Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.446
Filtrar
1.
Nat Commun ; 15(1): 4235, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762489

RESUMEN

Inflammation induced by lung infection is a double-edged sword, moderating both anti-viral and immune pathogenesis effects; the mechanism of the latter is not fully understood. Previous studies suggest the vasculature is involved in tissue injury. Here, we report that expression of Sparcl1, a secreted matricellular protein, is upregulated in pulmonary capillary endothelial cells (EC) during influenza-induced lung injury. Endothelial overexpression of SPARCL1 promotes detrimental lung inflammation, with SPARCL1 inducing 'M1-like' macrophages and related pro-inflammatory cytokines, while SPARCL1 deletion alleviates these effects. Mechanistically, SPARCL1 functions through TLR4 on macrophages in vitro, while TLR4 inhibition in vivo ameliorates excessive inflammation caused by endothelial Sparcl1 overexpression. Finally, SPARCL1 expression is increased in lung ECs from COVID-19 patients when compared with healthy donors, while fatal COVID-19 correlates with higher circulating SPARCL1 protein levels in the plasma. Our results thus implicate SPARCL1 as a potential prognosis biomarker for deadly COVID-19 pneumonia and as a therapeutic target for taming hyperinflammation in pneumonia.


Asunto(s)
COVID-19 , Células Endoteliales , Pulmón , Activación de Macrófagos , SARS-CoV-2 , Animales , Humanos , COVID-19/inmunología , COVID-19/virología , COVID-19/metabolismo , COVID-19/patología , Ratones , Células Endoteliales/metabolismo , Células Endoteliales/virología , Células Endoteliales/inmunología , SARS-CoV-2/fisiología , Pulmón/virología , Pulmón/patología , Pulmón/inmunología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Ratones Endogámicos C57BL , Neumonía Viral/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , Neumonía Viral/metabolismo , Masculino , Macrófagos/metabolismo , Macrófagos/inmunología , Femenino , Ratones Noqueados , Proteínas de la Matriz Extracelular
2.
J Virol ; 98(5): e0176223, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38563762

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and is responsible for the largest human pandemic in 100 years. Thirty-four vaccines are currently approved for use worldwide, and approximately 67% of the world population has received a complete primary series of one, yet countries are dealing with new waves of infections, variant viruses continue to emerge, and breakthrough infections are frequent secondary to waning immunity. Here, we evaluate a measles virus (MV)-vectored vaccine expressing a stabilized prefusion SARS-CoV-2 spike (S) protein (MV-ATU3-S2PΔF2A; V591) with demonstrated immunogenicity in mouse models (see companion article [J. Brunet, Z. Choucha, M. Gransagne, H. Tabbal, M.-W. Ku et al., J Virol 98:e01693-23, 2024, https://doi.org/10.1128/jvi.01693-23]) in an established African green monkey model of disease. Animals were vaccinated with V591 or the control vaccine (an equivalent MV-vectored vaccine with an irrelevant antigen) intramuscularly using a prime/boost schedule, followed by challenge with an early pandemic isolate of SARS-CoV-2 at 56 days post-vaccination. Pre-challenge, only V591-vaccinated animals developed S-specific antibodies that had virus-neutralizing activity as well as S-specific T cells. Following the challenge, V591-vaccinated animals had lower infectious virus and viral (v) RNA loads in mucosal secretions and stopped shedding virus in these secretions earlier. vRNA loads were lower in these animals in respiratory and gastrointestinal tract tissues at necropsy. This correlated with a lower disease burden in the lungs as quantified by PET/CT at early and late time points post-challenge and by pathological analysis at necropsy.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the largest human pandemic in 100 years. Even though vaccines are currently available, countries are dealing with new waves of infections, variant viruses continue to emerge, breakthrough infections are frequent, and vaccine hesitancy persists. This study uses a safe and effective measles vaccine as a platform for vaccination against SARS-CoV-2. The candidate vaccine was used to vaccinate African green monkeys (AGMs). All vaccinated AGMs developed robust antigen-specific immune responses. After challenge, these AGMs produced less virus in mucosal secretions, for a shorter period, and had a reduced disease burden in the lungs compared to control animals. At necropsy, lower levels of viral RNA were detected in tissue samples from vaccinated animals, and the lungs of these animals lacked the histologic hallmarks of SARS-CoV-2 disease observed exclusively in the control AGMs.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Virus del Sarampión , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Animales , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Chlorocebus aethiops , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/inmunología , COVID-19/virología , Virus del Sarampión/inmunología , Virus del Sarampión/genética , Vacunas contra la COVID-19/inmunología , Humanos , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Vectores Genéticos , Células Vero , Pandemias/prevención & control , Femenino , Betacoronavirus/inmunología , Betacoronavirus/genética , Neumonía Viral/prevención & control , Neumonía Viral/virología , Neumonía Viral/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/veterinaria , Vacunas Virales/inmunología , Vacunas Virales/genética , Vacunas Virales/administración & dosificación , Modelos Animales de Enfermedad
3.
Int J Biol Macromol ; 267(Pt 1): 131427, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583833

RESUMEN

Due to the health emergency created by SARS-CoV-2, the virus that causes the COVID-19 disease, the rapid implementation of a new vaccine technology was necessary. mRNA vaccines, being one of the cutting-edge new technologies, attracted significant interest and offered a lot of hope. The potential of these vaccines in preventing admission to hospitals and serious illness in people with comorbidities has recently been called into question due to the vaccines' rapidly waning immunity. Mounting evidence indicates that these vaccines, like many others, do not generate sterilizing immunity, leaving people vulnerable to recurrent infections. Additionally, it has been discovered that the mRNA vaccines inhibit essential immunological pathways, thus impairing early interferon signaling. Within the framework of COVID-19 vaccination, this inhibition ensures an appropriate spike protein synthesis and a reduced immune activation. Evidence is provided that adding 100 % of N1-methyl-pseudouridine (m1Ψ) to the mRNA vaccine in a melanoma model stimulated cancer growth and metastasis, while non-modified mRNA vaccines induced opposite results, thus suggesting that COVID-19 mRNA vaccines could aid cancer development. Based on this compelling evidence, we suggest that future clinical trials for cancers or infectious diseases should not use mRNA vaccines with a 100 % m1Ψ modification, but rather ones with the lower percentage of m1Ψ modification to avoid immune suppression.


Asunto(s)
COVID-19 , Neoplasias , Seudouridina , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2/inmunología , Neoplasias/inmunología , Seudouridina/metabolismo , Vacunas contra la COVID-19/inmunología , Animales , Vacunas de ARNm , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/virología , Neumonía Viral/prevención & control , Betacoronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología
4.
Obstet Gynecol ; 143(6): e149-e152, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38574363

RESUMEN

BACKGROUND: Since the onset of the coronavirus disease (COVID-19) pandemic, a variety of long-COVID-19 symptoms and autoimmune complications have been recognized. CASES: We report three cases of autoimmune premature poor ovarian response in patients aged 30-37 years after mild to asymptomatic COVID-19 before vaccination, with nucleotide antibody confirmation. Two patients failed to respond to maximum-dose gonadotropins for more than 4 weeks, despite a recent history of response before having COVID-19. After a month of prednisone 30 mg, these two patients had normal follicle-stimulating hormone (FSH) levels, high oocyte yield, and blastocyst formation in successful in vitro fertilization cycles. All three patients have above-average anti-müllerian hormone levels that persisted throughout their clinical ovarian insufficiency. Two patients had elevated FSH levels, perhaps resulting from FSH receptor blockade. One patient, with a history of high response to gonadotropins 75 international units per day and below-normal FSH levels, had no ovarian response to more than a month of gonadotropins (525 international units daily), suggesting autoimmune block of the FSH glycoprotein and possible FSH receptor blockade. CONCLUSION: Auto-antibody production in response to COVID-19 before vaccination may be a rare cause of autoimmune poor ovarian response. Although vaccination is likely protective, further study will be required to evaluate the effect of vaccination and duration of autoimmune FSH or FSH receptor blockade.


Asunto(s)
COVID-19 , Insuficiencia Ovárica Primaria , Receptores de HFE , SARS-CoV-2 , Humanos , Femenino , COVID-19/inmunología , COVID-19/complicaciones , Insuficiencia Ovárica Primaria/inmunología , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Adulto , SARS-CoV-2/inmunología , Pandemias , Hormona Folículo Estimulante/sangre , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/inmunología , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Betacoronavirus
5.
Mol Ther ; 32(5): 1510-1525, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38454605

RESUMEN

The acute respiratory virus infection can induce uncontrolled inflammatory responses, such as cytokine storm and viral pneumonia, which are the major causes of death in clinical cases. Cyclophilin A (CypA) is mainly distributed in the cytoplasm of resting cells and released into the extracellular space in response to inflammatory stimuli. Extracellular CypA (eCypA) is upregulated and promotes inflammatory response in severe COVID-19 patients. However, how eCypA promotes virus-induced inflammatory response remains elusive. Here, we observe that eCypA is induced by influenza A and B viruses and SARS-CoV-2 in cells, mice, or patients. Anti-CypA mAb reduces pro-inflammatory cytokines production, leukocytes infiltration, and lung injury in virus-infected mice. Mechanistically, eCypA binding to integrin ß2 triggers integrin activation, thereby facilitating leukocyte trafficking and cytokines production via the focal adhesion kinase (FAK)/GTPase and FAK/ERK/P65 pathways, respectively. These functions are suppressed by the anti-CypA mAb that specifically blocks eCypA-integrin ß2 interaction. Overall, our findings reveal that eCypA-integrin ß2 signaling mediates virus-induced inflammatory response, indicating that eCypA is a potential target for antibody therapy against viral pneumonia.


Asunto(s)
COVID-19 , Ciclofilina A , Ciclofilina A/metabolismo , Animales , Humanos , Ratones , COVID-19/metabolismo , COVID-19/virología , COVID-19/inmunología , Antígenos CD18/metabolismo , SARS-CoV-2 , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Neumonía Viral/metabolismo , Neumonía Viral/inmunología , Citocinas/metabolismo , Anticuerpos Monoclonales/farmacología , Transducción de Señal , Virus de la Influenza A , Modelos Animales de Enfermedad
6.
J Innate Immun ; 16(1): 133-142, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38325356

RESUMEN

INTRODUCTION: Coronavirus disease 2019 caused by coronavirus-2 (SARS-CoV-2) has emerged as an aggressive viral pandemic. Health care providers confront a challenging task for rapid development of effective strategies to combat this and its long-term after effects. Virus entry into host cells involves interaction between receptor-binding domain (RBD) of spike (S) protein S1 subunit with angiotensin converting enzyme present on host cells. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a moonlighting enzyme involved in cellular glycolytic energy metabolism and micronutrient homeostasis. It is deployed in various cellular compartments and the extra cellular milieu. Though it is known to moonlight as a component of mammalian innate immune defense machinery, till date its role in viral restriction remains unknown. METHOD: Recombinant S protein, the RBD, and human GAPDH protein were used for solid phase binding assays and biolayer interferometry. Pseudovirus particles expressing four different strain variants of S protein all harboring ZsGreen gene as marker of infection were used for flow cytometry-based infectivity assays. RESULTS: Pseudovirus entry into target cells in culture was significantly inhibited by addition of human GAPDH into the extracellular medium. Binding assays demonstrated that human GAPDH binds to S protein and RBD of SARS-CoV-2 with nanomolar affinity. CONCLUSIONS: Our investigations suggest that this interaction of GAPDH interferes in the viral docking with hACE2 receptors, thereby affecting viral ingress into mammalian cells.


Asunto(s)
COVID-19 , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante) , Unión Proteica , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus , Humanos , Glicoproteína de la Espiga del Coronavirus/metabolismo , SARS-CoV-2/fisiología , COVID-19/virología , Células HEK293 , Betacoronavirus/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Neumonía Viral/virología , Neumonía Viral/inmunología , Pandemias , Infecciones por Coronavirus/virología , Enzima Convertidora de Angiotensina 2/metabolismo
7.
Nature ; 623(7988): 803-813, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938781

RESUMEN

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Asunto(s)
Autoanticuerpos , Predisposición Genética a la Enfermedad , Interferón Tipo I , FN-kappa B , Humanos , Autoanticuerpos/inmunología , COVID-19/genética , COVID-19/inmunología , Mutación con Ganancia de Función , Heterocigoto , Proteínas I-kappa B/deficiencia , Proteínas I-kappa B/genética , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Mutación con Pérdida de Función , FN-kappa B/deficiencia , FN-kappa B/genética , Subunidad p52 de NF-kappa B/deficiencia , Subunidad p52 de NF-kappa B/genética , Neumonía Viral/genética , Neumonía Viral/inmunología , Timo/anomalías , Timo/inmunología , Timo/patología , Células Epiteliales Tiroideas/metabolismo , Células Epiteliales Tiroideas/patología , Proteína AIRE , Quinasa de Factor Nuclear kappa B
8.
Proc Natl Acad Sci U S A ; 119(37): e2121385119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067309

RESUMEN

Interferon (IFN) regulatory factor 3 (IRF3) is a transcription factor activated by phosphorylation in the cytoplasm of a virus-infected cell; by translocating to the nucleus, it induces transcription of IFN-ß and other antiviral genes. We have previously reported IRF3 can also be activated, as a proapoptotic factor, by its linear polyubiquitination mediated by the RIG-I pathway. Both transcriptional and apoptotic functions of IRF3 contribute to its antiviral effect. Here, we report a nontranscriptional function of IRF3, namely, the repression of IRF3-mediated NF-κB activity (RIKA), which attenuated viral activation of NF-κB and the resultant inflammatory gene induction. In Irf3-/- mice, consequently, Sendai virus infection caused enhanced inflammation in the lungs. Mechanistically, RIKA was mediated by the direct binding of IRF3 to the p65 subunit of NF-κB in the cytoplasm, which prevented its nuclear import. A mutant IRF3 defective in both the transcriptional and the apoptotic activities was active in RIKA and inhibited virus replication. Our results demonstrated IRF3 deployed a three-pronged attack on virus replication and the accompanying inflammation.


Asunto(s)
Inmunidad Innata , Factor 3 Regulador del Interferón , FN-kappa B , Neumonía Viral , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Expresión Génica , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Ratones , FN-kappa B/metabolismo , Neumonía Viral/genética , Neumonía Viral/inmunología , Virus Sendai
9.
BMC Infect Dis ; 22(1): 343, 2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35382755

RESUMEN

BACKGROUND: Pneumonia is a common complication of influenza and closely related to mortality in influenza patients. The present study examines cytokines as predictors of the prognosis of influenza-associated pneumonia. METHODS: This study included 101 inpatients with influenza (64 pneumonia and 37 non-pneumonia patients). 48 cytokines were detected in the serum samples of the patients and the clinical characteristics were analyzed. The correlation between them was analyzed to identify predictive biomarkers for the prognosis of influenza-associated pneumonia. RESULTS: Seventeen patients had poor prognosis and developed pneumonia. Among patients with influenza-associated pneumonia, the levels of 8 cytokines were significantly higher in those who had a poor prognosis: interleukin-6 (IL-6), interferon-γ (IFN-γ), granulocyte colony-stimulating factor (G-CSF), monocyte colony-stimulating factor (M-CSF), monocyte chemoattractant protein-1 (MCP-1), monocyte chemoattractant protein-3, Interleukin-2 receptor subunit alpha and Hepatocyte growth factor. Correlation analysis showed that the IL-6, G-CSF, M-CSF, IFN-γ, and MCP-1 levels had positive correlations with the severity of pneumonia. IL-6 and G-CSF showed a strong and positive correlation with poor prognosis in influenza-associated pneumonia patients. The combined effect of the two cytokines resulted in the largest area (0.926) under the receiver-operating characteristic curve. CONCLUSION: The results indicate that the probability of poor prognosis in influenza patients with pneumonia is significantly increased. IL-6, G-CSF, M-CSF, IFN-γ, and MCP-1 levels had a positive correlation with the severity of pneumonia. Importantly, IL-6 and G-CSF were identified as significant predictors of the severity of influenza-associated pneumonia.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Gripe Humana , Interleucina-6 , Neumonía Viral , Citocinas/sangre , Factor Estimulante de Colonias de Granulocitos/sangre , Humanos , Gripe Humana/complicaciones , Gripe Humana/inmunología , Interleucina-6/sangre , Neumonía Viral/diagnóstico , Neumonía Viral/inmunología , Pronóstico
10.
J Immunol Res ; 2021: 2958394, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926703

RESUMEN

Adenovirus (Ad) is a major causal agent of acute respiratory infections. However, they are a powerful delivery system for gene therapy and vaccines. Some Ad serotypes antagonize the immune system leading to meningitis, conjunctivitis, gastroenteritis, and/or acute hemorrhagic cystitis. Studies have shown that the release of small, membrane-derived extracellular vesicles (EVs) may offer a mechanism by which viruses can enter cells via receptor-independent entry and how they influence disease pathogenesis and/or host protection considering their existence in almost all bodily fluids. We proposed that Ad3 could alter EV biogenesis, composition, and trafficking and may stimulate various immune responses in vitro. In the present study, we evaluated the impact of in vitro infection with Ad3 vector on EV biogenesis and composition in the human adenocarcinoma lung epithelial cell line A549. Cells were infected in an exosome-free media at different multiplicity of infections (MOIs) and time points. The cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and fluorometric calcein-AM. EVs were isolated via ultracentrifugation. Isolated EV proteins were quantified and evaluated via nanoparticle tracking, transmission electron microscopy, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and immunoblotting assays. The cell viability significantly decreased with an increase in MOI and incubation time. A significant increase in particle mean sizes, concentrations, and total EV protein content was detected at higher MOIs when compared to uninfected cells (control group). A549 cell-derived EVs revealed the presence of TSG101, tetraspanins CD9 and CD63, and heat shock proteins 70 and 100 with significantly elevated levels of Rab5, 7, and 35 at higher MOIs (300, 750, and 1500) when compared to the controls. Our findings suggested Ad3 could modulate EV biogenesis, composition, and trafficking which could impact infection pathogenesis and disease progression. This study might suggest EVs could be diagnostic and therapeutic advancement to Ad infections and other related viral infections. However, further investigation is warranted to explore the underlying mechanism(s).


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Vesículas Extracelulares/inmunología , Pulmón/patología , Neumonía Viral/inmunología , Células A549 , Infecciones por Adenovirus Humanos/patología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/genética , Supervivencia Celular/inmunología , Vesículas Extracelulares/metabolismo , Humanos , Pulmón/citología , Pulmón/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , Serogrupo
11.
Viruses ; 13(12)2021 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-34960654

RESUMEN

The host immunity of patients with adenovirus pneumonia in different severity of illness is unclear. This study compared the routine laboratory tests and the host immunity of human adenovirus (HAdV) patients with different severity of illness. A co-cultured cell model in vitro was established to verify the T cell response in vitro. Among 140 patients with confirmed HAdV of varying severity, the number of lymphocytes in the severe patients was significantly reduced to 1.91 × 109/L compared with the healthy control (3.92 × 109/L) and the mild patients (4.27 × 109/L). The levels of IL-6, IL-10, and IFN-γ in patients with adenovirus pneumonia were significantly elevated with the severity of the disease. Compared with the healthy control (20.82%) and the stable patients (33.96%), the percentage of CD8+ T cells that produced IFN-γ increased to 56.27% in the progressing patients. Adenovirus infection increased the percentage of CD8+ T and CD4+ T cells that produce IFN-γ in the co-culture system. The hyperfunction of IFN-γ+ CD8+ T cells might be related to the severity of adenovirus infection. The in vitro co-culture cell model could also provide a usable cellular model for subsequent experiments.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/fisiología , Linfocitos T CD8-positivos/microbiología , Interferón gamma/inmunología , Neumonía Viral/inmunología , Infecciones por Adenovirus Humanos/genética , Infecciones por Adenovirus Humanos/patología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/genética , Niño , Preescolar , Femenino , Humanos , Lactante , Interferón gamma/genética , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Recuento de Linfocitos , Masculino , Gravedad del Paciente , Neumonía Viral/genética , Neumonía Viral/patología , Neumonía Viral/virología
12.
mBio ; 12(6): e0274921, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34749524

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a historic pandemic of respiratory disease (coronavirus disease 2019 [COVID-19]), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2 signaling restricts the viral burden in the lung. We find that a recently developed mouse-adapted SARS-CoV-2 (MA-SARS-CoV-2) strain as well as the emerging B.1.351 variant trigger an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using intravital antibody labeling, we demonstrate that MA-SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Single-cell RNA sequencing (scRNA-Seq) analysis of lung homogenates identified a hyperinflammatory monocyte profile. We utilize this model to demonstrate that mechanistically, CCR2 signaling promotes the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified a potential CCR2-monocyte axis that is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection. IMPORTANCE SARS-CoV-2 has caused a historic pandemic of respiratory disease (COVID-19), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2-dependent infiltration of monocytes restricts the viral burden in the lung. We find that SARS-CoV-2 triggers an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using RNA sequencing and flow cytometry approaches, we demonstrate that SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Mechanistically, CCR2 signaling promoted the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified that the CCR2 pathway is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection.


Asunto(s)
Pulmón/inmunología , Neumonía Viral/prevención & control , Receptores CCR2/inmunología , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Animales , COVID-19 , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunidad Innata , Inflamación , Pulmón/citología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Neumonía Viral/inmunología , Neumonía Viral/virología , Receptores CCR2/genética , Receptores CCR2/metabolismo , SARS-CoV-2/genética , Carga Viral , Replicación Viral/inmunología
13.
Expert Rev Gastroenterol Hepatol ; 15(11): 1281-1294, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34654347

RESUMEN

INTRODUCTION: Human gut microbiota plays a crucial role in providing protective responses against pathogens, particularly by regulating immune system homeostasis. There is a reciprocal interaction between the gut and lung microbiota, called the gut-lung axis (GLA). Any alteration in the gut microbiota or their metabolites can cause immune dysregulation, which can impair the antiviral activity of the immune system against respiratory viruses such as severe acute respiratory syndrome coronavirus (SARS-CoV) and SARS-CoV-2. AREAS COVERED: This narrative review mainly outlines emerging data on the mechanisms underlying the interactions between the immune system and intestinal microbial dysbiosis, which is caused by an imbalance in the levels of essential metabolites. The authors will also discuss the role of probiotics in restoring the balance of the gut microbiota and modulation of cytokine storm. EXPERT OPINION: Microbiota-derived signals regulate the immune system and protect different tissues during severe viral respiratory infections. The GLA's equilibration could help manage the mortality and morbidity rates associated with SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/inmunología , Neumonía Viral/inmunología , Humanos , SARS-CoV-2
14.
Bioengineered ; 12(2): 10335-10344, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34699306

RESUMEN

It focused on the antiviral immune regulation of biofilm-localized protein kinase Dbf2p-related kinase 1 (NDR1) in viral pneumonia. Mouse alveolar monocyte RAW264.7 was used as blank control, and viral pneumonia cell model was prepared by infecting cells with respiratory syncytial virus (RSV). NDR1 overexpression vector and siRNA interference sequences were synthesized, and overexpression/silence NDR1 cell model was fabricated. About 50 ng/mL interleukin 17 (IL-17) was given to stimulate. Enzyme-linked immunosorbent assay (ELISA), quantitative reverse transcription PCR (RT-qRCR), and Western blot were performed to detect cytokines and chemokines, mRNA of inflammatory factors, and signal molecule protein expression. Notably, RSV infection increased RSV-F mRNA in RAW264.7 cells and reduced NDR1 mRNA and protein. Secretion levels of IL-6, interferon ß (IFN-ß), chemokine (C-X-C motif) ligand 2 (CXCL2), and chemokine (C-C motif) ligand 2 (CCL20) increased in the model group versus blank control (P< 0.05). IL-6, IFN-ß, tumor necrosis factor α (TNF-α), and toll-like receptor 3 (TLR3) mRNA were up-regulated (P < 0.05). Extracellular signal-regulated kinase (ERK1/2), p38 protein phosphorylation, human recombinant 1 (TBK1), and nuclear factor kappa-B (NF-κB) protein levels increased (P < 0.05). After overexpression of NDR1, the secretion levels of cytokines and chemokines, inflammatory factors mRNA, and signal molecule protein increased significantly. After NDR1 was silenced, cytokines and chemokines, inflammatory factors mRNA, and signal molecule protein were not significantly different versus blank control group (P > 0.05). In short, NDR1 regulated innate immune response to viral pneumonia induced by IL-17, which can be used as a new target for the treatment of IL-17-induced inflammatory response and autoimmune diseases.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Inmunidad Innata , Interleucina-17/metabolismo , Neumonía Viral/enzimología , Neumonía Viral/inmunología , Animales , Proliferación Celular , Quimiocinas/metabolismo , Inflamación/patología , Ratones , Proteínas Serina-Treonina Quinasas , Células RAW 264.7 , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Transducción de Señal
15.
Eur J Immunol ; 51(12): 3194-3201, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34564853

RESUMEN

Accelerate lung repair in SARS-CoV-2 pneumonia is essential for pandemic handling. Innate lymphoid cells (ILCs) are likely players, given their role in mucosal protection and tissue homeostasis. We studied ILC subpopulations at two time points in a cohort of patients admitted in the hospital due to SARS-CoV-2 infection. COVID-19 patients with moderate/severe respiratory failure featured profound depletion of circulating ILCs at hospital admission, in agreement with overall lymphocyte depletion. However, ILCs recovered in direct correlation with lung function improvement as measured by oxygenation index and in negative association with inflammatory and lung/endothelial damage markers like RAGE. While both ILC1 and ILC2 expanded, ILC2 showed the most striking phenotype changes, with CCR10 upregulation in strong correlation with these parameters. Overall, CCR10+ ILC2 emerge as relevant contributors to SARS-CoV-2 pneumonia recovery.


Asunto(s)
Biomarcadores/metabolismo , COVID-19/inmunología , Pulmón/patología , Linfocitos/inmunología , Neumonía Viral/inmunología , Receptores CCR10/metabolismo , SARS-CoV-2/fisiología , Adulto , Anciano , Antígenos de Neoplasias/metabolismo , Proliferación Celular , Citocinas/metabolismo , Femenino , Humanos , Inmunidad Innata , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Recuperación de la Función , Células Th2/inmunología , Regulación hacia Arriba
16.
Front Immunol ; 12: 704427, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489955

RESUMEN

Viral pneumonias are a major cause of morbidity and mortality, owing in part to dysregulated excessive lung inflammation, and therapies to modulate host responses to viral lung injury are urgently needed. Protectin conjugates in tissue regeneration 1 (PCTR1) and protectin D1 (PD1) are specialized pro-resolving mediators (SPMs) whose roles in viral pneumonia are of interest. In a mouse model of Respiratory Syncytial Virus (RSV) pneumonia, intranasal PCTR1 and PD1 each decreased RSV genomic viral load in lung tissue when given after RSV infection. Concurrent with enhanced viral clearance, PCTR1 administration post-infection, decreased eosinophils, neutrophils, and NK cells, including NKG2D+ activated NK cells, in the lung. Intranasal PD1 administration post-infection decreased lung eosinophils and Il-13 expression. PCTR1 increased lung expression of cathelicidin anti-microbial peptide and decreased interferon-gamma production by lung CD4+ T cells. PCTR1 and PD1 each increased interferon-lambda expression in human bronchial epithelial cells in vitro and attenuated RSV-induced suppression of interferon-lambda in mouse lung in vivo. Liquid chromatography coupled with tandem mass spectrometry of RSV-infected and untreated mouse lungs demonstrated endogenous PCTR1 and PD1 that decreased early in the time course while cysteinyl-leukotrienes (cys-LTs) increased during early infection. As RSV infection resolved, PCTR1 and PD1 increased and cys-LTs decreased to pre-infection levels. Together, these results indicate that PCTR1 and PD1 are each regulated during RSV pneumonia, with overlapping and distinct mechanisms for PCTR1 and PD1 during the resolution of viral infection and its associated inflammation.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Pulmón/inmunología , Neumonía Viral , Infecciones por Virus Sincitial Respiratorio , Virus Sincitiales Respiratorios/inmunología , Administración Intranasal , Animales , Inflamación/inmunología , Inflamación/prevención & control , Pulmón/virología , Masculino , Ratones , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Carga Viral/efectos de los fármacos
17.
Front Immunol ; 12: 727941, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34504501

RESUMEN

Influenza A virus infection is usually associated with acute lung injury, which is typically characterized by tracheal mucosal barrier damage and an interleukin 17A (IL-17A)-mediated inflammatory response in lung tissues. Although targeting IL-17A has been proven to be beneficial for attenuating inflammation around lung cells, it still has a limited effect on pulmonary tissue recovery after influenza A virus infection. In this research, interleukin 22 (IL-22), a cytokine involved in the repair of the pulmonary mucosal barrier, was fused to the C-terminus of the anti-IL-17A antibody vunakizumab to endow the antibody with a tissue recovery function. The vunakizumab-IL22 (vmab-IL-22) fusion protein exhibits favorable stability and retains the biological activities of both the anti-IL-17A antibody and IL-22 in vitro. Mice infected with lethal H1N1 influenza A virus and treated with vmab-mIL22 showed attenuation of lung index scores and edema when compared to those of mice treated with saline or vmab or mIL22 alone. Our results also illustrate that vmab-mIL22 triggers the upregulation of MUC2 and ZO1, as well as the modulation of cytokines such as IL-1ß, HMGB1 and IL-10, indicating the recovery of pulmonary goblet cells and the suppression of excessive inflammation in mice after influenza A virus infection. Moreover, transcriptome profiling analysis suggest the downregulation of fibrosis-related genes and signaling pathways, including genes related to focal adhesion, the inflammatory response pathway, the TGF-ß signaling pathway and lung fibrosis upon vmab-mIL22 treatment, which indicates that the probable mechanism of vmab-mIL22 in ameliorating H1N1 influenza A-induced lung injury. Our results reveal that the bifunctional fusion protein vmab-mIL22 can trigger potent therapeutic effects in H1N1-infected mice by enhancing lung tissue recovery and inhibiting pulmonary inflammation, which highlights a potential approach for treating influenza A virus infection by targeting IL-17A and IL-22 simultaneously.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Anticuerpos Monoclonales/inmunología , Antivirales/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A , Interleucina-17/inmunología , Interleucinas/inmunología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Anticuerpos Monoclonales/genética , Antivirales/farmacología , Células CHO , Cricetulus , Células HT29 , Células Hep G2 , Humanos , Interleucinas/genética , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/genética , Neumonía Viral/inmunología , Neumonía Viral/patología , Proteínas Recombinantes de Fusión/farmacología , Transcriptoma/efectos de los fármacos , Interleucina-22
18.
Cell Rep ; 37(1): 109798, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34587481

RESUMEN

Despite the worldwide effect of the coronavirus disease 2019 (COVID-19) pandemic, the underlying mechanisms of fatal viral pneumonia remain elusive. Here, we show that critical COVID-19 is associated with enhanced eosinophil-mediated inflammation when compared to non-critical cases. In addition, we confirm increased T helper (Th)2-biased adaptive immune responses, accompanying overt complement activation, in the critical group. Moreover, enhanced antibody responses and complement activation are associated with disease pathogenesis as evidenced by formation of immune complexes and membrane attack complexes in airways and vasculature of lung biopsies from six fatal cases, as well as by enhanced hallmark gene set signatures of Fcγ receptor (FcγR) signaling and complement activation in myeloid cells of respiratory specimens from critical COVID-19 patients. These results suggest that SARS-CoV-2 infection may drive specific innate immune responses, including eosinophil-mediated inflammation, and subsequent pulmonary pathogenesis via enhanced Th2-biased immune responses, which might be crucial drivers of critical disease in COVID-19 patients.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Proteínas del Sistema Complemento/inmunología , Eosinófilos/inmunología , Inflamación/inmunología , Neumonía Viral/inmunología , SARS-CoV-2/inmunología , Inmunidad Adaptativa , Adulto , Anciano , Anciano de 80 o más Años , Complejo Antígeno-Anticuerpo/metabolismo , COVID-19/metabolismo , COVID-19/virología , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Eosinófilos/virología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Masculino , Persona de Mediana Edad , Neumonía Viral/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Células Th2/inmunología , Carga Viral , Adulto Joven
19.
Biomed Res Int ; 2021: 9987931, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34423043

RESUMEN

OBJECTIVE: Respiratory failure is the leading cause of mortality in COVID-19 patients, characterized by a generalized disbalance of inflammation. The aim of this study was to investigate the relationship between immune-inflammatory index and mortality in PSI IV-V patients with COVID-19. METHODS: We retrospectively reviewed the medical records of COVID-19 patients from Feb. to Apr. 2020 in the Zhongfa Xincheng Branch of Tongji Hospital, Wuhan, China. Patients who presented high severity of COVID-19-related pneumonia were enrolled for further analysis according to the Pneumonia Severity Index (PSI) tool. RESULTS: A total of 101 patients diagnosed with COVID-19 were identified at initial research. The survival analysis revealed that mortality of the PSI IV-V cohort was significantly higher than the PSI I-III group (p = 0.0003). The overall mortality in PSI IV-V patients was 32.1% (9/28). The fatal cases of the PSI IV-V group had a higher level of procalcitonin (p = 0.022) and neutrophil-to-lymphocyte ratio (p = 0.033) compared with the survivors. Procalcitonin was the most sensitive predictor of mortality for the severe COVID-19 population with area under receiver operating characteristic curve of 0.78, higher than the neutrophil-to-lymphocyte ratio (0.75) and total lymphocyte (0.68) and neutrophil (0.67) counts. CONCLUSION: Procalcitonin and neutrophil-to-lymphocyte ratio may potentially be effective predictors for mortality in PSI IV-V patients with COVID-19. Increased procalcitonin and neutrophil-to-lymphocyte ratio were associated with greater risk of mortality.


Asunto(s)
COVID-19/inmunología , COVID-19/fisiopatología , Pandemias , SARS-CoV-2 , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , COVID-19/mortalidad , China/epidemiología , Estudios de Cohortes , Femenino , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neumonía Viral/inmunología , Neumonía Viral/mortalidad , Neumonía Viral/fisiopatología , Polipéptido alfa Relacionado con Calcitonina/sangre , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
20.
J Immunol ; 207(5): 1229-1238, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34348975

RESUMEN

Infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or seasonal influenza may lead to respiratory failure requiring intubation and mechanical ventilation. The pathophysiology of this respiratory failure is attributed to local immune dysregulation, but how the immune response to viral infection in the lower airways of the human lung differs between individuals with respiratory failure and those without is not well understood. We used quantitative multiparameter flow cytometry and multiplex cytokine assays to evaluate matched blood and bronchoalveolar lavage (BAL) samples from control human subjects, subjects with symptomatic seasonal influenza who did not have respiratory failure, and subjects with severe seasonal influenza or SARS-CoV-2 infection with respiratory failure. We find that severe cases are associated with an influx of nonclassical monocytes, activated T cells, and plasmablast B cells into the lower airways. Cytokine concentrations were not elevated in the lower airways of moderate influenza patients compared with controls; however, 28 of 35 measured cytokines were significantly elevated in severe influenza, severe SARS-CoV-2 infection, or both. We noted the largest elevations in IL-6, IP-10, MCP-1, and IL-8. IL-1 family cytokines and RANTES were higher in severe influenza infection than severe SARS-CoV-2 infection. Interestingly, only the concentration of IP-10-correlated between blood and BAL during severe infection. Our results demonstrate inflammatory immune dysregulation in the lower airways during severe viral pneumonia that is distinct from lower airway responses seen in human patients with symptomatic, but not severe, illness and suggest that measurement of blood IP-10 concentration may predict this unique dysregulation.


Asunto(s)
COVID-19/inmunología , Virus de la Influenza A/fisiología , Neumonía Viral/inmunología , Sistema Respiratorio/inmunología , SARS-CoV-2/fisiología , Adulto , Anciano , Proteínas Sanguíneas/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , COVID-19/diagnóstico , Quimiocina CXCL10/metabolismo , Estudios de Cohortes , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Gripe Humana/inmunología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Insuficiencia Respiratoria , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA