Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
STAR Protoc ; 5(3): 103142, 2024 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-39305487

RESUMEN

Here, we present a protocol for primary, human immune cell isolation and stimulation for lipid mediator profiling. We describe steps for the isolation of monocytes from human leukocyte concentrates via density centrifugation and differentiation/polarization toward M1- or M2-monocyte-derived macrophages (MDMs). We detail stimulation approaches of MDMs with live bacteria or influenza A virus for lipid mediator profiling and sample preparation for subsequent analysis, such as enzyme expression, mRNA analysis, or surface marker determination. For complete details on the use and execution of this protocol, please refer to Jordan et al.1.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos , Monocitos , Humanos , Macrófagos/metabolismo , Macrófagos/inmunología , Interacciones Huésped-Patógeno/inmunología , Monocitos/metabolismo , Lípidos/química , Fenotipo , Diferenciación Celular
2.
Front Microbiol ; 15: 1422651, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39206370

RESUMEN

Introduction: Treatment of influenza A virus infections is currently limited to few direct acting antiviral substances. Repurposing other established pharmaceuticals as antivirals could aid in improving treatment options. Methods: This study investigates the antiviral properties of ProcCluster® and procaine hydrochloride, two derivatives of the local anesthetic procaine, in influenza A virus infection of A549, Calu-3 and MDCK cells. Results: Both substances inhibit replication in all three of these cell lines in multi-cycle experiments. However, cell line-dependent differences in the effects of the substances on viral RNA replication and subsequent protein synthesis, as well as release of progeny viruses in single-cycle experiments can be observed. Both ProcCluster® and procaine hydrochloride delay endosome fusion of the virus early in the replication cycle, possibly due to the alkaline nature of the active component procaine. In A549 and Calu-3 cells an additional effect of the substances can be observed at late stages in the first replication cycle. Interestingly, this effect is absent in MDCK cells. We demonstrate that ProcCluster® and procaine hydrochloride inhibit phospholipase A2 (PLA2) enzymes from A549 but not MDCK cells and confirm that specific inhibition of calcium independent PLA2 but not cytosolic PLA2 has antiviral effects. Discussion: We show that ProcCluster® and procaine hydrochloride inhibit influenza A virus infection at several stages of the replication cycle and have potential as antiviral substances.

3.
iScience ; 27(1): 108775, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38261967

RESUMEN

Respiratory influenza A virus (IAV) infections are major health concerns worldwide, where bacterial superinfections substantially increase morbidity and mortality. The underlying mechanisms of how IAV impairs host defense remain elusive. Macrophages are pivotal for the innate immune response and crucially regulate the entire inflammatory process, occurring as inflammatory M1- or pro-resolving M2-like phenotypes. Lipid mediators (LM), produced from polyunsaturated fatty acids by macrophages, are potent immune regulators and impact all stages of inflammation. Using LM metabololipidomics, we show that human pro-resolving M2-macrophages respond to IAV infections with specific and robust production of prostaglandin (PG)E2 along with upregulation of cyclooxygenase-2 (COX-2), which persists after co-infection with Staphylococcus aureus. In contrast, cytokine/interferon production in macrophages was essentially unaffected by IAV infection, and the functionality of M1-macrophages was not influenced. Conclusively, IAV infection of M2-macrophages selectively elevates PGE2 formation, suggesting inhibition of the COX-2/PGE2 axis as strategy to limit IAV exacerbation.

4.
Microbes Infect ; 26(3): 105264, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38008399

RESUMEN

Seasonal influenza A virus (IAV) infections still pose a major burden for public health worldwide. Severe disease progression or even death is often related to superinfections of the virus and a secondary bacterial pathogen. However, fungi, especially Aspergillus fumigatus, are also frequently diagnosed during IAV infection. Although, clinical studies have reported the severity of influenza-associated pulmonary aspergillosis, the molecular mechanisms underlying this type of disease are poorly understood. Here, a new in vitro model is introduced that allows the investigation of complex pathogen-host and pathogen-pathogen interactions during coinfection of lung epithelial cells with IAV and A. fumigatus. Our data reveal a reduced IAV load and IAV-induced cytokine and chemokine expression in the presence of A. fumigatus. At the same time, IAV infection promotes the growth of A. fumigatus. Even in the absence of the human host cell, purified IAV particles are able to induce hyphal growth, due to a direct interaction of the virus particles with the fungal surface. Thus, our study gives first insights into the complex interplay between IAV, A. fumigatus and the host cell as well as the two pathogens alone.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Humanos , Aspergillus fumigatus , Pulmón/microbiología , Células Epiteliales
5.
Int J Mol Sci ; 24(19)2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37834031

RESUMEN

As vaccination efforts against SARS-CoV-2 progress in many countries, there is still an urgent need for efficient antiviral treatment strategies for those with severer disease courses, and lately, considerable efforts have been undertaken to repurpose existing drugs as antivirals. The local anaesthetic procaine has been investigated for antiviral properties against several viruses over the past decades. Here, we present data on the inhibitory effect of the procaine prodrugs ProcCluster® and procaine hydrochloride on SARS-CoV-2 infection in vitro. Both procaine prodrugs limit SARS-CoV-2 progeny virus titres as well as reduce interferon and cytokine responses in a proportional manner to the virus load. The addition of procaine during the early stages of the SARS-CoV-2 replication cycle in a cell culture first limits the production of subgenomic RNA transcripts, and later affects the replication of the viral genomic RNA. Interestingly, procaine additionally exerts a prominent effect on SARS-CoV-2 progeny virus release when added late during the replication cycle, when viral RNA production and protein production are already largely completed.


Asunto(s)
COVID-19 , Profármacos , Animales , Chlorocebus aethiops , SARS-CoV-2 , Antivirales/farmacología , Anestésicos Locales/farmacología , Profármacos/farmacología , Células Vero , Procaína/farmacología , Replicación Viral
6.
Int J Obes (Lond) ; 47(11): 1088-1099, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37587162

RESUMEN

OBJECTIVE: Obesity is an independent risk factor for severe influenza virus and COVID-19 infections. There might be an interplay between adipose tissue and respiratory pathogens, although the mechanism is unknown. Proinflammatory factors secreted by the adipose tissue are often discussed to serve as indirect contributor to virus infection. However, the direct potential of adipose tissue to serve as a viral niche has not yet been investigated. METHODS: Two murine obesity models (DIO and ob/ob) were infected with influenza A virus (IAV) and monitored for 3 weeks. p.i. Lung and adipose tissue were harvested, and the viral load was analysed. Direct replication of IAV in vitro was investigated in human derived primary adipocytes and macrophages. The indirect impact of the secretory products of adipocytes during infection was analysed in a co-culture system with lung fibroblasts. Moreover, lung and adipose tissue was harvested from deceased patients infected with SARS-CoV-2 omicron variant. Additionally, replication of SARS-CoV-2 alpha, delta, and omicron variants was investigated in vitro in adipocytes and macrophages. RESULTS: Both murine obesity models presented high IAV titers compared to non-obese mice. Interestingly, adipose tissue adjacent to the lungs was a focal point for influenza virus replication in mice. We further detected IAV replication and antiviral response in human adipocytes. Co-cultivation of adipocytes and lung fibroblasts led to increased IL-8 concentration during infection. Though we observed SARS-CoV-2 in the thoracic adipose tissue of COVID-19 patients, no active replication was found in adipocytes in vitro. However, SARS-CoV-2 was detected in the macrophages and this finding was associated with increased inflammation. CONCLUSIONS: Our study revealed that thoracic adipose tissue contributes to respiratory virus infection. Besides indirect induction of proinflammatory factors during infection, adipocytes and macrophages within the tissue can directly support viral replication.


Asunto(s)
COVID-19 , Virus de la Influenza A , Gripe Humana , Humanos , Ratones , Animales , Pulmón , Tejido Adiposo , Virus de la Influenza A/fisiología , Obesidad
7.
Int J Mol Sci ; 23(13)2022 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-35805887

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing the coronavirus disease-19 (COVID-19) is still challenging healthcare systems and societies worldwide. While vaccines are available, therapeutic strategies are developing and need to be adapted to each patient. Many clinical approaches focus on the repurposing of approved therapeutics against other diseases. However, the efficacy of these compounds on viral infection or even harmful secondary effects in the context of SARS-CoV-2 infection are sparsely investigated. Similarly, adverse effects of commonly used therapeutics against lifestyle diseases have not been studied in detail. Using mono cell culture systems and a more complex chip model, we investigated the effects of the acetylsalicylic acid (ASA) salt D,L-lysine-acetylsalicylate + glycine (LASAG) on SARS-CoV-2 infection in vitro. ASA is commonly known as Aspirin® and is one of the most frequently used medications worldwide. Our data indicate an inhibitory effect of LASAG on SARS-CoV-2 replication and SARS-CoV-2-induced expression of pro-inflammatory cytokines and coagulation factors. Remarkably, our data point to an additive effect of the combination of LASAG and the antiviral acting drug remdesivir on SARS-CoV-2 replication in vitro.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Antivirales/uso terapéutico , Aspirina/farmacología , Aspirina/uso terapéutico , Glicina/farmacología , Glicina/uso terapéutico , Humanos , Lisina
8.
Viruses ; 14(4)2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35458484

RESUMEN

Omsk haemorrhagic fever virus (OHFV) is the agent leading to Omsk haemorrhagic fever (OHF), a viral disease currently only known in Western Siberia in Russia. The symptoms include fever, headache, nausea, muscle pain, cough and haemorrhages. The transmission cycle of OHFV is complex. Tick bites or contact with infected small mammals are the main source of infection. The Republic of Kazakhstan is adjacent to the endemic areas of OHFV in Russia and febrile diseases with haemorrhages occur throughout the country-often with unclear aetiology. In this study, we examined human cerebrospinal fluid samples of patients with suspected meningitis or meningoencephalitis with unknown origins for the presence of OHFV RNA. Further, reservoir hosts such as rodents and ticks from four Kazakhstan regions were screened for OHFV RNA to clarify if this virus could be the causative agent for many undiagnosed cases of febrile diseases in humans in Kazakhstan. Out of 130 cerebrospinal fluid samples, two patients (1.53%) originating from Almaty city were positive for OHFV RNA. Screening of tick samples revealed positive pools from different areas in the Akmola region. Of the caught rodents, 1.1% out of 621 were positive for OHFV at four trapping areas from the West Kazakhstan region. In this paper, we present a broad investigation of the spread of OHFV in Kazakhstan in human cerebrospinal fluid samples, rodents and ticks. Our study shows for the first time that OHFV can not only be found in the area of Western Siberia in Russia, but can also be detected up to 1.600 km away in the Almaty region in patients and natural foci.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas , Garrapatas , Animales , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Humanos , Mamíferos , ARN , Federación de Rusia/epidemiología , Siberia/epidemiología
9.
Nutrients ; 14(7)2022 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-35406128

RESUMEN

While nutrition during pregnancy is critical for the health of both mother and child, little is known about the diet quality of women during pregnancy, its correlation with gestational weight gain (GWG)/body composition, and chosen maternal adipokines. Therefore, we evaluated the Healthy Eating Index (HEI) of 110 pregnant women and analyzed its correlation with GWG/body composition, physical activity, leptin, resistin, adiponectin, and interleukin 6 (IL-6), respectively. Diet quality was medium in 63% of women, characterized by a high intake of animal-based products. HEI was negatively influenced by pre-pregnancy obesity (ß = −0.335, p = 0.004), and positively influenced by higher age (>35 yrs., ß = 0.365, p ≤ 0.001), upper arm circumference (ß = 0.222, p = 0.052), and total activity during the third trimester (ß = 0.258, p = 0.008). GWG was associated with pre-pregnancy obesity (ß = −0.512, p ≤ 0.001), thigh circumference (ß = 0.342, p = 0.007), upper arm fat area (ß = 0.208, p = 0.092), and maternal age group (>35 yrs. ß = −0.166, p = 0.082), but not with HEI. Leptin and IL-6 displayed associations with variables representative of body composition, such as pre-pregnancy BMI, thigh circumference, upper arm fat area, and upper arm circumference, but were not influenced by HEI. Neither were adiponectin and resistin. IL-6 was also associated with total activity. In conclusion, GWG, leptin, and IL-6 were influenced by nutritional status (body composition/pre-pregnancy BMI), not by maternal diet. Physical activity level also had an impact on IL-6. Thus, efforts should be intensified to improve diet quality and participation in sports before and during pregnancy, particularly in overweight or obese women.


Asunto(s)
Adipoquinas , Dieta , Ganancia de Peso Gestacional , Adiponectina , Índice de Masa Corporal , Estudios Transversales , Femenino , Alemania , Humanos , Interleucina-6 , Leptina , Obesidad , Embarazo , Resistina
10.
Artículo en Inglés | MEDLINE | ID: mdl-34948770

RESUMEN

Physical activity (PA) during pregnancy is beneficial for mother and child. Little is known regarding the effects of PA on specific adipokines/myokines and their impact during pregnancy. This study investigates the correlation between PA during late pregnancy, body composition, and maternal levels of leptin, IL-6, and TNF-α at delivery. In a cross-sectional study of 91 pregnant participants (mean age 33.9 ± 4.6 years) without gestational diabetes mellitus or preeclampsia, anthropometric data and blood samples were taken at delivery. PA during the third trimester was measured via the Pregnancy Physical Activity Questionnaire. Activities were ranked by intensity: sedentary (<1.5 metabolic equivalent (METs)), light (1.5-3.0 METs), moderate (3.0-6.0 METs), and vigorous activity (>6.0 METs). Leptin at delivery correlated positively with body composition and negatively with light PA intensity. Sedentary behaviour showed a positive correlation with IL-6 levels at delivery. Moderate activity during the last trimester, sedentary activity levels, and body composition had the greatest influence on maternal IL-6 at delivery. Completed weeks of pregnancy, moderate and light PA, and sedentary activity had the greatest influence on maternal TNF-α at delivery. PA during late pregnancy potentially affects circulating (adipo-)/myokines. Further studies are needed to examine causal relationships and the impact on maternal and new-born health.


Asunto(s)
Ejercicio Físico , Conducta Sedentaria , Adulto , Composición Corporal , Niño , Estudios Transversales , Femenino , Humanos , Equivalente Metabólico , Embarazo
11.
Int J Mol Sci ; 22(11)2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34067487

RESUMEN

Influenza virus is a well-known respiratory pathogen, which still leads to many severe pulmonary infections in the human population every year. Morbidity and mortality rates are further increased if virus infection coincides with co-infections or superinfections caused by bacteria such as Streptococcus pneumoniae (S. pneumoniae) and Staphylococcus aureus (S. aureus). This enhanced pathogenicity is due to complex interactions between the different pathogens and the host and its immune system and is mainly governed by altered intracellular signaling processes. In this review, we summarize the recent findings regarding the innate and adaptive immune responses during co-infection with influenza virus and S. pneumoniae or S. aureus, describing the signaling pathways involved and how these interactions influence disease outcomes.


Asunto(s)
Coinfección/inmunología , Inmunidad/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Neumocócicas/inmunología , Transducción de Señal/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Humanos , Orthomyxoviridae/inmunología , Staphylococcus aureus/inmunología , Streptococcus pneumoniae/inmunología
12.
Biomolecules ; 11(6)2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072389

RESUMEN

Influenza virus (IV) infections are considered to cause severe diseases of the respiratory tract. Beyond mild symptoms, the infection can lead to respiratory distress syndrome and multiple organ failure. Occurrence of resistant seasonal and pandemic strains against the currently licensed antiviral medications points to the urgent need for new and amply available anti-influenza drugs. Interestingly, the virus-supportive function of the cellular phosphatidylinositol 3-kinase (PI3K) suggests that this signaling module may be a potential target for antiviral intervention. In the sense of repurposing existing drugs for new indications, we used Pictilisib, a known PI3K inhibitor to investigate its effect on IV infection, in mono-cell-culture studies as well as in a human chip model. Our results indicate that Pictilisib is a potent inhibitor of IV propagation already at early stages of infection. In a murine model of IV pneumonia, the in vitro key findings were verified, showing reduced viral titers as well as inflammatory response in the lung after delivery of Pictilisib. Our data identified Pictilisib as a promising drug candidate for anti-IV therapies that warrant further studying. These results further led to the conclusion that the repurposing of previously approved substances represents a cost-effective and efficient way for development of novel antiviral strategies.


Asunto(s)
Indazoles/farmacología , Virus de la Influenza A/metabolismo , Pulmón , Infecciones por Orthomyxoviridae , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Neumonía Viral , Sulfonamidas/farmacología , Células A549 , Animales , Perros , Humanos , Pulmón/enzimología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/virología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/enzimología , Neumonía Viral/virología
13.
Elife ; 102021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33781385

RESUMEN

Clinical observations indicate that COVID-19 is a systemic disease. An investigation of the viral distribution within the human body and its correlation with tissue damage can aid in understanding the pathophysiology of SARS-CoV-2 infection. We present a detailed mapping of the viral RNA in 61 tissues and organs of 11 deceased patients with COVID-19. The autopsies were performed within the early postmortem interval (between 1.5 and 15 hr, mean: 5.6 hr) to minimize the bias due to viral RNA and tissue degradation. Very high viral loads (>104copies/ml) were detected in most patients' lungs, and the presence of intact viral particles in the lung tissue could be verified by transmission electron microscopy. Interestingly, viral RNA was detected throughout various extrapulmonary tissues and organs without visible tissue damage. The dissemination of SARS-CoV-2-RNA throughout the body supports the hypothesis that there is a maladaptive host response with viremia and multiorgan dysfunction.


Since the discovery of the new coronavirus that causes COVID-19, scientists have been scrambling to understand the different features of the virus. While a lot more is now known about SARS-CoV-2, several key questions have proved more difficult to answer. For example, it remained unclear where the virus travels to in the body and causes the most harm. To help answer this question, Deinhardt-Emmer, Wittschieber et al. performed postmortem examinations on 11 patients who had recently died of COVID-19. After sampling 61 different organs and tissues from each patient, several tests were used to detect traces of SARS-CoV-2. The experiments showed that the largest pool of SARS-CoV-2 was present in the lungs, where it had caused severe damage to the alveolae, the delicate air sacs at the end of the lungs' main air tubes. Small amounts of the virus were also detected in other organs and tissues, but no severe tissue damage was seen. In addition, Deinhardt-Emmer, Wittschieber et al. found that each patient had increased levels of some of the proteins involved in inflammation and blood clotting circulating their bloodstream. This suggests that the inflammation caused by SARS-CoV-2 leads to an excessive immune reaction throughout the entire body. This research provides important new insights into which areas of the body are most impacted by SARS-CoV-2. These findings may help to design more effective drug treatments that target the places SARS-CoV-2 is most likely to accumulate and help patients fight off the infection at these regions.


Asunto(s)
Autopsia , COVID-19/patología , SARS-CoV-2/aislamiento & purificación , Anciano , Anciano de 80 o más Años , COVID-19/sangre , COVID-19/inmunología , COVID-19/terapia , Causas de Muerte , Comorbilidad , Femenino , Humanos , Inflamación/sangre , Inflamación/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , ARN Viral/sangre , Carga Viral
14.
Cell Microbiol ; 23(6): e13323, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33655690

RESUMEN

Nonsense-mediated mRNA decay (NMD) was identified as a process to degrade flawed cellular messenger RNA (mRNA). Within the last decades it was also shown that NMD carries virus-restricting capacities and thus could be considered a part of the cellular antiviral system. As this was shown to affect primarily positive-sense single stranded RNA ((+)ssRNA) viruses there is only scarce knowledge if this also applies to negative-sense single stranded RNA ((-)ssRNA) viruses. Influenza A viruses (IAVs) harbour a segmented (-)ssRNA genome. During their replication IAVs produce numerous RNA transcripts and simultaneously impair cellular transcription and translation. The viral mRNAs hold several molecular patterns which can elicit NMD and in turn would lead to their degradation. This, in consequence, may mitigate viral propagation. Thus, we examined if a knockdown or a pharmacological inhibition of NMD key components may influence IAV replication. Additionally, we performed similar experiments with respiratory syncytial virus (RSV), another (-)ssRNA virus, but with a non-segmented genome. Although it seemed that a knockdown of up-frameshift protein 1 (UPF1), the central NMD factor, slightly increased viral mRNA and protein levels, no significant alteration of viral replication could be observed, implying that the NMD machinery may not have restricting capacities against (-)ssRNA viruses.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Células A549 , Silenciador del Gen , Humanos , ARN Helicasas/genética , ARN Viral/genética , Virus Sincitiales Respiratorios/genética , Transactivadores/genética , Replicación Viral
15.
J Virol ; 95(10)2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637603

RESUMEN

Infections with SARS-CoV-2 can be asymptomatic, but they can also be accompanied by a variety of symptoms that result in mild to severe coronavirus disease-19 (COVID-19) and are sometimes associated with systemic symptoms. Although the viral infection originates in the respiratory system, it is unclear how the virus can overcome the alveolar barrier, which is observed in severe COVID-19 disease courses. To elucidate the viral effects on the barrier integrity and immune reactions, we used mono-cell culture systems and a complex human chip model composed of epithelial, endothelial, and mononuclear cells. Our data show that SARS-CoV-2 efficiently infected epithelial cells with high viral loads and inflammatory response, including interferon expression. By contrast, the adjacent endothelial layer was neither infected nor did it show productive virus replication or interferon release. With prolonged infection, both cell types were damaged, and the barrier function was deteriorated, allowing the viral particles to overbear. In our study, we demonstrate that although SARS-CoV-2 is dependent on the epithelium for efficient replication, the neighboring endothelial cells are affected, e.g., by the epithelial cytokines or components induced during infection, which further results in the damage of the epithelial/endothelial barrier function and viral dissemination.IMPORTANCESARS-CoV-2 challenges healthcare systems and societies worldwide in unprecedented ways. Although numerous new studies have been conducted, research to better understand the molecular pathogen-host interactions are urgently needed. For this, experimental models have to be developed and adapted. In the present study we used mono cell-culture systems and we established a complex chip model, where epithelial and endothelial cells are cultured in close proximity. We demonstrate that epithelial cells can be infected with SARS-CoV-2, while the endothelium did not show any infection signs. Since SARS-CoV-2 is able to establish viremia, the link to thromboembolic events in severe COVID-19 courses is evident. However, whether the endothelial layer is damaged by the viral pathogens or whether other endothelial-independent homeostatic factors are induced by the virus is essential for understanding the disease development. Therefore, our study is important as it demonstrates that the endothelial layer could not be infected by SARS-CoV-2 in our in vitro experiments, but we were able to show the destruction of the epithelial-endothelial barrier in our chip model. From our experiments we can assume that virus-induced host factors disturbed the epithelial-endothelial barrier function and thereby promote viral spread.

16.
Planta Med ; 87(10-11): 818-826, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32781473

RESUMEN

In a cytopathic effect inhibition assay, a standardized Rhodiola rosea root and rhizome extract, also known as roseroot extract (SHR-5), exerted distinct anti-influenza A virus activity against HK/68 (H3N2) (IC50 of 2.8 µg/mL) without being cytotoxic. For fast and efficient isolation and identification of the extract's bioactive constituents, a high-performance countercurrent chromatographic separation method was developed. It resulted in a three-stage gradient elution program using a mobile phase solvent system composed of ethyl acetate/n-butanol/water (1 : 4 : 5 → 2 : 3 : 5 → 3 : 2 : 5) in the reversed-phase mode. The elaborated high-performance countercurrent chromatographic method allowed for fractionation of the complex roseroot extract in a single chromatographic step in a way that only one additional orthogonal isolation/purification step per fraction yielded 12 isolated constituents. They cover a broad polarity range and belong to different structural classes, namely, the phenylethanoid tyrosol and its glucoside salidroside, the cinnamyl alcohol glycosides rosavin, rosarin, and rosin as well as gallic acid, the cyanogenic glucoside lotaustralin, the monoterpene glucosides rosiridin and kenposide A, and the flavonoids tricin, tricin-5-O-ß-D-glucopyranoside, and rhodiosin. The most promising anti-influenza activities were determined for rhodiosin, tricin, and tricin-5-O-ß-D-glucopyranoside with IC50 values of 7.9, 13, and 15 µM, respectively. The herein established high-performance countercurrent chromatographic protocol enables fast and scalable access to major as well as minor roseroot constituents. This is of particular relevance for extract standardization, quality control, and further in-depth pharmacological investigations of the metabolites of this popular traditional herbal remedy.


Asunto(s)
Rhodiola , Distribución en Contracorriente , Glicósidos , Subtipo H3N2 del Virus de la Influenza A , Raíces de Plantas
17.
Microorganisms ; 8(12)2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33333815

RESUMEN

Human beings are exposed to microorganisms every day. Among those, diverse commensals and potential pathogens including Staphylococcus aureus (S. aureus) compose a significant part of the respiratory tract microbiota. Remarkably, bacterial colonization is supposed to affect the outcome of viral respiratory tract infections, including those caused by influenza viruses (IV). Since 30% of the world's population is already colonized with S. aureus that can develop metabolically inactive dormant phenotypes and seasonal IV circulate every year, super-infections are likely to occur. Although IV and S. aureus super-infections are widely described in the literature, the interactions of these pathogens with each other and the host cell are only scarcely understood. Especially, the effect of quasi-dormant bacterial subpopulations on IV infections is barely investigated. In the present study, we aimed to investigate the impact of S. aureus small colony variants on the cell intrinsic immune response during a subsequent IV infection in vitro. In fact, we observed a significant impact on the regulation of pro-inflammatory factors, contributing to a synergistic effect on cell intrinsic innate immune response and induction of harmful cell death. Interestingly, the cytopathic effect, which was observed in presence of both pathogens, was not due to an increased pathogen load.

18.
PLoS One ; 15(5): e0233052, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32413095

RESUMEN

Severe influenza virus (IV) infections still represent a major challenge to public health. To combat IV infections, vaccines and antiviral compounds are available. However, vaccine efficacies vary with very limited to no protection against newly emerging zoonotic IV introductions. In addition, the development of resistant virus variants against currently available antivirals can be rapidly detected, in consequence demanding the design of novel antiviral strategies. Virus supportive cellular signaling cascades, such as the NF-κB pathway, have been identified to be promising antiviral targets against IV in in vitro and in vivo studies and clinical trials. While administration of NF-κB pathway inhibiting agents, such as LASAG results in decreased IV replication, it remained unclear whether blocking of NF-κB might sensitize cells to secondary bacterial infections, which often come along with viral infections. Thus, we examined IV and Staphylococcus aureus growth during LASAG treatment. Interestingly, our data reveal that the presence of LASAG during superinfection still leads to reduced IV titers. Furthermore, the inhibition of the NF-κB pathway resulted in decreased intracellular Staphylococcus aureus loads within epithelial cells, indicating a dependency on the pathway for bacterial uptake. Unfortunately, so far it is not entirely clear if this phenomenon might be a drawback in bacterial clearance during infection.


Asunto(s)
Antivirales/efectos adversos , Aspirina/análogos & derivados , Infecciones Bacterianas/etiología , Glicina/efectos adversos , Gripe Humana/tratamiento farmacológico , Lisina/análogos & derivados , FN-kappa B/antagonistas & inhibidores , Células A549 , Aspirina/efectos adversos , Combinación de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Técnicas de Silenciamiento del Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/complicaciones , Gripe Humana/virología , Lisina/efectos adversos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/etiología , Sobreinfección/etiología , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Replicación Viral/efectos de los fármacos
19.
PLoS One ; 15(4): e0231138, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32243477

RESUMEN

In respect to the heterogeneity among influenza A virus strains and the shortcomings of current vaccination programs, there is a huge interest in the development of alternative vaccines that provide a broader and more long-lasting protection. Gene-based approaches are considered as promising candidates for such flu vaccines. In our study, innate signalling molecules from the RIG-I and the NALP3 pathways were evaluated as genetic adjuvants in intramuscular DNA immunizations. Plasmids encoding a constitutive active form of RIG-I (cRIG-I), IPS-1, IL-1ß, or IL-18 were co-administered with plasmids encoding the hemagglutinin and nucleoprotein derived from H1N1/Puerto Rico/8/1934 via electroporation in BALB/c mice. Immunogenicity was analysed in detail and efficacy was demonstrated in homologous and heterologous influenza challenge experiments. Although the biological activities of the adjuvants have been confirmed by in vitro reporter assays, their single or combined inclusion in the vaccine did not result in superior vaccine efficacy. With the exception of significantly increased levels of antigen-specific IgG1 after the co-administration of IL-1ß, there were only minor alterations concerning the immunogenicity. Since DNA electroporation alone induced substantial inflammation at the injection site, as demonstrated in this study using Mx2-Luc reporter mice, it might override the adjuvants´ contribution to the inflammatory microenvironment and thereby minimizes the influence on the immunogenicity. Taken together, the DNA immunization was protective against subsequent challenge infections but could not be further improved by the genetic adjuvants analysed in this study.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Inmunidad Innata , Vacunas contra la Influenza/inmunología , Transducción de Señal , Vacunas de ADN/inmunología , Animales , Antígenos/inmunología , Linfocitos T CD8-positivos , Bovinos , Línea Celular , Perros , Femenino , Inmunidad Humoral , Inmunización , Inflamación/patología , Virus de la Influenza B/inmunología , Cinética , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae
20.
Microorganisms ; 8(4)2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316261

RESUMEN

Pneumonia is the leading cause of hospitalization worldwide. Besides viruses, bacterial co-infections dramatically exacerbate infection. In general, surfactant protein-A (SP-A) represents a first line of immune defense. In this study, we analyzed whether influenza A virus (IAV) and/or Staphylococcus aureus (S. aureus) infections affect SP-A expression. To closely reflect the situation in the lung, we used a human alveolus-on-a-chip model and a murine pneumonia model. Our results show that S. aureus can reduce extracellular levels of SP-A, most likely attributed to bacterial proteases. Mono-epithelial cell culture experiments reveal that the expression of SP-A is not directly affected by IAV or S. aureus. Yet, the mRNA expression of SP-A is strongly down-regulated by TNF-α, which is highly produced by professional phagocytes in response to bacterial infection. By using the human alveolus-on-a-chip model, we show that the down-regulation of SP-A is strongly dependent on macrophages. In a murine model of pneumonia, we can confirm that S. aureus decreases SP-A levels in vivo. These findings indicate that (I) complex interactions of epithelial and immune cells induce down-regulation of SP-A expression and (II) bacterial mono- and super-infections reduce SP-A expression in the lung, which might contribute to a severe outcome of bacterial pneumonia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA