Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 23(15)2022 Aug 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1987828

RESUMEN

Blood group antigen is a class of heritable antigenic substances present on the erythrocyte membrane. However, the role of blood group antigens in cancer prognosis is still largely unclear. In this study, we investigated the expression of 33 blood group antigen genes and their association with the prognosis of 30 types of cancers in 31,870 tumor tissue samples. Our results revealed that blood group antigens are abnormally expressed in a variety of cancers. The high expression of these antigen genes was mainly related to the activation of the epithelial-mesenchymal transition (EMT) pathway. High expression of seven antigen genes, i.e., FUT7, AQP1, P1, C4A, AQP3, KEL and DARC, were significantly associated with good OS (Overall Survival) in six types of cancers, while ten genes, i.e., AQP1, P1, C4A, AQP3, BSG, CD44, CD151, LU, FUT2, and SEMA7A, were associated with poor OS in three types of cancers. Kidney renal clear cell carcinoma (KIRC) is associated with the largest number (14 genes) of prognostic antigen genes, i.e., CD44, CD151, SEMA7A, FUT7, CR1, AQP1, GYPA, FUT3, FUT6, FUT1, SLC14A1, ERMAP, C4A, and B3GALT3. High expression of SEMA7A gene was significantly correlated with a poor prognosis of KIRC in this analysis but has not been reported previously. SEMA7A might be a putative biomarker for poor prognosis in KIRC. In conclusion, our analysis indicates that blood group antigens may play functional important roles in tumorigenesis, progression, and especially prognosis. These results provide data to support prognostic marker development and future clinical management.


Asunto(s)
Antígenos de Grupos Sanguíneos , Carcinoma de Células Renales , Neoplasias Renales , Semaforinas , Antígenos CD , Biomarcadores , Carcinoma de Células Renales/patología , Proteínas Ligadas a GPI , Humanos , Riñón/metabolismo , Neoplasias Renales/metabolismo , Pronóstico , Semaforinas/genética
2.
J Cell Physiol ; 237(8): 3394-3407, 2022 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1905874

RESUMEN

Purinergic signaling modulates immune function and is involved in the immunopathogenesis of several viral infections. This study aimed to investigate alterations in purinergic pathways in coronavirus disease 2019 (COVID-19) patients. Mild and severe COVID-19 patients had lower extracellular adenosine triphosphate and adenosine levels, and higher cytokines than healthy controls. Mild COVID-19 patients presented lower frequencies of CD4+ CD25+ CD39+ (activated/memory regulatory T cell [mTreg]) and increased frequencies of high-differentiated (CD27- CD28- ) CD8+ T cells compared with healthy controls. Severe COVID-19 patients also showed higher frequencies of CD4+ CD39+ , CD4+ CD25- CD39+ (memory T effector cell), and high-differentiated CD8+ T cells (CD27- CD28- ), and diminished frequencies of CD4+ CD73+ , CD4+ CD25+ CD39+ mTreg cell, CD8+ CD73+ , and low-differentiated CD8+ T cells (CD27+ CD28+ ) in the blood in relation to mild COVID-19 patients and controls. Moreover, severe COVID-19 patients presented higher expression of PD-1 on low-differentiated CD8+ T cells. Both severe and mild COVID-19 patients presented higher frequencies of CD4+ Annexin-V+ and CD8+ Annexin-V+ T cells, indicating increased T-cell apoptosis. Plasma samples collected from severe COVID-19 patients were able to decrease the expression of CD73 on CD4+ and CD8+ T cells of a healthy donor. Interestingly, the in vitro incubation of peripheral blood mononuclear cell from severe COVID-19 patients with adenosine reduced the nuclear factor-κB activation in T cells and monocytes. Together, these data add new knowledge to the COVID-19 immunopathology through purinergic regulation.


Asunto(s)
5'-Nucleotidasa , Apirasa , COVID-19 , Linfocitos T , 5'-Nucleotidasa/metabolismo , Adenosina/sangre , Adenosina Trifosfato/sangre , Anexinas , Apirasa/metabolismo , Antígenos CD28/metabolismo , COVID-19/inmunología , Citocinas/sangre , Proteínas Ligadas a GPI/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Receptores Purinérgicos , Transducción de Señal , Linfocitos T/inmunología
3.
Nat Immunol ; 23(5): 679-691, 2022 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1878539

RESUMEN

Here we report the identification of human CD66b-CD64dimCD115- neutrophil-committed progenitor cells (NCPs) within the SSCloCD45dimCD34+ and CD34dim/- subsets in the bone marrow. NCPs were either CD45RA+ or CD45RA-, and in vitro experiments showed that CD45RA acquisition was not mandatory for their maturation process. NCPs exclusively generated human CD66b+ neutrophils in both in vitro differentiation and in vivo adoptive transfer experiments. Single-cell RNA-sequencing analysis indicated NCPs fell into four clusters, characterized by different maturation stages and distributed along two differentiation routes. One of the clusters was characterized by an interferon-stimulated gene signature, consistent with the reported expansion of peripheral mature neutrophil subsets that express interferon-stimulated genes in diseased individuals. Finally, comparison of transcriptomic and phenotypic profiles indicated NCPs represented earlier neutrophil precursors than the previously described early neutrophil progenitors (eNePs), proNeus and COVID-19 proNeus. Altogether, our data shed light on the very early phases of neutrophil ontogeny.


Asunto(s)
Antígenos CD , Médula Ósea , Moléculas de Adhesión Celular , Diferenciación Celular , Neutrófilos , Receptor de Factor Estimulante de Colonias de Macrófagos , Receptores de IgG , Células de la Médula Ósea , COVID-19 , Proteínas Ligadas a GPI , Humanos , Interferones , Neutrófilos/citología
4.
Viruses ; 14(1)2021 12 28.
Artículo en Inglés | MEDLINE | ID: covidwho-1715736

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) infection induces elevated levels of inflammatory cytokines, which are mainly produced by the innate response to the virus. The role of NK cells, which are potent producers of IFN-γ and cytotoxicity, has not been sufficiently studied in the setting of SARS-CoV-2 infection. We confirmed a different distribution of NK cell subsets in hospitalized COVID-19 patients despite their NK cell deficiency. The impairment of this innate defense is mainly focused on the cytotoxic capacity of the CD56dim NK cells. On the one hand, we found an expansion of the CD56dimCD16neg NK subset, lower cytotoxic capacities, and high frequencies of inhibitory 2DL1 and 2DL1/S1 KIR receptors in COVID-19 patients. On the other hand, the depletion of CD56dimCD16dim/bright NK cell subsets, high cytotoxic capacities, and high frequencies of inhibitory 2DL1 KIR receptors were found in COVID-19 patients. In contrast, no differences in the distribution of CD56bright NK cell subsets were found in this study. These alterations in the distribution and phenotype of NK cells might enhance the impairment of this crucial innate line of defense during COVID-19 infection.


Asunto(s)
COVID-19/inmunología , Células Asesinas Naturales/metabolismo , Subgrupos Linfocitarios/metabolismo , Receptores KIR/metabolismo , Anciano , Antígeno CD56/metabolismo , COVID-19/sangre , Femenino , Proteínas Ligadas a GPI/metabolismo , Hospitalización , Humanos , Inflamación , Masculino , Persona de Mediana Edad , Receptores de IgG/metabolismo , SARS-CoV-2
5.
Genome Med ; 14(1): 16, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: covidwho-1690882

RESUMEN

BACKGROUND: Understanding the host genetic architecture and viral immunity contributes to the development of effective vaccines and therapeutics for controlling the COVID-19 pandemic. Alterations of immune responses in peripheral blood mononuclear cells play a crucial role in the detrimental progression of COVID-19. However, the effects of host genetic factors on immune responses for severe COVID-19 remain largely unknown. METHODS: We constructed a computational framework to characterize the host genetics that influence immune cell subpopulations for severe COVID-19 by integrating GWAS summary statistics (N = 969,689 samples) with four independent scRNA-seq datasets containing healthy controls and patients with mild, moderate, and severe symptom (N = 606,534 cells). We collected 10 predefined gene sets including inflammatory and cytokine genes to calculate cell state score for evaluating the immunological features of individual immune cells. RESULTS: We found that 34 risk genes were significantly associated with severe COVID-19, and the number of highly expressed genes increased with the severity of COVID-19. Three cell subtypes that are CD16+monocytes, megakaryocytes, and memory CD8+T cells were significantly enriched by COVID-19-related genetic association signals. Notably, three causal risk genes of CCR1, CXCR6, and ABO were highly expressed in these three cell types, respectively. CCR1+CD16+monocytes and ABO+ megakaryocytes with significantly up-regulated genes, including S100A12, S100A8, S100A9, and IFITM1, confer higher risk to the dysregulated immune response among severe patients. CXCR6+ memory CD8+ T cells exhibit a notable polyfunctionality including elevation of proliferation, migration, and chemotaxis. Moreover, we observed an increase in cell-cell interactions of both CCR1+ CD16+monocytes and CXCR6+ memory CD8+T cells in severe patients compared to normal controls among both PBMCs and lung tissues. The enhanced interactions of CXCR6+ memory CD8+T cells with epithelial cells facilitate the recruitment of this specific population of T cells to airways, promoting CD8+T cell-mediated immunity against COVID-19 infection. CONCLUSIONS: We uncover a major genetics-modulated immunological shift between mild and severe infection, including an elevated expression of genetics-risk genes, increase in inflammatory cytokines, and of functional immune cell subsets aggravating disease severity, which provides novel insights into parsing the host genetic determinants that influence peripheral immune cells in severe COVID-19.


Asunto(s)
Linfocitos T CD8-positivos/virología , COVID-19/genética , COVID-19/patología , Monocitos/virología , Análisis de la Célula Individual/métodos , COVID-19/inmunología , Biología Computacional/métodos , Proteínas Ligadas a GPI/metabolismo , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Células Progenitoras de Megacariocitos/inmunología , Células Progenitoras de Megacariocitos/virología , Monocitos/metabolismo , Sitios de Carácter Cuantitativo , Receptores CCR1/inmunología , Receptores CCR1/metabolismo , Receptores CXCR6/inmunología , Receptores CXCR6/metabolismo , Receptores de IgG/metabolismo , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad
6.
Sci Adv ; 8(5): eabl8920, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1673337

RESUMEN

Dexamethasone is widely used as an immunosuppressive therapy and recently as COVID-19 treatment. Here, we demonstrate that dexamethasone sensitizes to ferroptosis, a form of iron-catalyzed necrosis, previously suggested to contribute to diseases such as acute kidney injury, myocardial infarction, and stroke, all of which are triggered by glutathione (GSH) depletion. GSH levels were significantly decreased by dexamethasone. Mechanistically, we identified that dexamethasone up-regulated the GSH metabolism regulating protein dipeptidase-1 (DPEP1) in a glucocorticoid receptor (GR)-dependent manner. DPEP1 knockdown reversed the phenotype of dexamethasone-induced ferroptosis sensitization. Ferroptosis inhibitors, the DPEP1 inhibitor cilastatin, or genetic DPEP1 inactivation reversed the dexamethasone-induced increase in tubular necrosis in freshly isolated renal tubules. Our data indicate that dexamethasone sensitizes to ferroptosis by a GR-mediated increase in DPEP1 expression and GSH depletion. Together, we identified a previously unknown mechanism of glucocorticoid-mediated sensitization to ferroptosis bearing clinical and therapeutic implications.


Asunto(s)
Dexametasona/farmacología , Dipeptidasas/genética , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Receptores de Glucocorticoides/metabolismo , Carbolinas/efectos adversos , Carbolinas/farmacología , Línea Celular , Dipeptidasas/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunofenotipificación , Oxidación-Reducción/efectos de los fármacos , Piperazinas/efectos adversos , Piperazinas/farmacología
7.
Front Immunol ; 12: 799558, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1662582

RESUMEN

The poor outcome of the coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, is associated with systemic hyperinflammatory response and immunopathology. Although inflammasome and oxidative stress have independently been implicated in COVID-19, it is poorly understood whether these two pathways cooperatively contribute to disease severity. Herein, we found an enrichment of CD14highCD16- monocytes displaying inflammasome activation evidenced by caspase-1/ASC-speck formation in severe COVID-19 patients when compared to mild ones and healthy controls, respectively. Those cells also showed aberrant levels of mitochondrial superoxide and lipid peroxidation, both hallmarks of the oxidative stress response, which strongly correlated with caspase-1 activity. In addition, we found that NLRP3 inflammasome-derived IL-1ß secretion by SARS-CoV-2-exposed monocytes in vitro was partially dependent on lipid peroxidation. Importantly, altered inflammasome and stress responses persisted after short-term patient recovery. Collectively, our findings suggest oxidative stress/NLRP3 signaling pathway as a potential target for host-directed therapy to mitigate early COVID-19 hyperinflammation and also its long-term outcomes.


Asunto(s)
COVID-19/metabolismo , Inflamasomas/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Monocitos/metabolismo , Estrés Oxidativo/fisiología , Receptores de IgG/metabolismo , Anciano , COVID-19/patología , Caspasa 1/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/patología , Monocitos/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , SARS-CoV-2/metabolismo , Transducción de Señal/fisiología
8.
ChemistryOpen ; 10(11): 1133-1141, 2021 11.
Artículo en Inglés | MEDLINE | ID: covidwho-1520270

RESUMEN

We present in this work a first X-ray Absorption Spectroscopy study of the interactions of Zn with human BST2/tetherin and SARS-CoV-2 orf7a proteins as well as with some of their complexes. The analysis of the XANES region of the measured spectra shows that Zn binds to BST2, as well as to orf7a, thus resulting in the formation of BST2-orf7a complexes. This structural information confirms the the conjecture, recently put forward by some of the present Authors, according to which the accessory orf7a (and possibly also orf8) viral protein are capable of interfering with the BST2 antiviral activity. Our explanation for this behavior is that, when BST2 gets in contact with Zn bound to the orf7a Cys15 ligand, it has the ability of displacing the metal owing to the creation of a new disulfide bridge across the two proteins. The formation of this BST2-orf7a complex destabilizes BST2 dimerization, thus impairing the antiviral activity of the latter.


Asunto(s)
Antígenos CD/metabolismo , SARS-CoV-2/química , Proteínas Virales/metabolismo , Zinc/metabolismo , Cisteína/química , Proteínas Ligadas a GPI/metabolismo , Histidina/química , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Espectroscopía de Absorción de Rayos X
9.
Sci Rep ; 11(1): 21514, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1500512

RESUMEN

Coronavirus disease 2019 (COVID-19) is associated with systemic inflammation. A wide range of adipokines activities suggests they influence pathogenesis and infection course. The aim was to assess concentrations of chemerin, omentin, and vaspin among COVID-19 patients with an emphasis on adipokines relationship with COVID-19 severity, concomitant metabolic abnormalities and liver dysfunction. Serum chemerin, omentin and vaspin concentrations were measured in serum collected from 70 COVID-19 patients at the moment of admission to hospital, before any treatment was applied and 20 healthy controls. Serum chemerin and omentin concentrations were significantly decreased in COVID-19 patients compared to healthy volunteers (271.0 vs. 373.0 ng/ml; p < 0.001 and 482.1 vs. 814.3 ng/ml; p = 0.01, respectively). There were no correlations of analyzed adipokines with COVID-19 severity based on the presence of pneumonia, dyspnea, or necessity of Intensive Care Unit hospitalization (ICU). Liver test abnormalities did not influence adipokines levels. Elevated GGT activity was associated with ICU admission, presence of pneumonia and elevated concentrations of CRP, ferritin and interleukin 6. Chemerin and omentin depletion in COVID-19 patients suggests that this adipokines deficiency play influential role in disease pathogenesis. However, there was no relationship between lower adipokines level and frequency of COVID-19 symptoms as well as disease severity. The only predictive factor which could predispose to a more severe COVID-19 course, including the presence of pneumonia and ICU hospitalization, was GGT activity.


Asunto(s)
Adipoquinas/sangre , Quimiocinas/sangre , Citocinas/sangre , Lectinas/sangre , Serpinas/sangre , Anciano , Índice de Masa Corporal , Proteína C-Reactiva/análisis , COVID-19/complicaciones , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Estudios de Casos y Controles , Femenino , Proteínas Ligadas a GPI/sangre , Hospitalización , Humanos , Hígado/metabolismo , Masculino , Síndrome Metabólico/complicaciones , Persona de Mediana Edad , SARS-CoV-2/aislamiento & purificación , gamma-Glutamiltransferasa/metabolismo
10.
Cardiovasc Res ; 117(1): 224-239, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1387842

RESUMEN

AIMS: To elucidate the prognostic role of monocytes in the immune response of patients with coronary artery disease (CAD) at risk for life-threatening heart and lung injury as major complications of SARS-CoV-2 infection. METHODS AND RESULTS: From February to April 2020, we prospectively studied a cohort of 96 participants comprising 47 consecutive patients with CAD and acute SARS-CoV-2 infection (CAD + SARS-CoV-2), 19 CAD patients without infections, and 30 healthy controls. Clinical assessment included blood sampling, echocardiography, and electrocardiography within 12 h of admission. Respiratory failure was stratified by the Horovitz Index (HI) as moderately/severely impaired when HI ≤200 mmHg. The clinical endpoint (EP) was defined as HI ≤200 mmHg with subsequent mechanical ventilation within a follow-up of 30 days. The numbers of CD14dimCD16+ non-classical monocytes in peripheral blood were remarkably low in CAD + SARS-CoV-2 compared with CAD patients without infection and healthy controls (P < 0.0001). Moreover, these CD14dimCD16 monocytes showed decreased expression of established markers of adhesion, migration, and T-cell activation (CD54, CD62L, CX3CR1, CD80, and HLA-DR). Decreased numbers of CD14dimCD16+ monocytes were associated with the occurrence of EP. Kaplan-Meier curves illustrate that CAD + SARS-CoV-2 patients with numbers below the median of CD14dimCD16+ monocytes (median 1443 cells/mL) reached EP significantly more often compared to patients with numbers above the median (log-rank 5.03, P = 0.025). CONCLUSION: Decreased numbers of CD14dimCD16+ monocytes are associated with rapidly progressive respiratory failure in CAD + SARS-CoV-2 patients. Intensified risk assessments comprising monocyte sub- and phenotypes may help to identify patients at risk for respiratory failure.


Asunto(s)
COVID-19/complicaciones , Enfermedad de la Arteria Coronaria/complicaciones , Receptores de Lipopolisacáridos/análisis , Monocitos/fisiología , Receptores de IgG/análisis , SARS-CoV-2 , Anciano , Anciano de 80 o más Años , COVID-19/inmunología , Enfermedad de la Arteria Coronaria/inmunología , Femenino , Proteínas Ligadas a GPI/análisis , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Fenotipo , Estudios Retrospectivos
11.
J Infect Dis ; 224(4): 575-585, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1358459

RESUMEN

Severe coronavirus disease 2019 (COVID-19) is associated with an overactive inflammatory response mediated by macrophages. Here, we analyzed the phenotype and function of neutrophils in patients with COVID-19. We found that neutrophils from patients with severe COVID-19 express high levels of CD11b and CD66b, spontaneously produce CXCL8 and CCL2, and show a strong association with platelets. Production of CXCL8 correlated with plasma concentrations of lactate dehydrogenase and D-dimer. Whole blood assays revealed that neutrophils from patients with severe COVID-19 show a clear association with immunoglobulin G (IgG) immune complexes. Moreover, we found that sera from patients with severe disease contain high levels of immune complexes and activate neutrophils through a mechanism partially dependent on FcγRII (CD32). Interestingly, when integrated in immune complexes, anti-severe acute respiratory syndrome coronavirus 2 IgG antibodies from patients with severe COVID-19 displayed a higher proinflammatory profile compared with antibodies from patients with mild disease. Our study suggests that IgG immune complexes might promote the acquisition of an inflammatory signature by neutrophils, worsening the course of COVID-19.


Asunto(s)
Anticuerpos Antivirales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , COVID-19/inmunología , Inmunoglobulina G/inmunología , Activación Neutrófila/inmunología , Adulto , Anciano , Anticuerpos Antivirales/sangre , Complejo Antígeno-Anticuerpo/sangre , Antígenos CD/inmunología , Antígeno CD11b/inmunología , Moléculas de Adhesión Celular/inmunología , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunoglobulina G/sangre , Interleucina-8/inmunología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Receptores de IgG/inmunología , SARS-CoV-2/inmunología , Adulto Joven
12.
Dis Markers ; 2021: 5566826, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1341351

RESUMEN

An excess formation of neutrophil extracellular traps (NETs), previously shown to be strongly associated with cytokine storm and acute respiratory distress syndrome (ARDS) with prevalent endothelial dysfunction and thrombosis, has been postulated to be a central factor influencing the pathophysiology and clinical presentation of severe COVID-19. A growing number of serological and morphological evidence has added to this assumption, also in regard to potential treatment options. In this study, we used immunohistochemistry and histochemistry to trace NETs and their molecular markers in autopsy lung tissue from seven COVID-19 patients. Quantification of key immunomorphological features enabled comparison with non-COVID-19 diffuse alveolar damage. Our results strengthen and extend recent findings, confirming that NETs are abundantly present in seriously damaged COVID-19 lung tissue, especially in association with microthrombi of the alveolar capillaries. In addition, we provide evidence that low-density neutrophils (LDNs), which are especially prone to NETosis, contribute substantially to COVID-19-associated lung damage in general and vascular blockages in particular.


Asunto(s)
COVID-19/patología , Trampas Extracelulares , Lesión Pulmonar/patología , Neutrófilos/patología , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Autopsia , Moléculas de Adhesión Celular/metabolismo , Trampas Extracelulares/virología , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunohistoquímica , Pulmón/patología , Pulmón/virología , Lesión Pulmonar/virología , Masculino , Neutrófilos/metabolismo , Neutrófilos/virología , Peroxidasa/metabolismo
13.
Front Immunol ; 12: 665773, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1264333

RESUMEN

The COVID-19 pandemic has caused more than three million deaths globally. The severity of the disease is characterized, in part, by a dysregulated immune response. CD16+ monocytes are innate immune cells involved in inflammatory responses to viral infections, and tissue repair, among other functions. We characterized the transcriptional changes in CD16+ monocytes from PBMC of people with COVID-19, and from healthy individuals using publicly available single cell RNA sequencing data. CD16+ monocytes from people with COVID-19 compared to those from healthy individuals expressed transcriptional changes indicative of increased cell activation, and induction of a migratory phenotype. We also analyzed COVID-19 cases based on severity of the disease and found that mild cases were characterized by upregulation of interferon response and MHC class II related genes, whereas the severe cases had dysregulated expression of mitochondrial and antigen presentation genes, and upregulated inflammatory, cell movement, and apoptotic gene signatures. These results suggest that CD16+ monocytes in people with COVID-19 contribute to a dysregulated host response characterized by decreased antigen presentation, and an elevated inflammatory response with increased monocytic infiltration into tissues. Our results show that there are transcriptomic changes in CD16+ monocytes that may impact the functions of these cells, contributing to the pathogenesis and severity of COVID-19.


Asunto(s)
COVID-19/virología , Monocitos/virología , Receptores de IgG/metabolismo , SARS-CoV-2/patogenicidad , Transcripción Genética , Transcriptoma , Adulto , Anciano , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , COVID-19/genética , COVID-19/inmunología , COVID-19/metabolismo , Estudios de Casos y Controles , Citocinas/genética , Citocinas/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , RNA-Seq , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Adulto Joven
14.
JCI Insight ; 6(9)2021 05 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1228934

RESUMEN

SARS coronavirus 2 (SARS-CoV-2) is a novel viral pathogen that causes a clinical disease called coronavirus disease 2019 (COVID-19). Although most COVID-19 cases are asymptomatic or involve mild upper respiratory tract symptoms, a significant number of patients develop severe or critical disease. Patients with severe COVID-19 commonly present with viral pneumonia that may progress to life-threatening acute respiratory distress syndrome (ARDS). Patients with COVID-19 are also predisposed to venous and arterial thromboses that are associated with a poorer prognosis. The present study identified the emergence of a low-density inflammatory neutrophil (LDN) population expressing intermediate levels of CD16 (CD16Int) in patients with COVID-19. These cells demonstrated proinflammatory gene signatures, activated platelets, spontaneously formed neutrophil extracellular traps, and enhanced phagocytic capacity and cytokine production. Strikingly, CD16Int neutrophils were also the major immune cells within the bronchoalveolar lavage fluid, exhibiting increased CXCR3 but loss of CD44 and CD38 expression. The percentage of circulating CD16Int LDNs was associated with D-dimer, ferritin, and systemic IL-6 and TNF-α levels and changed over time with altered disease status. Our data suggest that the CD16Int LDN subset contributes to COVID-19-associated coagulopathy, systemic inflammation, and ARDS. The frequency of that LDN subset in the circulation could serve as an adjunct clinical marker to monitor disease status and progression.


Asunto(s)
Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/etiología , COVID-19/sangre , COVID-19/complicaciones , Neutrófilos/inmunología , SARS-CoV-2 , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Trastornos de la Coagulación Sanguínea/inmunología , COVID-19/inmunología , Citocinas/sangre , Femenino , Proteínas Ligadas a GPI/sangre , Hospitalización , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Neutrófilos/clasificación , Pandemias , Fagocitosis , Activación Plaquetaria , Receptores de IgG/sangre , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/inmunología , Índice de Severidad de la Enfermedad
15.
IUBMB Life ; 73(6): 843-854, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1219298

RESUMEN

The 78 kDa glucose-regulated protein (GRP78) is an endoplasmic reticulum (ER)-resident molecular chaperone. GRP78 is a member of the 70 kDa heat shock family of proteins involved in correcting and clearing misfolded proteins in the ER. In response to cellular stress, GRP78 escapes from the ER and moves to the plasma membrane where it (a) functions as a receptor for many ligands, and (b) behaves as an autoantigen for autoantibodies that contribute to human disease and cancer. Cell surface GRP78 (csGRP78) associates with the major histocompatibility complex class I (MHC-I), and is the port of entry for several viruses, including the predictive binding of the novel SARS-CoV-2. Furthermore, csGRP78 is found in association with partners as diverse as the teratocarcinoma-derived growth factor 1 (Cripto), the melanocortin-4 receptor (MC4R) and the DnaJ-like protein MTJ-1. CsGRP78 also serves as a receptor for a large variety of ligands including activated α2 -macroglobulin (α2 M*), plasminogen kringle 5 (K5), microplasminogen, the voltage-dependent anion channel (VDAC), tissue factor (TF), and the prostate apoptosis response-4 protein (Par-4). In this review, we discuss the mechanisms involved in the translocation of GRP78 from the ER to the cell surface, and the role of secreted GRP78 and its autoantibodies in cancer and neurological disorders.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/inmunología , COVID-19/transmisión , Proteínas de Choque Térmico/fisiología , Proteínas de Neoplasias/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores de Superficie Celular/fisiología , Receptores Virales/fisiología , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Supervivencia Celular , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/fisiología , Exosomas , Proteínas Ligadas a GPI/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/inmunología , Humanos , Ligandos , Invasividad Neoplásica , Proteínas de Neoplasias/inmunología , Proteínas del Tejido Nervioso/inmunología , Dominios Proteicos , Transporte de Proteínas , Transducción de Señal , Microambiente Tumoral , Respuesta de Proteína Desplegada/fisiología , Internalización del Virus
16.
Front Immunol ; 12: 651656, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1211812

RESUMEN

Although immune dysfunction is a key feature of coronavirus disease 2019 (COVID-19), the metabolism-related mechanisms remain elusive. Here, by reanalyzing single-cell RNA sequencing data, we delineated metabolic remodeling in peripheral blood mononuclear cells (PBMCs) to elucidate the metabolic mechanisms that may lead to the progression of severe COVID-19. After scoring the metabolism-related biological processes and signaling pathways, we found that mono-CD14+ cells expressed higher levels of glycolysis-related genes (PKM, LDHA and PKM) and PPP-related genes (PGD and TKT) in severe patients than in mild patients. These genes may contribute to the hyperinflammation in mono-CD14+ cells of patients with severe COVID-19. The mono-CD16+ cell population in COVID-19 patients showed reduced transcription levels of genes related to lysine degradation (NSD1, KMT2E, and SETD2) and elevated transcription levels of genes involved in OXPHOS (ATP6V1B2, ATP5A1, ATP5E, and ATP5B), which may inhibit M2-like polarization. Plasma cells also expressed higher levels of the OXPHOS gene ATP13A3 in COVID-19 patients, which was positively associated with antibody secretion and survival of PCs. Moreover, enhanced glycolysis or OXPHOS was positively associated with the differentiation of memory B cells into plasmablasts or plasma cells. This study comprehensively investigated the metabolic features of peripheral immune cells and revealed that metabolic changes exacerbated inflammation in monocytes and promoted antibody secretion and cell survival in PCs in COVID-19 patients, especially those with severe disease.


Asunto(s)
COVID-19/inmunología , Glucólisis/genética , Lisina/metabolismo , Monocitos/metabolismo , Análisis de la Célula Individual/métodos , Adenosina Trifosfatasas/sangre , Adenosina Trifosfatasas/genética , Anticuerpos/metabolismo , COVID-19/metabolismo , COVID-19/fisiopatología , Bases de Datos Genéticas , Proteínas Ligadas a GPI/metabolismo , Ontología de Genes , Hematopoyesis/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Receptores de Lipopolisacáridos/metabolismo , Lisina/genética , Proteínas de Transporte de Membrana/sangre , Proteínas de Transporte de Membrana/genética , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/fisiología , Monocitos/inmunología , Monocitos/patología , Fosforilación Oxidativa , RNA-Seq , Receptores de IgG/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma/genética
17.
Mol Cell ; 81(12): 2656-2668.e8, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: covidwho-1179919

RESUMEN

A deficient interferon (IFN) response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated as a determinant of severe coronavirus disease 2019 (COVID-19). To identify the molecular effectors that govern IFN control of SARS-CoV-2 infection, we conducted a large-scale gain-of-function analysis that evaluated the impact of human IFN-stimulated genes (ISGs) on viral replication. A limited subset of ISGs were found to control viral infection, including endosomal factors inhibiting viral entry, RNA binding proteins suppressing viral RNA synthesis, and a highly enriched cluster of endoplasmic reticulum (ER)/Golgi-resident ISGs inhibiting viral assembly/egress. These included broad-acting antiviral ISGs and eight ISGs that specifically inhibited SARS-CoV-2 and SARS-CoV-1 replication. Among the broad-acting ISGs was BST2/tetherin, which impeded viral release and is antagonized by SARS-CoV-2 Orf7a protein. Overall, these data illuminate a set of ISGs that underlie innate immune control of SARS-CoV-2/SARS-CoV-1 infection, which will facilitate the understanding of host determinants that impact disease severity and offer potential therapeutic strategies for COVID-19.


Asunto(s)
Antígenos CD/genética , Interacciones Huésped-Patógeno/genética , Factores Reguladores del Interferón/genética , Interferón Tipo I/genética , SARS-CoV-2/genética , Proteínas Virales/genética , Animales , Antígenos CD/química , Antígenos CD/inmunología , Sitios de Unión , Línea Celular Tumoral , Chlorocebus aethiops , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/virología , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Aparato de Golgi/virología , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Factores Reguladores del Interferón/clasificación , Factores Reguladores del Interferón/inmunología , Interferón Tipo I/inmunología , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , SARS-CoV-2/inmunología , Transducción de Señal , Células Vero , Proteínas Virales/química , Proteínas Virales/inmunología , Internalización del Virus , Liberación del Virus/genética , Liberación del Virus/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
18.
Curr Drug Targets ; 22(17): 2006-2020, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1124782

RESUMEN

BACKGROUND: By the end of 2019, the sudden outbreak of the novel coronavirus disease (COVID-19) has become a global threat. It is called COVID-19 because it was caused by the novel coronavirus (SARS-COV-2) in 2019. A total of 1.9 M deaths and 87.9 M cases have been reported all over the world, where 49M cases have recovered so far. Scientists are working hard to find chemotherapeutics and vaccines for COVID-19. Mutations in SARS-CoV-2 have been observed in a combination of several hazardous stresses, making them more resistant and beneficial. So to break down the viral system, the disease targets are examined. OBJECTIVE: In today's review, a comprehensive study of spike protein explains the main purpose of the novel coronavirus and how to prevent the spread of the disease virus cross-transmission from infected to a healthy person. METHODS: Covid-19 has already been declared a pandemic by the World Health Organization (WHO) due to its result in causing death and severe illness globally. SARS-CoV-2 is highly contagious; however, the intermediate host of the novel coronavirus is not clear. To explore the mechanisms of disease, one of the viral targets, such as the spike protein that binds to human cells and causes the disease by altering its genetic structure which is considered along with potential inhibitors. RESULTS: It has been shown that the interaction of receptor-binding domain (RBD) protein of SARS- CoV-2 spike and the angiotensin-converting enzyme 2 (ACE2) host receptor and further replication of coronavirus spike protein causes its invasion in the host cell. The human Lymphocyte antigen 6 complex, Locus E (LY6E), inhibits the entry of CoV into host cells by interfering with the human gene, inducing spike protein-mediated membrane fusion. Some natural formulations have also been shown to prevent spike protein from binding to the host cell. CONCLUSION: With the development of the LY6E gene activator that can inhibit spike protein- ACE2-mediated membrane fusion, new opportunities for SARS-CoV-2 treatment may emerge. Existing antiviral fusion inhibitors and natural compounds targeting spike resistance can serve as a template for further SARS-CoV-2 drug formulation.


Asunto(s)
Antivirales , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus , Enzima Convertidora de Angiotensina 2 , Antígenos de Superficie , Antivirales/farmacología , Productos Biológicos/farmacología , COVID-19 , Proteínas Ligadas a GPI , Humanos , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores
19.
Front Immunol ; 11: 595950, 2020.
Artículo en Inglés | MEDLINE | ID: covidwho-1110296

RESUMEN

In recent years, the success of immunotherapy targeting immunoregulatory receptors (immune checkpoints) in cancer have generated enthusiastic support to target these receptors in a wide range of other immune related diseases. While the overwhelming focus has been on blockade of these inhibitory pathways to augment immunity, agonistic triggering via these receptors offers the promise of dampening pathogenic inflammatory responses. V-domain Ig suppressor of T cell activation (VISTA) has emerged as an immunoregulatory receptor with constitutive expression on both the T cell and myeloid compartments, and whose agonistic targeting has proven a unique avenue relative to other checkpoint pathways to suppress pathologies mediated by the innate arm of the immune system. VISTA agonistic targeting profoundly changes the phenotype of human monocytes towards an anti-inflammatory cell state, as highlighted by striking suppression of the canonical markers CD14 and Fcγr3a (CD16), and the almost complete suppression of both the interferon I (IFN-I) and antigen presentation pathways. The insights from these very recent studies highlight the impact of VISTA agonistic targeting of myeloid cells, and its potential therapeutic implications in the settings of hyperinflammatory responses such as cytokine storms, driven by dysregulated immune responses to viral infections (with a focus on COVID-19) and autoimmune diseases. Collectively, these findings suggest that the VISTA pathway plays a conserved, non-redundant role in myeloid cell function.


Asunto(s)
Antígenos B7/agonistas , COVID-19/patología , Síndrome de Liberación de Citoquinas/prevención & control , Células Mieloides/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos B7/antagonistas & inhibidores , Antígenos B7/inmunología , Linfocitos T CD4-Positivos/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/patología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Humanos , Inmunoterapia , Interferón Tipo I/antagonistas & inhibidores , Receptores de Lipopolisacáridos/antagonistas & inhibidores , Activación de Linfocitos/inmunología , Ratones , Receptores de IgG/antagonistas & inhibidores , SARS-CoV-2/inmunología
20.
Med Hypotheses ; 144: 110012, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-997289

RESUMEN

The most serious health issue today is the rapid outbreak of Coronavirus Disease 2019 (COVID-19). More than 6,973,427 confirmed cases were diagnosed in nearly 213 countries and territories around the world and two international conveyances, causing globally over 400,000 deaths. Epidemiology, risk factors, and clinical characteristics of COVID-19 patients have been identified, but the factors influencing the immune system against COVID-19 have not been well established. Upon infection or cell damage, high amounts of adenosine triphosphate (ATP) are released from damaged cells, which serve as mediators of inflammation through purinergic cell surface receptor signaling. As a protective mechanism to prevent excessive damage to host tissue, adenosine counteracts ATP's effects by adenosine receptor stimulation to suppress the pro-inflammatory response. Adenosine is seen as a major obstacle to the efficacy of immune therapies, and the adenosinergic axis components are critical therapeutic targets for cancer and microbial infections. Pharmacologic inhibitors or antibodies specific to adenosinergic pathway components or adenosine receptors in microbial and tumor therapy have shown efficacy in pre-clinical studies and are entering the clinical arena. In this review, we provide a novel hypothesis explaining the potential for improving the efficiency of innate and adaptive immune systems by targeting adenosinergic pathway components and adenosine A2A receptor signaling for the treatment of COVID-19.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Pandemias , Receptor de Adenosina A2A/fisiología , 5'-Nucleotidasa/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina Trifosfato/metabolismo , Apirasa/metabolismo , COVID-19/epidemiología , COVID-19/inmunología , COVID-19/metabolismo , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón beta/fisiología , Modelos Inmunológicos , Terapia Molecular Dirigida , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA