Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
2.
Microbiol Spectr ; 10(5): e0237122, 2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: covidwho-2008768

RESUMEN

Diverse severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have emerged since the beginning of the COVID-19 pandemic. We investigated the immunological and pathological peculiarity of the SARS-CoV-2 beta variant of concern (VoC) compared to the ancestral strain. Comparative analysis of phenotype and pathology revealed that the beta VoC induces slower disease progression and a prolonged presymptomatic period in the early stages of SARS-CoV-2 infection but ultimately causes sudden death in the late stages of infection in the K18-hACE2 mouse model. The beta VoC induced enhanced activation of CXCL1/2-CXCR2-NLRP3-IL-1ß signal cascade accelerating neutrophil recruitment and lung pathology in beta variant-infected mice, as evidenced by multiple analyses of SARS-CoV-2-induced inflammatory cytokines and transcriptomes. CCL2 was one of the most highly secreted cytokines in the early stages of infection. Its blockade reduced virus-induced weight loss and delayed mortality. Our study provides a better understanding of the variant characteristics and need for treatment. IMPORTANCE Since the outbreak of COVID-19, diverse SARS-CoV-2 variants have been identified. These variants have different infectivity and transmissibility from the ancestral strains. However, underlying molecular mechanisms have not yet been fully elucidated. In our study, the beta variant showed distinct pathological conditions and cytokine release kinetics from an ancestral strain in a mouse model. It was associated with higher neutrophil recruitment by increased levels of CXCL1/2, CXCR2, and interleukin 1ß (IL-1ß) at a later stage of viral infection. Our study will provide a better understanding of SARS-CoV-2 pathogenesis.


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Humanos , Animales , Pandemias , Interleucina-1beta/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Citocinas , Modelos Animales de Enfermedad
3.
J Virol ; 96(18): e0102422, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: covidwho-2008764

RESUMEN

Zoonotic coronaviruses represent an ongoing threat to public health. The classical porcine epidemic diarrhea virus (PEDV) first appeared in the early 1970s. Since 2010, outbreaks of highly virulent PEDV variants have caused great economic losses to the swine industry worldwide. However, the strategies by which PEDV variants escape host immune responses are not fully understood. Complement component 3 (C3) is considered a central component of the three complement activation pathways and plays a crucial role in preventing viral infection. In this study, we found that C3 significantly inhibited PEDV replication in vitro, and both variant and classical PEDV strains induced high levels of interleukin-1ß (IL-1ß) in Huh7 cells. However, the PEDV variant strain reduces C3 transcript and protein levels induced by IL-1ß compared with the PEDV classical strain. Examination of key molecules of the C3 transcriptional signaling pathway revealed that variant PEDV reduced C3 by inhibiting CCAAT/enhancer-binding protein ß (C/EBP-ß) phosphorylation. Mechanistically, PEDV nonstructural protein 1 (NSP1) inhibited C/EBP-ß phosphorylation via amino acid residue 50. Finally, we constructed recombinant PEDVs to verify the critical role of amino acid 50 of NSP1 in the regulation of C3 expression. In summary, we identified a novel antiviral role of C3 in inhibiting PEDV replication and the viral immune evasion strategies of PEDV variants. Our study reveals new information on PEDV-host interactions and furthers our understanding of the pathogenic mechanism of this virus. IMPORTANCE The complement system acts as a vital link between the innate and the adaptive immunity and has the ability to recognize and neutralize various pathogens. Activation of the complement system acts as a double-edged sword, as appropriate levels of activation protect against pathogenic infections, but excessive responses can provoke a dramatic inflammatory response and cause tissue damage, leading to pathological processes, which often appear in COVID-19 patients. However, how PEDV, as the most severe coronavirus causing diarrhea in piglets, regulates the complement system has not been previously reported. In this study, for the first time, we identified a novel mechanism of a PEDV variant in the suppression of C3 expression, showing that different coronaviruses and even different subtype strains differ in regulation of C3 expression. In addition, this study provides a deeper understanding of the mechanism of the PEDV variant in immune escape and enhanced virulence.


Asunto(s)
Complemento C3 , Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Proteínas no Estructurales Virales , Replicación Viral , Animales , Antivirales , COVID-19/inmunología , Línea Celular Tumoral , Complemento C3/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología
4.
Stem Cell Reports ; 17(3): 538-555, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1692861

RESUMEN

To date, the direct causative mechanism of SARS-CoV-2-induced endotheliitis remains unclear. Here, we report that human ECs barely express surface ACE2, and ECs express less intracellular ACE2 than non-ECs of the lungs. We ectopically expressed ACE2 in hESC-ECs to model SARS-CoV-2 infection. ACE2-deficient ECs are resistant to the infection but are more activated than ACE2-expressing ones. The virus directly induces endothelial activation by increasing monocyte adhesion, NO production, and enhanced phosphorylation of p38 mitogen-associated protein kinase (MAPK), NF-κB, and eNOS in ACE2-expressing and -deficient ECs. ACE2-deficient ECs respond to SARS-CoV-2 through TLR4 as treatment with its antagonist inhibits p38 MAPK/NF-κB/ interleukin-1ß (IL-1ß) activation after viral exposure. Genome-wide, single-cell RNA-seq analyses further confirm activation of the TLR4/MAPK14/RELA/IL-1ß axis in circulating ECs of mild and severe COVID-19 patients. Circulating ECs could serve as biomarkers for indicating patients with endotheliitis. Together, our findings support a direct role for SARS-CoV-2 in mediating endothelial inflammation in an ACE2-dependent or -independent manner.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Modelos Biológicos , SARS-CoV-2/fisiología , Receptor Toll-Like 4/metabolismo , Enzima Convertidora de Angiotensina 2/genética , COVID-19/patología , COVID-19/virología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , SARS-CoV-2/aislamiento & purificación , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Sci Rep ; 11(1): 24432, 2021 12 24.
Artículo en Inglés | MEDLINE | ID: covidwho-1585772

RESUMEN

Despite the initial success of some drugs and vaccines targeting COVID-19, understanding the mechanism underlying SARS-CoV-2 disease pathogenesis remains crucial for the development of further approaches to treatment. Some patients with severe Covid-19 experience a cytokine storm and display evidence of inflammasome activation leading to increased levels of IL-1ß and IL-18; however, other reports have suggested reduced inflammatory responses to Sars-Cov-2. In this study we have examined the effects of the Sars-Cov-2 envelope (E) protein, a virulence factor in coronaviruses, on inflammasome activation and pulmonary inflammation. In cultured macrophages the E protein suppressed inflammasome priming and NLRP3 inflammasome activation. Similarly, in mice transfected with E protein and treated with poly(I:C) to simulate the effects of viral RNA, the E protein, in an NLRP3-dependent fashion, reduced expression of pro-IL-1ß, levels of IL-1ß and IL-18 in broncho-alveolar lavage fluid, and macrophage infiltration in the lung. To simulate the effects of more advanced infection, macrophages were treated with both LPS and poly(I:C). In this setting the E protein increased NLRP3 inflammasome activation in both murine and human macrophages. Thus, the Sars-Cov-2 E protein may initially suppress the host NLRP3 inflammasome response to viral RNA while potentially increasing NLRP3 inflammasome responses in the later stages of infection. Targeting the Sars-Cov-2 E protein especially in the early stages of infection may represent a novel approach to Covid-19 therapy.


Asunto(s)
Proteínas de la Envoltura de Coronavirus/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , SARS-CoV-2/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , COVID-19/patología , COVID-19/virología , Proteínas de la Envoltura de Coronavirus/genética , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico , Humanos , Inflamasomas/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Quinasas Janus/genética , Quinasas Janus/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Poli I-C/farmacología , ARN Viral/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/aislamiento & purificación
6.
Sci Rep ; 11(1): 23695, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1561520

RESUMEN

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread across the world. Inactivating the virus in saliva and the oral cavity represents a reasonable approach to prevent human-to-human transmission because the virus is easily transmitted through oral routes by dispersed saliva. Persimmon-derived tannin is a condensed type of tannin that has strong antioxidant and antimicrobial activity. In this study, we investigated the antiviral effects of persimmon-derived tannin against SARS-CoV-2 in both in vitro and in vivo models. We found that persimmon-derived tannin suppressed SARS-CoV-2 titers measured by plaque assay in vitro in a dose- and time-dependent manner. We then created a Syrian hamster model by inoculating SARS-CoV-2 into hamsters' mouths. Oral administration of persimmon-derived tannin dissolved in carboxymethyl cellulose before virus inoculation dramatically reduced the severity of pneumonia with lower virus titers compared with a control group inoculated with carboxymethyl cellulose alone. In addition, pre-administration of tannin to uninfected hamsters reduced hamster-to-hamster transmission of SARS-CoV-2 from a cohoused, infected donor cage mate. These data suggest that oral administration of persimmon-derived tannin may help reduce the severity of SARS-CoV-2 infection and transmission of the virus.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Diospyros/química , Taninos/uso terapéutico , Administración Oral , Animales , Antivirales/química , Antivirales/aislamiento & purificación , Antivirales/farmacología , COVID-19/patología , COVID-19/transmisión , COVID-19/virología , Cricetinae , Diospyros/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Mesocricetus , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad , Taninos/química , Taninos/aislamiento & purificación , Taninos/farmacología , Carga Viral/efectos de los fármacos
7.
Nutrients ; 13(11)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1512522

RESUMEN

BACKGROUND: We aimed to establish an acute treatment protocol to increase serum vitamin D, evaluate the effectiveness of vitamin D3 supplementation, and reveal the potential mechanisms in COVID-19. METHODS: We retrospectively analyzed the data of 867 COVID-19 cases. Then, a prospective study was conducted, including 23 healthy individuals and 210 cases. A total of 163 cases had vitamin D supplementation, and 95 were followed for 14 days. Clinical outcomes, routine blood biomarkers, serum levels of vitamin D metabolism, and action mechanism-related parameters were evaluated. RESULTS: Our treatment protocol increased the serum 25OHD levels significantly to above 30 ng/mL within two weeks. COVID-19 cases (no comorbidities, no vitamin D treatment, 25OHD <30 ng/mL) had 1.9-fold increased risk of having hospitalization longer than 8 days compared with the cases with comorbidities and vitamin D treatment. Having vitamin D treatment decreased the mortality rate by 2.14 times. The correlation analysis of specific serum biomarkers with 25OHD indicated that the vitamin D action in COVID-19 might involve regulation of INOS1, IL1B, IFNg, cathelicidin-LL37, and ICAM1. CONCLUSIONS: Vitamin D treatment shortened hospital stay and decreased mortality in COVID-19 cases, even in the existence of comorbidities. Vitamin D supplementation is effective on various target parameters; therefore, it is essential for COVID-19 treatment.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Vitamina D/administración & dosificación , Péptidos Catiónicos Antimicrobianos/sangre , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , COVID-19/complicaciones , COVID-19/mortalidad , Suplementos Dietéticos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/sangre , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/sangre , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-1beta/sangre , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Óxido Nítrico Sintasa de Tipo II/sangre , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estudios Prospectivos , Estudios Retrospectivos , Vitamina D/sangre , Vitamina D/farmacología , Vitaminas/administración & dosificación , Vitaminas/farmacología , Catelicidinas
8.
Mol Neurobiol ; 59(1): 445-458, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: covidwho-1491383

RESUMEN

In addition to respiratory complications produced by SARS-CoV-2, accumulating evidence suggests that some neurological symptoms are associated with the disease caused by this coronavirus. In this study, we investigated the effects of the SARS-CoV-2 spike protein S1 stimulation on neuroinflammation in BV-2 microglia. Analyses of culture supernatants revealed an increase in the production of TNF-α, IL-6, IL-1ß and iNOS/NO. S1 also increased protein levels of phospho-p65 and phospho-IκBα, as well as enhanced DNA binding and transcriptional activity of NF-κB. These effects of the protein were blocked in the presence of BAY11-7082 (1 µM). Exposure of S1 to BV-2 microglia also increased the protein levels of NLRP3 inflammasome and enhanced caspase-1 activity. Increased protein levels of p38 MAPK was observed in BV-2 microglia stimulated with the spike protein S1 (100 ng/ml), an action that was reduced in the presence of SKF 86,002 (1 µM). Results of immunofluorescence microscopy showed an increase in TLR4 protein expression in S1-stimulated BV-2 microglia. Furthermore, pharmacological inhibition with TAK 242 (1 µM) and transfection with TLR4 small interfering RNA resulted in significant reduction in TNF-α and IL-6 production in S1-stimulated BV-2 microglia. These results have provided the first evidence demonstrating S1-induced neuroinflammation in BV-2 microglia. We propose that induction of neuroinflammation by this protein in the microglia is mediated through activation of NF-κB and p38 MAPK, possibly as a result of TLR4 activation. These results contribute to our understanding of some of the mechanisms involved in CNS pathologies of SARS-CoV-2.


Asunto(s)
Microglía/metabolismo , Enfermedades Neuroinflamatorias/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Caspasa 1/metabolismo , Línea Celular , Furanos/farmacología , Indenos/farmacología , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-6/metabolismo , Ratones , Microglía/patología , FN-kappa B/metabolismo , Enfermedades Neuroinflamatorias/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitrilos/farmacología , ARN Interferente Pequeño , Proteínas Recombinantes/metabolismo , Sulfonamidas/farmacología , Sulfonas/farmacología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Molecules ; 26(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1470935

RESUMEN

Excessive host inflammation following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is associated with severity and mortality in coronavirus disease 2019 (COVID-19). We recently reported that the SARS-CoV-2 spike protein S1 subunit (S1) induces pro-inflammatory responses by activating toll-like receptor 4 (TLR4) signaling in macrophages. A standardized extract of Asparagus officinalis stem (EAS) is a unique functional food that elicits anti-photoaging effects by suppressing pro-inflammatory signaling in hydrogen peroxide and ultraviolet B-exposed skin fibroblasts. To elucidate its potential in preventing excessive inflammation in COVID-19, we examined the effects of EAS on pro-inflammatory responses in S1-stimulated macrophages. Murine peritoneal exudate macrophages were co-treated with EAS and S1. Concentrations and mRNA levels of pro-inflammatory cytokines were assessed using enzyme-linked immunosorbent assay and reverse transcription and real-time polymerase chain reaction, respectively. Expression and phosphorylation levels of signaling proteins were analyzed using western blotting and fluorescence immunomicroscopy. EAS significantly attenuated S1-induced secretion of interleukin (IL)-6 in a concentration-dependent manner without reducing cell viability. EAS also markedly suppressed the S1-induced transcription of IL-6 and IL-1ß. However, among the TLR4 signaling proteins, EAS did not affect the degradation of inhibitor κBα, nuclear translocation of nuclear factor-κB p65 subunit, and phosphorylation of c-Jun N-terminal kinase p54 subunit after S1 exposure. In contrast, EAS significantly suppressed S1-induced phosphorylation of p44/42 mitogen-activated protein kinase (MAPK) and Akt. Attenuation of S1-induced transcription of IL-6 and IL-1ß by the MAPK kinase inhibitor U0126 was greater than that by the Akt inhibitor perifosine, and the effects were potentiated by simultaneous treatment with both inhibitors. These results suggest that EAS attenuates S1-induced IL-6 and IL-1ß production by suppressing p44/42 MAPK and Akt signaling in macrophages. Therefore, EAS may be beneficial in regulating excessive inflammation in patients with COVID-19.


Asunto(s)
Asparagus/química , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Asparagus/metabolismo , Butadienos/farmacología , Supervivencia Celular/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-6/genética , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Extractos Vegetales/química , Tallos de la Planta/química , Tallos de la Planta/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glicoproteína de la Espiga del Coronavirus/farmacología , Receptor Toll-Like 4/metabolismo , Transcripción Genética/efectos de los fármacos
10.
Arterioscler Thromb Vasc Biol ; 41(9): 2509-2511, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1405256
11.
Am J Respir Cell Mol Biol ; 63(5): 707-709, 2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1388616
12.
Zool Res ; 42(5): 633-636, 2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: covidwho-1369995

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent responsible for the global coronavirus disease 2019 (COVID-19) pandemic. Numerous studies have demonstrated that cardiovascular disease may affect COVID-19 progression. In the present study, we investigated the effect of hypertension on viral replication and COVID-19 progression using a hypertensive mouse model infected with SARS-CoV-2. Results revealed that SARS-CoV-2 replication was delayed in hypertensive mouse lungs. In contrast, SARS-CoV-2 replication in hypertensive mice treated with the antihypertensive drug captopril demonstrated similar virus replication as SARS-CoV-2-infected normotensive mice. Furthermore, antihypertensive treatment alleviated lung inflammation induced by SARS-CoV-2 replication (interleukin (IL)-1ß up-regulation and increased immune cell infiltration). No differences in lung inflammation were observed between the SARS-CoV-2-infected normotensive mice and hypertensive mice. Our findings suggest that captopril treatment may alleviate COVID-19 progression but not affect viral replication.


Asunto(s)
Antihipertensivos/uso terapéutico , COVID-19/complicaciones , Captopril/uso terapéutico , Hipertensión/complicaciones , Enfermedades Pulmonares/tratamiento farmacológico , SARS-CoV-2 , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Antihipertensivos/farmacología , Captopril/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/virología , Ratones , Replicación Viral/efectos de los fármacos
13.
Mol Med Rep ; 24(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: covidwho-1271003

RESUMEN

Coronavirus disease 2019 (COVID­19), caused by the severe acute respiratory syndrome coronavirus­2 (SARS­CoV­2), led to an outbreak of viral pneumonia in December 2019. The present study aimed to investigate the host inflammatory response signature­caused by SARS­CoV­2 in human corneal epithelial cells (HCECs). The expression level of angiotensin­converting enzyme 2 (ACE2) in the human cornea was determined via immunofluorescence. In vitro experiments were performed in HCECs stimulated with the SARS­CoV­2 spike protein. Moreover, the expression levels of ACE2, IL­8, TNF­α, IL­6, gasdermin D (GSDMD) and IL­1ß in HCECs were detected using reverse transcription­quantitative PCR and/or western blotting. It was identified that ACE2 was expressed in normal human corneal epithelium and HCECs cultured in vitro. Furthermore, the expression levels of IL­8, TNF­α and IL­6 in HCECs were decreased following SARS­CoV­2 spike protein stimulation, while the expression levels of GSDMD and IL­1ß were increased. In conclusion, the present results demonstrated that the SARS­CoV­2 spike protein suppressed the host inflammatory response and induced pyroptosis in HCECs. Therefore, blocking the ACE2 receptor in HCECs may reduce the infection rate of COVID­19.


Asunto(s)
Epitelio Corneal/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Adulto , Anciano , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Células Cultivadas , Córnea/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Epitelio Corneal/virología , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo , Piroptosis , Glicoproteína de la Espiga del Coronavirus/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
14.
Immunity ; 54(7): 1463-1477.e11, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1263294

RESUMEN

Acute respiratory distress syndrome (ARDS), an inflammatory condition with high mortality rates, is common in severe COVID-19, whose risk is reduced by metformin rather than other anti-diabetic medications. Detecting of inflammasome assembly in post-mortem COVID-19 lungs, we asked whether and how metformin inhibits inflammasome activation while exerting its anti-inflammatory effect. We show that metformin inhibited NLRP3 inflammasome activation and interleukin (IL)-1ß production in cultured and alveolar macrophages along with inflammasome-independent IL-6 secretion, thus attenuating lipopolysaccharide (LPS)- and SARS-CoV-2-induced ARDS. By targeting electron transport chain complex 1 and independently of AMP-activated protein kinase (AMPK) or NF-κB, metformin blocked LPS-induced and ATP-dependent mitochondrial (mt) DNA synthesis and generation of oxidized mtDNA, an NLRP3 ligand. Myeloid-specific ablation of LPS-induced cytidine monophosphate kinase 2 (CMPK2), which is rate limiting for mtDNA synthesis, reduced ARDS severity without a direct effect on IL-6. Thus, inhibition of ATP and mtDNA synthesis is sufficient for ARDS amelioration.


Asunto(s)
Adenosina Trifosfato/metabolismo , ADN Mitocondrial/biosíntesis , Inflamasomas/efectos de los fármacos , Metformina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/prevención & control , Animales , COVID-19/metabolismo , COVID-19/prevención & control , Citocinas/genética , Citocinas/metabolismo , ADN Mitocondrial/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Metformina/uso terapéutico , Ratones , Nucleósido-Fosfato Quinasa/metabolismo , Neumonía/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2/patogenicidad
15.
Int J Mol Sci ; 22(10)2021 May 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1236794

RESUMEN

Acute lung injury (ALI) afflicts approximately 200,000 patients annually and has a 40% mortality rate. The COVID-19 pandemic has massively increased the rate of ALI incidence. The pathogenesis of ALI involves tissue damage from invading microbes and, in severe cases, the overexpression of inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß). This study aimed to develop a therapy to normalize the excess production of inflammatory cytokines and promote tissue repair in the lipopolysaccharide (LPS)-induced ALI. Based on our previous studies, we tested the insulin-like growth factor I (IGF-I) and BTP-2 therapies. IGF-I was selected, because we and others have shown that elevated inflammatory cytokines suppress the expression of growth hormone receptors in the liver, leading to a decrease in the circulating IGF-I. IGF-I is a growth factor that increases vascular protection, enhances tissue repair, and decreases pro-inflammatory cytokines. It is also required to produce anti-inflammatory 1,25-dihydroxyvitamin D. BTP-2, an inhibitor of cytosolic calcium, was used to suppress the LPS-induced increase in cytosolic calcium, which otherwise leads to an increase in proinflammatory cytokines. We showed that LPS increased the expression of the primary inflammatory mediators such as toll like receptor-4 (TLR-4), IL-1ß, interleukin-17 (IL-17), TNF-α, and interferon-γ (IFN-γ), which were normalized by the IGF-I + BTP-2 dual therapy in the lungs, along with improved vascular gene expression markers. The histologic lung injury score was markedly elevated by LPS and reduced to normal by the combination therapy. In conclusion, the LPS-induced increases in inflammatory cytokines, vascular injuries, and lung injuries were all improved by IGF-I + BTP-2 combination therapy.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Anilidas/farmacología , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Tiadiazoles/farmacología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Anilidas/uso terapéutico , Animales , COVID-19/complicaciones , Calcio/metabolismo , Canales de Calcio/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tiadiazoles/uso terapéutico , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Am J Obstet Gynecol ; 225(3): 301.e1-301.e14, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1226265

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2, the disease-causing pathogen of the coronavirus disease 2019 pandemic, has resulted in morbidity and mortality worldwide. Pregnant women are more susceptible to severe coronavirus disease 2019 and are at higher risk of preterm birth than uninfected pregnant women. Despite this evidence, the immunologic effects of severe acute respiratory syndrome coronavirus 2 infection during pregnancy remain understudied. OBJECTIVE: This study aimed to assess the impact of severe acute respiratory syndrome coronavirus 2 infection during pregnancy on inflammatory and humoral responses in maternal and fetal samples and compare antibody responses to severe acute respiratory syndrome coronavirus 2 among pregnant and nonpregnant women. STUDY DESIGN: Immune responses to severe acute respiratory syndrome coronavirus 2 were analyzed using samples from pregnant (n=33) and nonpregnant (n=17) women who tested either positive (pregnant, 22; nonpregnant, 17) or negative for severe acute respiratory syndrome coronavirus 2 (pregnant, 11) at Johns Hopkins Hospital. We measured proinflammatory and placental cytokine messenger RNAs, neonatal Fc receptor expression, and tetanus antibody transfer in maternal and cord blood samples. In addition, we evaluated antispike immunoglobulin G, antispike receptor-binding domain immunoglobulin G, and neutralizing antibody responses to severe acute respiratory syndrome coronavirus 2 in serum or plasma collected from nonpregnant women, pregnant women, and cord blood. RESULTS: Pregnant women with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection expressed more interleukin-1 beta, but not interleukin 6, in blood samples collected within 14 days vs >14 days after performing severe acute respiratory syndrome coronavirus 2 test. Pregnant women with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 infection also had reduced antispike receptor-binding domain immunoglobulin G titers and were less likely to have detectable neutralizing antibody than nonpregnant women. Although severe acute respiratory syndrome coronavirus 2 infection did not disrupt neonatal Fc receptor expression in the placenta, maternal transfer of severe acute respiratory syndrome coronavirus 2 neutralizing antibody was inhibited by infection during pregnancy. CONCLUSION: Severe acute respiratory syndrome coronavirus 2 infection during pregnancy was characterized by placental inflammation and reduced antiviral antibody responses, which may impact the efficacy of coronavirus disease 2019 treatment in pregnancy. In addition, the long-term implications of placental inflammation for neonatal health require greater consideration.


Asunto(s)
Anticuerpos Antivirales/sangre , COVID-19/inmunología , Inflamación/virología , Interleucina-1beta/genética , Complicaciones del Embarazo/virología , SARS-CoV-2/inmunología , Adulto , Anticuerpos Antivirales/inmunología , Proteínas de Arabidopsis/sangre , COVID-19/complicaciones , Femenino , Sangre Fetal/química , Expresión Génica , Humanos , Inmunoglobulina G/sangre , Interleucina-6/genética , Proteínas de la Membrana/sangre , Enfermedades Placentarias/virología , Embarazo , Complicaciones del Embarazo/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología
17.
Biofactors ; 46(6): 927-933, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-966303

RESUMEN

Recent articles report elevated markers of coagulation, endothelial injury, and microthromboses in lungs from deceased COVID-19 patients. However, there has been no discussion of what may induce intravascular coagulation. Platelets are critical in the formation of thrombi and their most potent trigger is platelet activating factor (PAF), first characterized by Demopoulos and colleagues in 1979. PAF is produced by cells involved in host defense and its biological actions bear similarities with COVID-19 disease manifestations. PAF can also stimulate perivascular mast cell activation, leading to inflammation implicated in severe acute respiratory syndrome (SARS). Mast cells are plentiful in the lungs and are a rich source of PAF and of inflammatory cytokines, such as IL-1ß and IL-6, which may contribute to COVID-19 and especially SARS. The histamine-1 receptor antagonist rupatadine was developed to have anti-PAF activity, and also inhibits activation of human mast cells in response to PAF. Rupatadine could be repurposed for COVID-19 prophylaxis alone or together with other PAF-inhibitors of natural origin such as the flavonoids quercetin and luteolin, which have antiviral, anti-inflammatory, and anti-PAF actions.


Asunto(s)
COVID-19/prevención & control , Ciproheptadina/análogos & derivados , Coagulación Intravascular Diseminada/prevención & control , Factor de Activación Plaquetaria/antagonistas & inhibidores , Embolia Pulmonar/prevención & control , SARS-CoV-2/patogenicidad , Síndrome Respiratorio Agudo Grave/prevención & control , Antivirales/uso terapéutico , Plaquetas/efectos de los fármacos , Plaquetas/patología , Plaquetas/virología , COVID-19/sangre , COVID-19/patología , COVID-19/virología , Ciproheptadina/uso terapéutico , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/patología , Coagulación Intravascular Diseminada/virología , Regulación de la Expresión Génica , Humanos , Inflamación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Luteolina/uso terapéutico , Mastocitos/efectos de los fármacos , Mastocitos/patología , Mastocitos/virología , Factor de Activación Plaquetaria/genética , Factor de Activación Plaquetaria/metabolismo , Embolia Pulmonar/sangre , Embolia Pulmonar/patología , Embolia Pulmonar/virología , Quercetina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Síndrome Respiratorio Agudo Grave/sangre , Síndrome Respiratorio Agudo Grave/patología , Síndrome Respiratorio Agudo Grave/virología
18.
Signal Transduct Target Ther ; 5(1): 235, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: covidwho-841900

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can lead to respiratory illness and multi-organ failure in critically ill patients. Although the virus-induced lung damage and inflammatory cytokine storm are believed to be directly associated with coronavirus disease 2019 (COVID-19) clinical manifestations, the underlying mechanisms of virus-triggered inflammatory responses are currently unknown. Here we report that SARS-CoV-2 infection activates caspase-8 to trigger cell apoptosis and inflammatory cytokine processing in the lung epithelial cells. The processed inflammatory cytokines are released through the virus-induced necroptosis pathway. Virus-induced apoptosis, necroptosis, and inflammation activation were also observed in the lung sections of SARS-CoV-2-infected HFH4-hACE2 transgenic mouse model, a valid model for studying SARS-CoV-2 pathogenesis. Furthermore, analysis of the postmortem lung sections of fatal COVID-19 patients revealed not only apoptosis and necroptosis but also massive inflammatory cell infiltration, necrotic cell debris, and pulmonary interstitial fibrosis, typical of immune pathogenesis in the lung. The SARS-CoV-2 infection triggered a dual mode of cell death pathways and caspase-8-dependent inflammatory responses may lead to the lung damage in the COVID-19 patients. These discoveries might assist the development of therapeutic strategies to treat COVID-19.


Asunto(s)
Apoptosis/inmunología , Betacoronavirus/patogenicidad , Caspasa 8/inmunología , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Necroptosis/inmunología , Neumonía Viral/inmunología , Fibrosis Pulmonar/inmunología , Animales , COVID-19 , Caspasa 8/genética , Línea Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/patología , Síndrome de Liberación de Citoquinas/virología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-7/genética , Interleucina-7/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Transgénicos , Pandemias , Neumonía Viral/genética , Neumonía Viral/patología , Neumonía Viral/virología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/virología , SARS-CoV-2 , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
19.
Signal Transduct Target Ther ; 5(1): 186, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: covidwho-744366

RESUMEN

Sterol regulatory element binding protein-2 (SREBP-2) is activated by cytokines or pathogen, such as virus or bacteria, but its association with diminished cholesterol levels in COVID-19 patients is unknown. Here, we evaluated SREBP-2 activation in peripheral blood mononuclear cells of COVID-19 patients and verified the function of SREBP-2 in COVID-19. Intriguingly, we report the first observation of SREBP-2 C-terminal fragment in COVID-19 patients' blood and propose SREBP-2 C-terminal fragment as an indicator for determining severity. We confirmed that SREBP-2-induced cholesterol biosynthesis was suppressed by Sestrin-1 and PCSK9 expression, while the SREBP-2-induced inflammatory responses was upregulated in COVID-19 ICU patients. Using an infectious disease mouse model, inhibitors of SREBP-2 and NF-κB suppressed cytokine storms caused by viral infection and prevented pulmonary damages. These results collectively suggest that SREBP-2 can serve as an indicator for severity diagnosis and therapeutic target for preventing cytokine storm and lung damage in severe COVID-19 patients.


Asunto(s)
Betacoronavirus/patogenicidad , Colesterol/biosíntesis , Infecciones por Coronavirus/genética , Síndrome de Liberación de Citoquinas/genética , Interacciones Huésped-Patógeno/genética , Leucocitos Mononucleares/inmunología , Neumonía Viral/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Betacoronavirus/inmunología , COVID-19 , Estudios de Casos y Controles , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/mortalidad , Síndrome de Liberación de Citoquinas/virología , Regulación de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Unidades de Cuidados Intensivos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , FN-kappa B/genética , FN-kappa B/inmunología , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Neumonía Viral/virología , Cultivo Primario de Células , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/inmunología , SARS-CoV-2 , Transducción de Señal , Proteína 2 de Unión a Elementos Reguladores de Esteroles/inmunología , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA