses implicated that similar evolutionary strategies were prevalent among clinical P. aeruginosa strains, indicative of parallelism between natural and experimental evolution. Divergence in the molecular bases illustrated the adaptive values of genomic plasticity for gaining competitive fitness in biofilm formation. Finally, we demonstrated that these fitness-adaptive mutations reduced bacterial virulence. Our findings revealed how the mutations intrinsically generated from the biofilm environment influence the evolution of P. aeruginosa.


Assuntos
Biofilmes , Pseudomonas aeruginosa , Biofilmes/crescimento & desenvolvimento , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiologia , Aptidão Genética , Adaptação Fisiológica , Virulência , Mutação , Bacteriófagos/genética , Bacteriófagos/fisiologia , GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Superinfecção/microbiologia , Evolução Biológica
8.
Front Immunol ; 15: 1364622, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38933273

RESUMO

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.


Assuntos
Anticorpos Monoclonais , Coinfecção , Streptococcus pneumoniae , Superinfecção , Animais , Humanos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/imunologia , Streptococcus pneumoniae/imunologia , Camundongos , Superinfecção/imunologia , Superinfecção/microbiologia , Coinfecção/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Metapneumovirus/imunologia , Vírus da Influenza A/imunologia , Modelos Animais de Doenças , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/prevenção & controle , Infecções Pneumocócicas/tratamento farmacológico , Feminino , Camundongos Endogâmicos BALB C , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/tratamento farmacológico , Anticorpos Antivirais/imunologia
9.
J Burn Care Res ; 45(5): 1356-1360, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-38915197

RESUMO

Mucormycosis is an uncommon infection but is increasing in prevalence. Cutaneous disease is associated with burns and traumatic injuries. Cutaneous mucormycosis is the least deadly form but mortality is still approximately 36%. Burn superinfection with mucormycosis is increasingly common and can be an insidious process that may not present until the disease disseminates. We present the case of a 30-year-old male who presented to the emergency department for a rash. A rash with yellow crusting was noted to involve his scalp, face, ear, right shoulder, and parts of both feet. He had been placed on antibiotics by an urgent care a few days prior to presenting. He denied systemic symptoms, chemical exposure, change in detergent, autoimmune diseases, or travel. The patient has a history of intravenous opioid and dissociative abuse and had multiple episodes of syncope-including at his work in a factory where there were hot metals, refrigerants, and numerous corrosive chemicals. Surgical debridement revealed mucormycosis on pathology. The patient was treated with isavuconazole, surgical debridement, and skin grafting. He experienced complete recovery.


Assuntos
Antifúngicos , Queimaduras Químicas , Mucormicose , Superinfecção , Humanos , Masculino , Mucormicose/diagnóstico , Mucormicose/terapia , Adulto , Superinfecção/microbiologia , Antifúngicos/uso terapêutico , Queimaduras Químicas/terapia , Queimaduras Químicas/microbiologia , Desbridamento , Triazóis/uso terapêutico , Transplante de Pele , Piridinas/uso terapêutico , Piridinas/efeitos adversos , Nitrilas/uso terapêutico
10.
Viruses ; 16(5)2024 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-38793665

RESUMO

Epidemiologic studies have established that mpox (formerly known as monkeypox) outbreaks worldwide in 2022-2023, due to Clade IIb mpox virus (MPXV), disproportionately affected gay, bisexual, and other men who have sex with men. More than 35% and 40% of the mpox cases suffer from co-infection with HIV and sexually transmitted infections (STIs) (e.g., Chlamydia trachomatis, Neisseria gonorrhoeae, Treponema pallidum, and herpes simplex virus), respectively. Bacterial superinfection can also occur. Co-infection of MPXV and other infectious agents may enhance disease severity, deteriorate outcomes, elongate the recovery process, and potentially contribute to the morbidity and mortality of the ensuing diseases. However, the interplays between MPXV and HIV, bacteria, other STI pathogens and host cells are poorly studied. There are many open questions regarding the impact of co-infections with HIV, STIs, or bacterial superinfections on the diagnosis and treatment of MPXV infections, including clinical and laboratory-confirmed mpox diagnosis, suboptimal treatment effectiveness, and induction of antiviral drug resistance. In this review article, we will discuss the progress and knowledge gaps in MPXV biology, antiviral therapy, pathogenesis of human MPXV and its co-infection with HIV, STIs, or bacterial superinfections, and the impact of the co-infections on the diagnosis and treatment of mpox disease. This review not only sheds light on the MPXV infection and co-infection of other etiologies but also calls for more research on MPXV life cycles and the molecular mechanisms of pathogenesis of co-infection of MPXV and other infectious agents, as well as research and development of a novel multiplex molecular testing panel for the detection of MPXV and other STI co-infections.


Assuntos
Coinfecção , Infecções por HIV , Infecções Sexualmente Transmissíveis , Humanos , Masculino , Coinfecção/microbiologia , Coinfecção/virologia , Infecções por HIV/complicações , Infecções por HIV/virologia , Monkeypox virus , Mpox/virologia , Infecções Sexualmente Transmissíveis/microbiologia , Infecções Sexualmente Transmissíveis/virologia , Infecções Sexualmente Transmissíveis/complicações , Superinfecção/microbiologia , Superinfecção/virologia , Feminino
11.
J Math Biol ; 89(1): 7, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38772937

RESUMO

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.


Assuntos
Modelos Epidemiológicos , Malária Vivax , Mosquitos Vetores , Plasmodium vivax , Humanos , Animais , Plasmodium vivax/crescimento & desenvolvimento , Plasmodium vivax/fisiologia , Malária Vivax/imunologia , Malária Vivax/parasitologia , Malária Vivax/transmissão , Mosquitos Vetores/parasitologia , Mosquitos Vetores/fisiologia , Superinfecção/imunologia , Superinfecção/parasitologia , Fígado/parasitologia , Probabilidade
12.
Viruses ; 16(5)2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38793618

RESUMO

Viral infection can regulate the cell cycle, thereby promoting viral replication. Hijacking and altering the cell cycle are important for the virus to establish and maintain a latent infection. Previously, Spodoptera exigua multiple nucleopolyhedrovirus (SeMNPV)-latently infected P8-Se301-C1 cells, which grew more slowly than Se301 cells and interfered with homologous SeMNNPV superinfection, were established. However, the effects of latent and superinfection with baculoviruses on cell cycle progression remain unknown. In this study, the cell cycle profiles of P8-Se301-C1 cells and SeMNPV or Autographa californica multiple nucleopolyhedrovirus (AcMNPV)-infected P8-Se301-C1 cells were characterized by flow cytometry. The results showed that replication-related genes MCM4, PCNA, and BAF were down-regulated (p < 0.05) in P8-Se301-C1 cells, and the S phase of P8-Se301-C1 cells was longer than that of Se301 cells. P8-Se301-C1 cells infected with SeMNPV did not arrest in the G2/M phase or affect the expression of Cyclin B and cyclin-dependent kinase 1 (CDK1). Furthermore, when P8-Se301-C1 cells were infected with SeMNPV after synchronized treatment with hydroxyurea and nocodazole, light microscopy and qRT-PCR analysis showed that, compared with unsynchronized cells and S and G2/M phase cells, SeMNPV-infected P8-Se301-C1 cells in G1 phase induced G2/M phase arrest, and the amount of virus adsorption and intracellular viral DNA replication were significantly increased (p < 0.05). In addition, budded virus (BV) production and occlusion body (OB)-containing cells were both increased at 120 h post-infection (p < 0.05). The expression of Cyclin B and CDK1 was significantly down-regulated at 48 h post-infection (p < 0.05). Finally, the arrest of SeMNPV-infected G1 phase cells in the G2/M phase increased BV production (p < 0.05) and the number of OB-containing cells. In conclusion, G1 phase infection and G2/M arrest are favorable to SeMNPV proliferation in P8-Se301-C1 cells, thereby alleviating the homologous superinfection exclusion. The results contribute to a better understanding of the relationship between baculoviruses and insect cell cycle progression and regulation.


Assuntos
Pontos de Checagem da Fase G2 do Ciclo Celular , Nucleopoliedrovírus , Spodoptera , Superinfecção , Replicação Viral , Animais , Nucleopoliedrovírus/fisiologia , Linhagem Celular , Spodoptera/virologia , Superinfecção/virologia , Fase G1
13.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38693120

RESUMO

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Assuntos
Antígeno CD47 , Células Epiteliais , Infecções Estafilocócicas , Staphylococcus aureus , Superinfecção , Antígeno CD47/metabolismo , Antígeno CD47/genética , Humanos , Animais , Superinfecção/microbiologia , Camundongos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Influenza Humana/metabolismo , Influenza Humana/imunologia , Influenza Humana/virologia , Aderência Bacteriana , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Camundongos Endogâmicos C57BL , Brônquios/metabolismo , Brônquios/citologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Camundongos Knockout , Vírus da Influenza A Subtipo H1N1
14.
Am J Respir Cell Mol Biol ; 71(1): 110-120, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38574279

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda , Modelos Animais de Doenças , Interferon gama , Interleucina-10 , Superinfecção , Animais , Interleucina-10/metabolismo , Interleucina-10/imunologia , Lesão Pulmonar Aguda/virologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/microbiologia , Interferon gama/metabolismo , Superinfecção/imunologia , Superinfecção/virologia , Camundongos , Camundongos Endogâmicos C57BL , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Coinfecção/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/virologia , Infecções Estafilocócicas/imunologia , Camundongos Knockout , Vírus da Influenza A/imunologia , Pulmão/virologia , Pulmão/patologia , Pulmão/imunologia , Pulmão/metabolismo
15.
Virus Res ; 345: 199375, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38642618

RESUMO

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.


Assuntos
COVID-19 , Infecções por Citomegalovirus , Citomegalovirus , Receptores de Lipopolissacarídeos , Monócitos , SARS-CoV-2 , Superinfecção , Humanos , Monócitos/virologia , Monócitos/imunologia , Citomegalovirus/imunologia , Receptores de Lipopolissacarídeos/metabolismo , SARS-CoV-2/imunologia , COVID-19/virologia , COVID-19/imunologia , Infecções por Citomegalovirus/virologia , Infecções por Citomegalovirus/imunologia , Superinfecção/virologia , Superinfecção/imunologia , Latência Viral , Inflamação , Coinfecção/virologia , Citocinas/metabolismo , Replicação Viral
16.
Am J Clin Dermatol ; 25(4): 623-637, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38578398

RESUMO

Secondary bacterial infections of common dermatoses such as atopic dermatitis, ectoparasitosis, and varicella zoster virus infections are frequent, with Staphylococcus aureus and Streptococcus pyogenes being the bacteria most involved. There are also Gram-negative infections secondary to common dermatoses such as foot dyshidrotic eczema and tinea pedis. Factors favoring secondary bacterial infections in atopic dermatitis, ectoparasitosis, and varicella zoster virus infections mainly include an epidermal barrier alteration as well as itch. Mite-bacteria interaction is also involved in scabies and some environmental factors can promote Gram-negative bacterial infections of the feet. Furthermore, the bacterial ecology of these superinfections may depend on the geographical origin of the patients, especially in ectoparasitosis. Bacterial superinfections can also have different clinical aspects depending on the underlying dermatoses. Subsequently, the choice of class, course, and duration of antibiotic treatment depends on the severity of the infection and the suspected bacteria, primarily targeting S. aureus. Prevention of these secondary bacterial infections depends first and foremost on the management of the underlying skin disorder. At the same time, educating the patient on maintaining good skin hygiene and reporting changes in the primary lesions is crucial. In the case of recurrent secondary infections, decolonization of S. aureus is deemed necessary, particularly in atopic dermatitis.


Assuntos
Antibacterianos , Dermatite Atópica , Humanos , Dermatite Atópica/complicações , Dermatite Atópica/microbiologia , Dermatite Atópica/diagnóstico , Antibacterianos/uso terapêutico , Superinfecção/microbiologia , Superinfecção/diagnóstico , Dermatopatias Bacterianas/microbiologia , Dermatopatias Bacterianas/diagnóstico , Dermatopatias Bacterianas/epidemiologia , Staphylococcus aureus/isolamento & purificação , Coinfecção/microbiologia
17.
Front Immunol ; 15: 1277447, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38633245

RESUMO

Modified vaccinia virus Ankara (MVA) has been widely tested in clinical trials as recombinant vector vaccine against infectious diseases and cancers in humans and animals. However, one biosafety concern about the use of MVA vectored vaccine is the potential for MVA to recombine with naturally occurring orthopoxviruses in cells and hosts in which it multiplies poorly and, therefore, producing viruses with mosaic genomes with altered genetic and phenotypic properties. We previously conducted co-infection and superinfection experiments with MVA vectored influenza vaccine (MVA-HANP) and a feline Cowpox virus (CPXV-No-F1) in Vero cells (that were semi-permissive to MVA infection) and showed that recombination occurred in both co-infected and superinfected cells. In this study, we selected the putative recombinant viruses and performed genomic characterization of these viruses. Some putative recombinant viruses displayed plaque morphology distinct of that of the parental viruses. Our analysis demonstrated that they had mosaic genomes of different lengths. The recombinant viruses, with a genome more similar to MVA-HANP (>50%), rescued deleted and/or fragmented genes in MVA and gained new host ranges genes. Our analysis also revealed that some MVA-HANP contained a partially deleted transgene expression cassette and one recombinant virus contained part of the transgene expression cassette similar to that incomplete MVA-HANP. The recombination in co-infected and superinfected Vero cells resulted in recombinant viruses with unpredictable biological and genetic properties as well as recovery of delete/fragmented genes in MVA and transfer of the transgene into replication competent CPXV. These results are relevant to hazard characterization and risk assessment of MVA vectored biologicals.


Assuntos
Coinfecção , Vacinas contra Influenza , Superinfecção , Chlorocebus aethiops , Animais , Gatos , Humanos , Vacinas contra Influenza/genética , Vírus da Varíola Bovina/genética , Células Vero , Vaccinia virus , Vacinas Sintéticas/genética , Sequenciamento Completo do Genoma
19.
J Gen Virol ; 105(3)2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38471041

RESUMO

Many viruses downregulate their cognate receptors, facilitating virus replication and pathogenesis via processes that are not yet fully understood. In the case of herpes simplex virus 1 (HSV1), the receptor binding protein glycoprotein D (gD) has been implicated in downregulation of its receptor nectin1, but current understanding of the process is limited. Some studies suggest that gD on the incoming virion is sufficient to achieve nectin1 downregulation, but the virus-encoded E3 ubiquitin ligase ICP0 has also been implicated. Here we have used the physiologically relevant nTERT human keratinocyte cell type - which we have previously shown to express readily detectable levels of endogenous nectin1 - to conduct a detailed investigation of nectin1 expression during HSV1 infection. In these cells, nectin1, but not nectin2 or the transferrin receptor, disappeared from the cell surface in a process that required virus protein synthesis rather than incoming virus, but did not involve virus-induced host shutoff. Furthermore, gD was not only required but was sufficient for nectin1 depletion, indicating that no other virus proteins are essential. NK cells were shown to be activated in the presence of keratinocytes, a process that was greatly inhibited in cells infected with wild-type virus. However, degranulation of NK cells was also inhibited in ΔgD-infected cells, indicating that blocking of NK cell activation was independent of gD downregulation of nectin1. By contrast, a superinfection time-course revealed that the ability of HSV1 infection to block subsequent infection of a GFP-expressing HSV1 was dependent on gD and occurred in line with the timing of nectin1 downregulation. Thus, the role of gD-dependent nectin1 impairment during HSV infection is important for virus infection, but not immune evasion, which is achieved by other mechanisms.


Assuntos
Herpes Simples , Herpesvirus Humano 1 , Superinfecção , Humanos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Regulação para Baixo , Herpesvirus Humano 1/fisiologia , Queratinócitos , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética
20.
Malar J ; 23(1): 68, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38443939

RESUMO

BACKGROUND: Genetic surveillance of the Plasmodium falciparum parasite shows great promise for helping National Malaria Control Programmes (NMCPs) assess parasite transmission. Genetic metrics such as the frequency of polygenomic (multiple strain) infections, genetic clones, and the complexity of infection (COI, number of strains per infection) are correlated with transmission intensity. However, despite these correlations, it is unclear whether genetic metrics alone are sufficient to estimate clinical incidence. METHODS: This study examined parasites from 3147 clinical infections sampled between the years 2012-2020 through passive case detection (PCD) across 16 clinic sites spread throughout Senegal. Samples were genotyped with a 24 single nucleotide polymorphism (SNP) molecular barcode that detects parasite strains, distinguishes polygenomic (multiple strain) from monogenomic (single strain) infections, and identifies clonal infections. To determine whether genetic signals can predict incidence, a series of Poisson generalized linear mixed-effects models were constructed to predict the incidence level at each clinical site from a set of genetic metrics designed to measure parasite clonality, superinfection, and co-transmission rates. RESULTS: Model-predicted incidence was compared with the reported standard incidence data determined by the NMCP for each clinic and found that parasite genetic metrics generally correlated with reported incidence, with departures from expected values at very low annual incidence (< 10/1000/annual [‰]). CONCLUSIONS: When transmission is greater than 10 cases per 1000 annual parasite incidence (annual incidence > 10‰), parasite genetics can be used to accurately infer incidence and is consistent with superinfection-based hypotheses of malaria transmission. When transmission was < 10‰, many of the correlations between parasite genetics and incidence were reversed, which may reflect the disproportionate impact of importation and focal transmission on parasite genetics when local transmission levels are low.


Assuntos
Malária , Superinfecção , Humanos , Senegal/epidemiologia , Incidência , Plasmodium falciparum/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...