Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Front Immunol ; 12: 656419, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1506563

RESUMEN

Tuberculosis (TB) is the global health problem with the second highest number of deaths from a communicable disease after COVID-19. Although TB is curable, poor health infrastructure, long and grueling TB treatments have led to the spread of TB pandemic with alarmingly increasing multidrug-resistant (MDR)-TB prevalence. Alternative host modulating therapies can be employed to improve TB drug efficacies or dampen the exaggerated inflammatory responses to improve lung function. Here, we investigated the adjunct therapy of natural immune-modulatory compound berberine in C57BL/6 mouse model of pulmonary TB. Berberine treatment did not affect Mtb growth in axenic cultures; however, it showed increased bacterial killing in primary murine bone marrow-derived macrophages and human monocyte-derived macrophages. Ad libitum berberine administration was beneficial to the host in combination with rifampicin and isoniazid. Berberine adjunctive treatment resulted in decreased lung pathology with no additive or synergistic effects on bacterial burdens in mice. Lung immune cell flow cytometry analysis showed that adjunctive berberine treatment decreased neutrophil, CD11b+ dendritic cell and recruited interstitial macrophage numbers. Late onset of adjunctive berberine treatment resulted in a similar phenotype with consistently reduced numbers of neutrophils both in lungs and the spleen. Together, our results suggest that berberine can be supplemented as an immunomodulatory agent depending on the disease stage and inflammatory status of the host.


Asunto(s)
Antituberculosos/uso terapéutico , Berberina/uso terapéutico , Factores Inmunológicos/uso terapéutico , Isoniazida/uso terapéutico , Rifampin/uso terapéutico , Tuberculosis Pulmonar/tratamiento farmacológico , Animales , Antituberculosos/farmacología , Berberina/farmacología , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Quimioterapia Combinada , Femenino , Humanos , Factores Inmunológicos/farmacología , Isoniazida/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Rifampin/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/microbiología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
2.
Adv Drug Deliv Rev ; 179: 114020, 2021 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1486938

RESUMEN

Adjuvant is an essential component in subunit vaccines. Many agonists of pathogen recognition receptors have been developed as potent adjuvants to optimize the immunogenicity and efficacy of vaccines. Recently discovered cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has attracted much attention as it is a key mediator for modulating immune responses. Vaccines adjuvanted with STING agonists are found to mediate a robust immune defense against infections and cancer. In this review, we first discuss the mechanisms of STING agonists in the context of vaccination. Next, we present recent progress in novel STING agonist discovery and the delivery strategies. We next highlight recent work in optimizing the efficacy while minimizing toxicity of STING agonist-assisted subunit vaccines for protection against infectious diseases or treatment of cancer. Finally, we share our perspectives of current issues and future directions in further developing STING agonists for adjuvanting subunit vaccines.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Vacunas de Subunidad/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Humanos , Inmunidad Humoral/efectos de los fármacos , Nucleotidiltransferasas/metabolismo
3.
Front Immunol ; 12: 732992, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1497075

RESUMEN

Chronic inflammatory disorders (CID), such as autoimmune diseases, are characterized by overactivation of the immune system and loss of immune tolerance. T helper 17 (Th17) cells are strongly associated with the pathogenesis of multiple CID, including psoriasis, rheumatoid arthritis, and inflammatory bowel disease. In line with the increasingly recognized contribution of innate immune cells to the modulation of dendritic cell (DC) function and DC-driven adaptive immune responses, we recently showed that neutrophils are required for DC-driven Th17 cell differentiation from human naive T cells. Consequently, recruitment of neutrophils to inflamed tissues and lymph nodes likely creates a highly inflammatory loop through the induction of Th17 cells that should be intercepted to attenuate disease progression. Tolerogenic therapy via DCs, the central orchestrators of the adaptive immune response, is a promising strategy for the treatment of CID. Tolerogenic DCs could restore immune tolerance by driving the development of regulatory T cells (Tregs) in the periphery. In this review, we discuss the effects of the tolerogenic adjuvants vitamin D3 (VD3), corticosteroids (CS), and retinoic acid (RA) on both DCs and neutrophils and their potential interplay. We briefly summarize how neutrophils shape DC-driven T-cell development in general. We propose that, for optimization of tolerogenic DC therapy for the treatment of CID, both DCs for tolerance induction and the neutrophil inflammatory loop should be targeted while preserving the potential Treg-enhancing effects of neutrophils.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Autoinmunidad/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
4.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1409704

RESUMEN

Autotaxin (ATX; ENPP2) is a secreted lysophospholipase D catalyzing the extracellular production of lysophosphatidic acid (LPA), a pleiotropic signaling phospholipid. Genetic and pharmacologic studies have previously established a pathologic role for ATX and LPA signaling in pulmonary injury, inflammation, and fibrosis. Here, increased ENPP2 mRNA levels were detected in immune cells from nasopharyngeal swab samples of COVID-19 patients, and increased ATX serum levels were found in severe COVID-19 patients. ATX serum levels correlated with the corresponding increased serum levels of IL-6 and endothelial damage biomarkers, suggesting an interplay of the ATX/LPA axis with hyperinflammation and the associated vascular dysfunction in COVID-19. Accordingly, dexamethasone (Dex) treatment of mechanically ventilated patients reduced ATX levels, as shown in two independent cohorts, indicating that the therapeutic benefits of Dex include the suppression of ATX. Moreover, large scale analysis of multiple single cell RNA sequencing datasets revealed the expression landscape of ENPP2 in COVID-19 and further suggested a role for ATX in the homeostasis of dendritic cells, which exhibit both numerical and functional deficits in COVID-19. Therefore, ATX has likely a multifunctional role in COVID-19 pathogenesis, suggesting that its pharmacological targeting might represent an additional therapeutic option, both during and after hospitalization.


Asunto(s)
COVID-19/diagnóstico , Células Dendríticas/inmunología , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/sangre , SARS-CoV-2/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , COVID-19/sangre , COVID-19/inmunología , COVID-19/terapia , Estudios de Cohortes , Conjuntos de Datos como Asunto , Células Dendríticas/efectos de los fármacos , Dexametasona/farmacología , Dexametasona/uso terapéutico , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Femenino , Humanos , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , RNA-Seq , Respiración Artificial , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Análisis de la Célula Individual
5.
Adv Mater ; 33(40): e2102528, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: covidwho-1358054

RESUMEN

Dendritic cell (DC) vaccines are used for cancer and infectious diseases, albeit with limited efficacy. Modulating the formation of DC-T-cell synapses may greatly increase their efficacy. The effects of graphene oxide (GO) nanosheets on DCs and DC-T-cell synapse formation are evaluated. In particular, size-dependent interactions are observed between GO nanosheets and DCs. GOs with diameters of >1 µm (L-GOs) demonstrate strong adherence to the DC surface, inducing cytoskeletal reorganization via the RhoA-ROCK-MLC pathway, while relatively small GOs (≈500 nm) are predominantly internalized by DCs. Furthermore, L-GO treatment enhances DC-T-cell synapse formation via cytoskeleton-dependent membrane positioning of integrin ICAM-1. L-GO acts as a "nanozipper," facilitating the aggregation of DC-T-cell clusters to produce a stable microenvironment for T cell activation. Importantly, L-GO-adjuvanted DCs promote robust cytotoxic T cell immune responses against SARS-CoV-2 spike 1, leading to >99.7% viral RNA clearance in mice infected with a clinically isolated SARS-CoV-2 strain. These findings highlight the potential value of nanomaterials as DC vaccine adjuvants for modulating DC-T-cell synapse formation and provide a basis for the development of effective COVID-19 vaccines.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra la COVID-19/uso terapéutico , COVID-19/prevención & control , Células Dendríticas/inmunología , Grafito/uso terapéutico , Nanoestructuras/uso terapéutico , Adyuvantes Inmunológicos/química , Animales , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Células Dendríticas/efectos de los fármacos , Grafito/química , Humanos , Ratones , Nanoestructuras/química , SARS-CoV-2/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
7.
Front Immunol ; 12: 635018, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1211810

RESUMEN

Objective: Bacterial and viral infectious triggers are linked to spondyloarthritis (SpA) including psoriatic arthritis (PsA) development, likely via dendritic cell activation. We investigated spinal entheseal plasmacytoid dendritic cells (pDCs) toll-like receptor (TLR)-7 and 9 activation and therapeutic modulation, including JAK inhibition. We also investigated if COVID-19 infection, a potent TLR-7 stimulator triggered PsA flares. Methods: Normal entheseal pDCs were characterized and stimulated with imiquimod and CpG oligodeoxynucleotides (ODN) to evaluate TNF and IFNα production. NanoString gene expression assay of total pDCs RNA was performed pre- and post- ODN stimulation. Pharmacological inhibition of induced IFNα protein was performed with Tofacitinib and PDE4 inhibition. The impact of SARS-CoV2 viral infection on PsA flares was evaluated. Results: CD45+HLA-DR+CD123+CD303+CD11c- entheseal pDCs were more numerous than blood pDCs (1.9 ± 0.8% vs 0.2 ± 0.07% of CD45+ cells, p=0.008) and showed inducible IFNα and TNF protein following ODN/imiquimod stimulation and were the sole entheseal IFNα producers. NanoString data identified 11 significantly upregulated differentially expressed genes (DEGs) including TNF in stimulated pDCs. Canonical pathway analysis revealed activation of dendritic cell maturation, NF-κB signaling, toll-like receptor signaling and JAK/STAT signaling pathways following ODN stimulation. Both tofacitinib and PDE4i strongly attenuated ODN induced IFNα. DAPSA scores elevations occurred in 18 PsA cases with SARS-CoV2 infection (9.7 ± 4 pre-infection and 35.3 ± 7.5 during infection). Conclusion: Entheseal pDCs link microbes to TNF/IFNα production. SARS-CoV-2 infection is associated with PsA Flares and JAK inhibition suppressed activated entheseal plasmacytoid dendritic Type-1 interferon responses as pointers towards a novel mechanism of PsA and SpA-related arthropathy.


Asunto(s)
Artritis Psoriásica/complicaciones , COVID-19/complicaciones , Células Dendríticas/metabolismo , Interferón-alfa/metabolismo , Quinasas Janus/antagonistas & inhibidores , Adyuvantes Inmunológicos/farmacología , Adulto , Anciano , COVID-19/genética , COVID-19/metabolismo , Biología Computacional , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Células Dendríticas/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Imiquimod/farmacología , Quinasas Janus/metabolismo , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Oligonucleótidos/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo
8.
Prostaglandins Leukot Essent Fatty Acids ; 161: 102177, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-796199

RESUMEN

As the infected cases of COVID-19 reach more than 20 million with more than 778,000 deaths globally, an increase in psychiatric disorders including anxiety and depression has been reported. Scientists globally have been searching for novel therapies and vaccines to fight against COVID-19. Improving innate immunity has been suggested to block progression of COVID-19 at early stages, while omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been shown to have immunomodulation effects. Moreover, n-3 PUFAs have also been shown to improve mood disorders, thus, future research is warranted to test if n-3 PUFAs may have the potential to improve our immunity to counteract both physical and mental impact of COVID-19.


Asunto(s)
Ansiedad/prevención & control , Infecciones por Coronavirus/prevención & control , Depresión/prevención & control , Suplementos Dietéticos , Ácidos Grasos Omega-3/administración & dosificación , Factores Inmunológicos/administración & dosificación , Pandemias/prevención & control , Neumonía Viral/prevención & control , Ansiedad/inmunología , Ansiedad/metabolismo , Ansiedad/virología , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Depresión/inmunología , Depresión/metabolismo , Depresión/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Ácidos Grasos Omega-3/inmunología , Ácidos Grasos Omega-3/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/virología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Neumonía Viral/inmunología , Neumonía Viral/metabolismo , Neumonía Viral/virología , SARS-CoV-2
9.
Vaccine ; 38(42): 6487-6499, 2020 09 29.
Artículo en Inglés | MEDLINE | ID: covidwho-720733

RESUMEN

The many carbohydrate chains on Covid-19 coronavirus SARS-CoV-2 and its S-protein form a glycan-shield that masks antigenic peptides and decreases uptake of inactivated virus or S-protein vaccines by APC. Studies on inactivated influenza virus and recombinant gp120 of HIV vaccines indicate that glycoengineering of glycan-shields to present α-gal epitopes (Galα1-3Galß1-4GlcNAc-R) enables harnessing of the natural anti-Gal antibody for amplifying vaccine efficacy, as evaluated in mice producing anti-Gal. The α-gal epitope is the ligand for the natural anti-Gal antibody which constitutes ~1% of immunoglobulins in humans. Upon administration of vaccines presenting α-gal epitopes, anti-Gal binds to these epitopes at the vaccination site and forms immune complexes with the vaccines. These immune complexes are targeted for extensive uptake by APC as a result of binding of the Fc portion of immunocomplexed anti-Gal to Fc receptors on APC. This anti-Gal mediated effective uptake of vaccines by APC results in 10-200-fold higher anti-viral immune response and in 8-fold higher survival rate following challenge with a lethal dose of live influenza virus, than same vaccines lacking α-gal epitopes. It is suggested that glycoengineering of carbohydrate chains on the glycan-shield of inactivated SARS-CoV-2 or on S-protein vaccines, for presenting α-gal epitopes, will have similar amplifying effects on vaccine efficacy. α-Gal epitope synthesis on coronavirus vaccines can be achieved with recombinant α1,3galactosyltransferase, replication of the virus in cells with high α1,3galactosyltransferase activity as a result of stable transfection of cells with several copies of the α1,3galactosyltransferase gene (GGTA1), or by transduction of host cells with replication defective adenovirus containing this gene. In addition, recombinant S-protein presenting multiple α-gal epitopes on the glycan-shield may be produced in glycoengineered yeast or bacteria expression systems containing the corresponding glycosyltransferases. Prospective Covid-19 vaccines presenting α-gal epitopes may provide better protection than vaccines lacking this epitope because of increased uptake by APC.


Asunto(s)
Antígenos Virales/genética , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/prevención & control , Glicoproteína de la Espiga del Coronavirus/genética , Trisacáridos/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/virología , Ingeniería Genética , Proteína p24 del Núcleo del VIH/química , Proteína p24 del Núcleo del VIH/genética , Proteína p24 del Núcleo del VIH/inmunología , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/inmunología , Humanos , Inmunogenicidad Vacunal , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Ratones , Neumonía Viral/inmunología , Neumonía Viral/virología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Trisacáridos/química , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis , Vacunas Virales/genética
10.
PLoS Pathog ; 16(7): e1008737, 2020 07.
Artículo en Inglés | MEDLINE | ID: covidwho-691046

RESUMEN

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the current COVID-19 pandemic. An unbalanced immune response, characterized by a weak production of type I interferons (IFN-Is) and an exacerbated release of proinflammatory cytokines, contributes to the severe forms of the disease. SARS-CoV-2 is genetically related to SARS-CoV and Middle East respiratory syndrome-related coronavirus (MERS-CoV), which caused outbreaks in 2003 and 2013, respectively. Although IFN treatment gave some encouraging results against SARS-CoV and MERS-CoV in animal models, its potential as a therapeutic against COVID-19 awaits validation. Here, we describe our current knowledge of the complex interplay between SARS-CoV-2 infection and the IFN system, highlighting some of the gaps that need to be filled for a better understanding of the underlying molecular mechanisms. In addition to the conserved IFN evasion strategies that are likely shared with SARS-CoV and MERS-CoV, novel counteraction mechanisms are being discovered in SARS-CoV-2-infected cells. Since the last coronavirus epidemic, we have made considerable progress in understanding the IFN-I response, including its spatiotemporal regulation and the prominent role of plasmacytoid dendritic cells (pDCs), which are the main IFN-I-producing cells. While awaiting the results of the many clinical trials that are evaluating the efficacy of IFN-I alone or in combination with antiviral molecules, we discuss the potential benefits of a well-timed IFN-I treatment and propose strategies to boost pDC-mediated IFN responses during the early stages of viral infection.


Asunto(s)
Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , Infecciones por Coronavirus/tratamiento farmacológico , Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Células Dendríticas/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/virología , Pronóstico , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19
11.
J Transl Med ; 18(1): 261, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: covidwho-617269

RESUMEN

Amino-bisphosphonates such as zoledronic acid (ZA) can possibly ameliorate or prevent severe COVID-19 disease by at least three distinct mechanisms: (1) as immunostimulants which could boost γδ T cell expansion, important in the acute response in the lung; (2) as DC modulators, limiting their ability to only partially activate T cells; and (3) as prenylation inhibitors of small GTPases in the endosomal pathway of the DC to prevent expulsion of lysosomes containing SARS-CoV-2 virions. Use of ZA or other amino-bisphosphonates as modulators of COVID-19 disease should be considered.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Células Dendríticas/virología , Difosfonatos/uso terapéutico , Endosomas/metabolismo , Factores Inmunológicos/farmacología , Neumonía Viral/inmunología , Neumonía Viral/virología , Animales , COVID-19 , Células Dendríticas/efectos de los fármacos , Endosomas/efectos de los fármacos , Humanos , Pandemias , SARS-CoV-2
12.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Artículo en Inglés | MEDLINE | ID: covidwho-186413

RESUMEN

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Citocinas/genética , Células Dendríticas/efectos de los fármacos , Factores Inmunológicos/farmacología , Macrófagos/efectos de los fármacos , Triterpenos/farmacología , Betacoronavirus/crecimiento & desarrollo , Betacoronavirus/inmunología , Diferenciación Celular/efectos de los fármacos , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/inmunología , Citocinas/clasificación , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Ebolavirus/efectos de los fármacos , Ebolavirus/crecimiento & desarrollo , Ebolavirus/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Virus de la Hepatitis E/efectos de los fármacos , Virus de la Hepatitis E/crecimiento & desarrollo , Virus de la Hepatitis E/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Especificidad de Órganos , Picornaviridae/efectos de los fármacos , Picornaviridae/crecimiento & desarrollo , Picornaviridae/inmunología , Cultivo Primario de Células , SARS-CoV-2 , Transducción de Señal , Virus Zika/efectos de los fármacos , Virus Zika/crecimiento & desarrollo , Virus Zika/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA