Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Front Immunol ; 13: 780839, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-1686482

RESUMEN

Macrophages are essential innate immune cells that contribute to host defense during infection. An important feature of macrophages is their ability to respond to extracellular cues and to adopt different phenotypes and functions in response to these stimuli. The evidence accumulated in the last decade has highlighted the crucial role of metabolic reprogramming during macrophage activation in infectious context. Thus, understanding and manipulation of macrophage immunometabolism during infection could be of interest to develop therapeutic strategies. In this review, we focus on 5 major metabolic pathways including glycolysis, pentose phosphate pathway, fatty acid oxidation and synthesis, tricarboxylic acid cycle and amino acid metabolism and discuss how they sustain and regulate macrophage immune function in response to parasitic, bacterial and viral infections as well as trained immunity. At the end, we assess whether some drugs including those used in clinic and in development can target macrophage immunometabolism for potential therapy during infection with an emphasis on SARS-CoV2 infection.


Asunto(s)
Infecciones/inmunología , Infecciones/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , COVID-19/inmunología , Humanos , Inmunidad Innata/inmunología , SARS-CoV-2
2.
Sci Immunol ; 7(68): eabf2846, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1685480

RESUMEN

Macrophages regulate protective immune responses to infectious microbes, but aberrant macrophage activation frequently drives pathological inflammation. To identify regulators of vigorous macrophage activation, we analyzed RNA-seq data from synovial macrophages and identified SLAMF7 as a receptor associated with a superactivated macrophage state in rheumatoid arthritis. We implicated IFN-γ as a key regulator of SLAMF7 expression and engaging SLAMF7 drove a strong wave of inflammatory cytokine expression. Induction of TNF-α after SLAMF7 engagement amplified inflammation through an autocrine signaling loop. We observed SLAMF7-induced gene programs not only in macrophages from rheumatoid arthritis patients but also in gut macrophages from patients with active Crohn's disease and in lung macrophages from patients with severe COVID-19. This suggests a central role for SLAMF7 in macrophage superactivation with broad implications in human disease pathology.


Asunto(s)
Inflamación/inmunología , Activación de Macrófagos/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Transcriptoma/inmunología , Enfermedad Aguda , Adulto , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , COVID-19/genética , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Enfermedad Crónica , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Activación de Macrófagos/genética , RNA-Seq/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Análisis de la Célula Individual/métodos , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Transcriptoma/genética
3.
Front Immunol ; 12: 828115, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1680008

RESUMEN

Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos/inmunología , Macrófagos/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Animales , Proteínas Portadoras , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Ligandos , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Mecanotransducción Celular , Terapia Molecular Dirigida , Unión Proteica
4.
Front Immunol ; 12: 763098, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1581339

RESUMEN

Although initial immunophenotypical studies on peripheral blood and bronchoalveolar lavage samples have provided a glimpse into the immunopathology of COVID-19, analyses of pulmonary draining lymph nodes are currently scarce. 22 lethal COVID-19 cases and 28 controls were enrolled in this study. Pulmonary draining lymph nodes (mediastinal, tracheal, peribronchial) were collected at autopsy. Control lymph nodes were selected from a range of histomorphological sequelae [unremarkable histology, infectious mononucleosis, follicular hyperplasia, non-SARS related HLH, extrafollicular plasmablast activation, non-SARS related diffuse alveolar damage (DAD), pneumonia]. Samples were mounted on a tissue microarray and underwent immunohistochemical staining for a selection of immunological markers and in-situ hybridization for Epstein Barr Virus (EBV) and SARS-CoV-2. Gene expression profiling was performed using the HTG EdgeSeq Immune Response Panel. Characteristic patterns of a dysregulated immune response were detected in COVID-19: 1. An accumulation of extrafollicular plasmablasts with a relative paucity or depletion of germinal centers. 2. Evidence of T-cell dysregulation demonstrated by immunohistochemical paucity of FOXP3+, Tbet+ and LEF1+ positive T-cells and a downregulation of key genes responsible for T-cell crosstalk, maturation and migration as well as a reactivation of herpes viruses in 6 COVID-19 lymph nodes (EBV, HSV). 3. Macrophage activation by a M2-polarized, CD163+ phenotype and increased incidence of hemophagocytic activity. 4. Microvascular dysfunction, evidenced by an upregulation of hemostatic (CD36, PROCR, VWF) and proangiogenic (FLT1, TEK) genes and an increase of fibrin microthrombi and CD105+ microvessels. Taken together, these findings imply widespread dysregulation of both innate and adoptive pathways with concordant microvascular dysfunction in severe COVID-19.


Asunto(s)
COVID-19/inmunología , COVID-19/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Pulmón , Activación de Macrófagos/inmunología , Masculino , Persona de Mediana Edad , SARS-CoV-2 , Linfocitos T/inmunología , Linfocitos T/patología , Tromboinflamación/inmunología , Tromboinflamación/patología , Tromboinflamación/virología
5.
J Med Virol ; 93(12): 6519-6524, 2021 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1544297

RESUMEN

The COVID-19 pandemic, which has ravaged our world for more than a year, still shapes our agenda with a scale of intensity that fluctuates over time. In our study, we aimed to determine the correlation between serum migration inhibitory factor (MIF) level and disease severity in COVID-19 with different prognoses. Between 15 October 2020 and 20 January 2021, 110 patients over the age of 18 who were diagnosed with COVID-19 and 40 volunteer healthcare personnel were included in our study. MIF levels were measured by enzyme-linked immunosorbent assay. In the comparison of serum MIF values in the patient and control group, it was observed that the MIF level was significantly higher in patients with both moderate and severe COVID-19 levels compared to the control group (p = 0.001, 0.001). In the comparison of serum MIF values of moderate to severe COVID-19 patients, it was observed that MIF level was higher in severe patients (p = 0.001). In the receiver operating characteristic curve analysis performed to differentiate between severe and moderate COVID-19 patients with MIF levels, the area under the curve was observed as 0.78. When the cutoff value of the MIF level was taken as 4.455 ng/ml, the sensitivity was 83% and the specificity was 62%. Failure to adequately balance the pro-inflammatory cytokines synthesized in COVID-19 with anti-inflammatory effect is the most important reason for the aggravation of the disease course. Playing a role in pro-inflammatory cytokine synthesis, MIF can provide important information about the disease prognosis in the early period.


Asunto(s)
COVID-19/patología , Síndrome de Liberación de Citoquinas/sangre , Oxidorreductasas Intramoleculares/sangre , Factores Inhibidores de la Migración de Macrófagos/sangre , Macrófagos/inmunología , SARS-CoV-2/inmunología , Estudios de Casos y Controles , Síndrome de Liberación de Citoquinas/patología , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Activación de Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad
6.
Front Immunol ; 12: 735922, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1477823

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major public health issue. COVID-19 is considered an airway/multi-systemic disease, and demise has been associated with an uncontrolled immune response and a cytokine storm in response to the virus. However, the lung pathology, immune response, and tissue damage associated with COVID-19 demise are poorly described and understood due to safety concerns. Using post-mortem lung tissues from uninfected and COVID-19 deadly cases as well as an unbiased combined analysis of histology, multi-viral and host markers staining, correlative microscopy, confocal, and image analysis, we identified three distinct phenotypes of COVID-19-induced lung damage. First, a COVID-19-induced hemorrhage characterized by minimal immune infiltration and large thrombus; Second, a COVID-19-induced immune infiltration with excessive immune cell infiltration but no hemorrhagic events. The third phenotype correspond to the combination of the two previous ones. We observed the loss of alveolar wall integrity, detachment of lung tissue pieces, fibroblast proliferation, and extensive fibrosis in all three phenotypes. Although lung tissues studied were from lethal COVID-19, a strong immune response was observed in all cases analyzed with significant B cell and poor T cell infiltrations, suggesting an exhausted or compromised immune cellular response in these patients. Overall, our data show that SARS-CoV-2-induced lung damage is highly heterogeneous. These individual differences need to be considered to understand the acute and long-term COVID-19 consequences.


Asunto(s)
COVID-19/mortalidad , COVID-19/patología , Lesión Pulmonar/patología , Alveolos Pulmonares/patología , Fibrosis Pulmonar/patología , Anciano , Anciano de 80 o más Años , Autopsia , Linfocitos T CD8-positivos/inmunología , Síndrome de Liberación de Citoquinas/mortalidad , Síndrome de Liberación de Citoquinas/patología , Células Epiteliales/patología , Femenino , Hemorragia/patología , Humanos , Inflamación/patología , Pulmón/patología , Lesión Pulmonar/virología , Linfopenia/patología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/patología , Neutrófilos/inmunología , SARS-CoV-2 , Trombosis/patología
7.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1304673

RESUMEN

Macrophages (Mφs) are instrumental regulators of the immune response whereby they acquire diverse functional phenotypes following their exposure to microenvironmental cues that govern their differentiation from monocytes and their activation. The complexity and diversity of the mycobacterial cell wall have empowered mycobacteria with potent immunomodulatory capacities. A heat-killed (HK) whole-cell preparation of Mycobacterium obuense (M. obuense) has shown promise as an adjunctive immunotherapeutic agent for the treatment of cancer. Moreover, HK M. obuense has been shown to trigger the differentiation of human monocytes into a monocyte-derived macrophage (MDM) type named Mob-MDM. However, the transcriptomic profile and functional properties of Mob-MDMs remain undefined during an activation state. Here, we characterized cytokine/chemokine release patterns and transcriptomic profiles of lipopolysaccharide (LPS)/interferon γ (IFNγ)-activated human MDMs that were differentiated with HK M. obuense (Mob-MDM(LPS/IFNγ)), macrophage colony-stimulating factor M-MDM(LPS/IFNγ)), or granulocyte/macrophage colony-stimulating factor (GM-MDM(LPS/IFNγ)). Mob-MDM(LPS/IFNγ) demonstrated a unique cytokine/chemokine release pattern (interleukin (IL)-10low, IL-12/23p40low, IL-23p19/p40low, chemokine (C-x-C) motif ligand (CXCL)9low) that was distinct from those of M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ). Furthermore, M-MDM(LPS/IFNγ) maintained IL-10 production at significantly higher levels compared to GM-MDM(LPS/IFNγ) and Mob-MDM(LPS/IFNγ) despite being activated with M1-Mφ-activating stimuli. Comparative RNA sequencing analysis pointed to a distinct transcriptome profile for Mob-MDM(LPS/IFNγ) relative to both M-MDM(LPS/IFNγ) and GM-MDM(LPS/IFNγ) that comprised 417 transcripts. Functional gene-set enrichment analysis revealed significant overrepresentation of signaling pathways and biological processes that were uniquely related to Mob-MDM(LPS/IFNγ). Our findings lay a foundation for the potential integration of HK M. obuense in specific cell-based immunotherapeutic modalities such as adoptive transfer of Mφs (Mob-MDM(LPS/IFNγ)) for cancer treatment.


Asunto(s)
Quimiocinas/metabolismo , Citocinas/metabolismo , Macrófagos/inmunología , Micobacterias no Tuberculosas/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacocinética , Humanos , Factores Inmunológicos/farmacología , Técnicas In Vitro , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Transcriptoma
8.
Microb Pathog ; 153: 104799, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: covidwho-1087152

RESUMEN

The outbreak of coronavirus disease 2019 (COVID-19), caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, has emerged in China in December 2019 and rapidly spread to more than 196 countries worldwide. The physiopathology of human SARS-CoV-2 has not been completely understood, but its pathogenesis has been linked to a disproportionate response of the immune system. Just as described for SARS and MERS, an uncontrolled systemic inflammatory response, known as cytokine release syndrome (CRS) was observed in severe COVID-19 patients. It results from the release by immune and non-immune effector cells of substantial amounts of pro-inflammatory cytokines and appears to contribute to SARS-CoV-2 pulmonary inflammation and extensive lung damage. In addition, hyper-coagulation and thrombosis resulted from the important release of pro-inflammatory cytokines contribute to the lethality of subjects severely infected with SARS-CoV-2. It is therefore essential to have a deep understanding of the various cytokines involved in this exacerbated immune response, and that could be targeted by potential immunological treatments. The aim of this review was to gather the current knowledge about the role of pro-inflammatory cytokines, namely IL-1ß, IL-6, IL-8, IL-17 and TNFα in SARS-CoV-2 CRS, the probable causes and clinical outcomes of this phenomenon in severe cases of COVID-19.


Asunto(s)
COVID-19/patología , Síndrome de Liberación de Citoquinas/patología , Citocinas/sangre , SARS-CoV-2/inmunología , Citocinas/inmunología , Humanos , Activación de Macrófagos/inmunología , Trombosis/patología
9.
Arthritis Rheumatol ; 73(1): 23-35, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-757767

RESUMEN

The clinical progression of the severe acute respiratory syndrome coronavirus 2 infection, coronavirus 2019 (COVID-19), to critical illness is associated with an exaggerated immune response, leading to magnified inflammation termed the "cytokine storm." This response is thought to contribute to the pathogenicity of severe COVID-19. There is an initial weak interferon response and macrophage activation that results in delayed neutrophil recruitment leading to impeded viral clearance. This causes prolonged immune stimulation and the release of proinflammatory cytokines. Elevated inflammatory markers in COVID-19 (e.g., d-dimer, C-reactive protein, lactate dehydrogenase, ferritin, and interleukin-6) are reminiscent of the cytokine storm seen in severe hyperinflammatory macrophage disorders. The dysfunctional immune response in COVID-19 also includes lymphopenia, reduced T cells, reduced natural killer cell maturation, and unmitigated plasmablast proliferation causing aberrant IgG levels. The progression to severe disease is accompanied by endotheliopathy, immunothrombosis, and hypercoagulability. Thus, both parts of the immune system-innate and adaptive-play a significant role in the cytokine storm, multiorgan dysfunction, and coagulopathy. This review highlights the importance of understanding the immunologic mechanisms of COVID-19 as they inform the clinical presentation and advise potential therapeutic targets.


Asunto(s)
Inmunidad Adaptativa/inmunología , COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Inmunidad Innata/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Formación de Anticuerpos , Antivirales/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/sangre , COVID-19/fisiopatología , Inactivadores del Complemento/uso terapéutico , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Humanos , Inmunidad Humoral/inmunología , Inmunoglobulina G/inmunología , Factores Inmunológicos/uso terapéutico , Memoria Inmunológica , Inmunosupresores/uso terapéutico , Interferones/inmunología , Células Asesinas Naturales/inmunología , Linfopenia/inmunología , Activación de Macrófagos/inmunología , Infiltración Neutrófila/inmunología , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Trombofilia/sangre , Trombofilia/inmunología , Trombosis/sangre , Trombosis/inmunología , Tratamiento Farmacológico de COVID-19
10.
Pediatr Allergy Immunol ; 32(1): 17-22, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-748746

RESUMEN

Multisystem inflammatory syndrome in children (MIS-C) during the COVID-19 pandemic raised a global alert from the Centers for Disease Control and Prevention's Health Alert Network. The main manifestations of MIS-C (also known as pediatric MIS (PMIS)) in the setting of a severe inflammatory state include fever, diarrhea, shock, and variable presence of rash, conjunctivitis, extremity edema, and mucous membrane changes. In some cases, these symptoms progressed to multi-organ failure. The low percentage of children with asymptomatic cases compared with mild illness and moderate illness could be correlated with the rare cases of MIS-C. One potential explanation for the progression to severe MIS-C disease despite the presence of readily detectable anti-SARS-CoV-2 antibodies could be due to the potential role of antibody-dependent enhancement (ADE). We reason that the incidence of the ADE phenomenon whereby the pathogen-specific antibodies can promote pathology should be considered in vaccine development against SARS-CoV-2.


Asunto(s)
COVID-19/epidemiología , Síndrome de Respuesta Inflamatoria Sistémica/epidemiología , Adolescente , Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo/inmunología , COVID-19/inmunología , Niño , Preescolar , Conjuntivitis/epidemiología , Diarrea/epidemiología , Exantema/epidemiología , Humanos , Lactante , Activación de Macrófagos/inmunología , Pandemias , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Adulto Joven
11.
Front Immunol ; 11: 1580, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-647056

RESUMEN

SARS-CoV-2 might directly activate NLRP3 inflammasome resulting in an endogenous adjuvant activity necessary to mount a proper adaptive immune response against the virus. Heterogeneous response of COVID-19 patients could be attributed to differences in not being able to properly downregulate NLRP3 inflammasome activation. This relates to the fitness of the immune system of the individual challenged by the virus. Patients with a reduced immune fitness can demonstrate a dysregulated NLRP3 inflammasome activity resulting in severe COVID-19 with tissue damage and a cytokine storm. We sketch the outlines of five possible scenarios for COVID-19 in medical practice and provide potential treatment options targeting dysregulated endogenous adjuvant activity in severe COVID-19 patients.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Proteína HMGB1/metabolismo , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/inmunología , Betacoronavirus/inmunología , COVID-19 , Infecciones por Coronavirus/patología , Citocinas/metabolismo , Humanos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Pandemias , Neumonía Viral/patología , SARS-CoV-2
12.
J Autoimmun ; 114: 102506, 2020 11.
Artículo en Inglés | MEDLINE | ID: covidwho-599328

RESUMEN

Coronavirus disease 2019 (COVID-19) has been categorized as evolving in overlapping phases. First, there is a viral phase that may well be asymptomatic or mild in the majority, perhaps 80% of patients. The pathophysiological mechanisms resulting in minimal disease in this initial phase are not well known. In the remaining 20% of cases, the disease may become severe and/or critical. In most patients of this latter group, there is a phase characterized by the hyperresponsiveness of the immune system. A third phase corresponds to a state of hypercoagulability. Finally, in the fourth stage organ injury and failure occur. Appearance of autoinflammatory/autoimmune phenomena in patients with COVID-19 calls attention for the development of new strategies for the management of life-threatening conditions in critically ill patients. Antiphospholipid syndrome, autoimmune cytopenia, Guillain-Barré syndrome and Kawasaki disease have each been reported in patients with COVID-19. Here we present a scoping review of the relevant immunological findings in COVID-19 as well as the current reports about autoinflammatory/autoimmune conditions associated with the disease. These observations have crucial therapeutic implications since immunomodulatory drugs are at present the most likely best candidates for COVID-19 therapy. Clinicians should be aware of these conditions in patients with COVID-19, and these observations should be considered in the current development of vaccines.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Neumonía Viral/inmunología , Inmunidad Adaptativa/genética , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/virología , Betacoronavirus/aislamiento & purificación , COVID-19 , Prueba de COVID-19 , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/terapia , Enfermedad Crítica , Síndrome de Liberación de Citoquinas/diagnóstico , Síndrome de Liberación de Citoquinas/terapia , Síndrome de Liberación de Citoquinas/virología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata/genética , Inmunización Pasiva/métodos , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Masculino , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/epidemiología , Neumonía Viral/terapia , Factores de Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Factores Sexuales , Sueroterapia para COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA