Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Immunol Lett ; 256-257: 34-41, 2023.
Artículo en Inglés | MEDLINE | ID: covidwho-2302009

RESUMEN

Kawasaki disease (KD), a rare multisystem inflammatory condition that predominantly affects children under six years of age, is the leading cause of childhood-acquired heart disease in developed countries. The pathogenesis is unknown, but studies support that an infectious stimulus triggers an autoimmune reaction in a genetically susceptible child. Recent studies demonstrated an association with autoantibody response to Del-1 (also known as EDIL3) in children with KD. Del-1 is an extracellular matrix protein that is expressed both in macrophages and vascular endothelium. Del-1 has an anti-inflammatory role by preventing leucocyte migration to inflammatory sites. Del-1 has two expression variants and genetic variants of Del-1 have been associated with the risk of intracranial aneurysms. Due to the physiologic plausibility for a role during KD, we chose to assess if autoantibodies against DEL-1 are seen in a larger cohort of children with KD and to assess if responses correlated to aneurysm formation. Contrary to prior findings, in comparison to febrile controls, autoantibodies were not overall higher in children with KD. Elevation in Post-IVIG samples in comparison to pre-IVIG and convalescent samples supports the commonality of anti-Del-1 antibodies. Autoantibodies were notably lower in children with KD who had coronary Z score elevations in comparison to those who did not.


Asunto(s)
Aneurisma Coronario , Síndrome Mucocutáneo Linfonodular , Niño , Humanos , Preescolar , Autoanticuerpos , Aneurisma Coronario/complicaciones , Aneurisma Coronario/prevención & control , Síndrome Mucocutáneo Linfonodular/genética , Inmunoglobulinas Intravenosas/uso terapéutico , Antiinflamatorios/uso terapéutico , Proteínas de Unión al Calcio , Moléculas de Adhesión Celular
2.
Vasa ; 51(6): 341-350, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-2096638

RESUMEN

Vascular adhesion protein-1 (VAP-1) also known as amino oxidase copper containing 3 (AOC3) is a pro-inflammatory and versatile molecule with adhesive and enzymatic properties. VAP-1 is a primary amine oxidase belonging to the semicarbazide-sensitive amine oxidase (SSAO) family, which catalyzes the oxidation of primary amines leading to the production of ammonium, formaldehyde, methylglyoxal, and hydrogen peroxide. VAP-1 is mainly expressed by endothelial cells, smooth muscle cells, adipocytes and pericytes. It is involved in a repertoire of biological functions, e.g., immune cell extravasation, angiogenesis, and vascularization. Research into VAP-1 has intensified within the last decade on its role as a novel clinical biomarker and as a potential therapeutic target of vascular inflammatory disorders such as atherosclerosis, stroke, diabetes, neurovascular disorders (e.g., Alzheimer's Disease), hepatic disease (e.g., non-alcoholic steatohepatitis), and skin conditions (e.g., psoriasis). This is the most up-to-date and comprehensive review on VAP-1 focusing on the translational aspects of VAP-1. Compared to recent reviews, our review provides novel insights on VAP-1 and heart failure, stroke and frailty, diabetes, endometriosis, osteoarthritis, COVID-19, conjunctivitis associated systemic lupus erythematosus, hematopoietic stem cells, gliomas, treatment of colorectal cancer with a novel VAP-1 inhibitor (U-V269), promoting recovery of motor functions and habit learning with a novel VAP-1 inhibitor (PXS-4681A), and 68Ga-DOTA-Siglec-9, a labelled peptide of Siglec-9 (a VAP-1 ligand), which appears to be a safe PET tracer for inflammation in rheumatoid arthritis. Finally, we present the emerging role of VAP-1 in pregnancy as a gatekeeper of immune cells, which are critical for spiral arterial remodeling, the deficiency of which could lead to vascular disorders of pregnancy such as preeclampsia. Future research should prioritize clinical trials on VAP-1 small-molecule inhibitors and monoclonal antibodies, thus, maximizing the potential of VAP-1 targeted therapy as well as research into sVAP-1 as a clinical biomarker of diseases and its prognosis.


Asunto(s)
Amina Oxidasa (conteniendo Cobre) , Aterosclerosis , COVID-19 , Diabetes Mellitus , Accidente Cerebrovascular , Femenino , Humanos , Células Endoteliales , Moléculas de Adhesión Celular/uso terapéutico , Amina Oxidasa (conteniendo Cobre)/uso terapéutico , Molécula 1 de Adhesión Celular Vascular , Biomarcadores , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/uso terapéutico
3.
Sci Rep ; 12(1): 14049, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: covidwho-1991667

RESUMEN

Even before the COVID-19 pandemic declines in life expectancy in the United States were attributed to increased mortality rates in midlife adults across racial and ethnic groups, indicating a need for markers to identify individuals at risk for early mortality. Extracellular vesicles (EVs) are small, lipid-bound vesicles capable of shuttling functional proteins, nucleic acids, and lipids. Given their role as intercellular communicators and potential biomarkers of disease, we explored whether circulating EVs may be markers of mortality in a prospective, racially, and socioeconomically diverse middle-aged cohort. We isolated plasma EVs from 76 individuals (mean age = 59.6 years) who died within a 5 year period and 76 surviving individuals matched by age, race, and poverty status. There were no significant differences in EV concentration, size, or EV-associated mitochondrial DNA levels associated with mortality. We found that several EV-associated inflammatory proteins including CCL23, CSF-1, CXCL9, GDNF, MCP-1, STAMBP, and 4E-BP1 were significantly associated with mortality. IL-10RB and CDCP1 were more likely to be present in plasma EVs from deceased individuals than in their alive counterparts. We also report differences in EV-associated inflammatory proteins with poverty status, race, and sex. Our results suggest that plasma EV-associated inflammatory proteins are promising potential clinical biomarkers of mortality.


Asunto(s)
COVID-19 , Vesículas Extracelulares , Adulto , Antígenos de Neoplasias/metabolismo , Biomarcadores , Proteínas Sanguíneas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Persona de Mediana Edad , Pandemias , Estudios Prospectivos
4.
Int J Mol Sci ; 21(15)2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1934096

RESUMEN

In physiology and pathophysiology the molecules involved in blood cell-blood cell and blood cell-endothelium interactions have been identified. Platelet aggregation and adhesion to the walls belonging to vessels involve glycoproteins (GP), GP llb and GP llla and the GP Ib-IX-V complex. Red blood cells (RBCs) in normal situations have little interaction with the endothelium. Abnormal adhesion of RBCs was first observed in sickle cell anemia involving vascular cell adhesion molecule (VCAM)-1, α4ß1, Lu/BCAM, and intercellular adhesion molecule (ICAM)-4. More recently RBC adhesion was found to be increased in retinal-vein occlusion (RVO) and in polycythemia vera (PV). The molecules which participate in this process are phosphatidylserine and annexin V in RVO, and phosphorylated Lu/BCAM and α5 laminin chain in PV. The additional adhesion in diabetes mellitus occurs due to the glycated RBC band 3 and the advanced glycation end-product receptors. The multiligand receptor binds advanced glycation end products (AGEs) or S100 calgranulins, or ß-amyloid peptide. This receptor for advanced glycation end products is known as RAGE. The binding to RAGE-activated endothelial cells leads to an inflammatory reaction and a prothrombotic state via NADPH activation and altered gene expression. RAGE blockade is a potential target for drugs preventing the deleterious consequences of RAGE activation.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Proteínas de Neoplasias/metabolismo , Policitemia Vera/metabolismo , Oclusión de la Vena Retiniana/metabolismo , Adhesión Celular , Células Endoteliales/patología , Eritrocitos/patología , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Policitemia Vera/patología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Oclusión de la Vena Retiniana/patología , Trombosis/metabolismo , Trombosis/patología
5.
Sci Rep ; 12(1): 9583, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1921701

RESUMEN

TACSTD2 encodes a transmembrane glycoprotein Trop2 commonly overexpressed in carcinomas. While the Trop2 protein was discovered already in 1981 and first antibody-drug conjugate targeting Trop2 were recently approved for cancer therapy, the physiological role of Trop2 is still not fully understood. In this article, we show that TACSTD2/Trop2 expression is evolutionarily conserved in lungs of various vertebrates. By analysis of publicly available transcriptomic data we demonstrate that TACSTD2 level consistently increases in lungs infected with miscellaneous, but mainly viral pathogens. Single cell and subpopulation based transcriptomic data revealed that the major source of TACSTD2 transcript are lung epithelial cells and their progenitors and that TACSTD2 is induced directly in lung epithelial cells following infection. Increase in TACSTD2 expression may represent a mechanism to maintain/restore epithelial barrier function and contribute to regeneration process in infected/damaged lungs.


Asunto(s)
Antígenos de Neoplasias , Moléculas de Adhesión Celular , Animales , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/metabolismo , Pulmón/metabolismo , Regulación hacia Arriba
6.
Comput Biol Med ; 146: 105537, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1899652

RESUMEN

Recently a novel coactivator, Leupaxin (LPXN), has been reported to interact with Androgen receptor (AR) and play a significant role in the invasion and progression of prostate cancer. The interaction between AR and LPXN occurs in a ligand-dependent manner and has been reported that the LIM domain in the Leupaxin interacts with the LDB (ligand-binding domain) domain AR. However, no detailed study is available on how the LPXN interacts with AR and increases the (prostate cancer) PCa progression. Considering the importance of the novel co-activator, LPXN, the current study also uses state-of-the-art methods to provide atomic-level insights into the binding of AR and LPXN and the impact of the most frequent clinical mutations H874Y, T877A, and T877S on the binding and function of LPXN. Protein coupling analysis revealed that the three mutants favour the robust binding of LPXN than the wild type by altering the hydrogen bonding network. Further understanding of the binding variations was explored through dissociation constant prediction which demonstrated similar reports as the docking results. A molecular simulation approaches further revealed the dynamic features which reported variations in the dynamics stability, protein packing, hydrogen bonding network, and residues flexibility index. Furthermore, we also assessed the protein motion and free energy landscape which also demonstrated variations in the internal dynamics. The binding free energy calculation revealed -32.95 ± 0.17 kcal/mol for the wild type, for H874Y the total binding energy (BFE) was -36.69 ± 0.11 kcal/mol, for T877A the BFE was calculated to be -38.78 ± 0.17 kcal/mol while for T877S the BFE -41.16 ± 0.12 kcal/mol. This shows that the binding of LPXN is increased by these mutations which consequently increase the PCa invasion and motility. In conclusion, the current study helps in understanding the protein networks and particular the coupling of AR-LPXN in prostate cancer and is of great interest in deciphering the molecular mechanism of disease and therapeutics developments.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Humanos , Ligandos , Masculino , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo
7.
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
8.
Physiol Rep ; 10(8): e15271, 2022 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1791646

RESUMEN

Acute respiratory distress syndrome (ARDS) is a major clinical problem without available therapies. Known risks for ARDS include severe sepsis, SARS-CoV-2, gram-negative bacteria, trauma, pancreatitis, and blood transfusion. During ARDS, blood fluids and inflammatory cells enter the alveoli, preventing oxygen exchange from air into blood vessels. Reduced pulmonary endothelial barrier function, resulting in leakage of plasma from blood vessels, is one of the major determinants in ARDS. It is, however, unknown why systemic inflammation particularly targets the pulmonary endothelium, as endothelial cells (ECs) line all vessels in the vascular system of the body. In this study, we examined ECs of pulmonary, umbilical, renal, pancreatic, and cardiac origin for upregulation of adhesion molecules, ability to facilitate neutrophil (PMN) trans-endothelial migration (TEM) and for endothelial barrier function, in response to the gram-negative bacterial endotoxin LPS. Interestingly, we found that upon LPS stimulation, pulmonary ECs showed increased levels of adhesion molecules, facilitated more PMN-TEM and significantly perturbed the endothelial barrier, compared to other types of ECs. These observations could partly be explained by a higher expression of the adhesion molecule ICAM-1 on the pulmonary endothelial surface compared to other ECs. Moreover, we identified an increased expression of Cadherin-13 in pulmonary ECs, for which we demonstrated that it aids PMN-TEM in pulmonary ECs stimulated with LPS. We conclude that pulmonary ECs are uniquely sensitive to LPS, and intrinsically different, compared to ECs from other vascular beds. This may add to our understanding of the development of ARDS upon systemic inflammation.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , SARS-CoV-2
9.
Nature ; 607(7917): 97-103, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1730298

RESUMEN

Critical COVID-19 is caused by immune-mediated inflammatory lung injury. Host genetic variation influences the development of illness requiring critical care1 or hospitalization2-4 after infection with SARS-CoV-2. The GenOMICC (Genetics of Mortality in Critical Care) study enables the comparison of genomes from individuals who are critically ill with those of population controls to find underlying disease mechanisms. Here we use whole-genome sequencing in 7,491 critically ill individuals compared with 48,400 controls to discover and replicate 23 independent variants that significantly predispose to critical COVID-19. We identify 16 new independent associations, including variants within genes that are involved in interferon signalling (IL10RB and PLSCR1), leucocyte differentiation (BCL11A) and blood-type antigen secretor status (FUT2). Using transcriptome-wide association and colocalization to infer the effect of gene expression on disease severity, we find evidence that implicates multiple genes-including reduced expression of a membrane flippase (ATP11A), and increased expression of a mucin (MUC1)-in critical disease. Mendelian randomization provides evidence in support of causal roles for myeloid cell adhesion molecules (SELE, ICAM5 and CD209) and the coagulation factor F8, all of which are potentially druggable targets. Our results are broadly consistent with a multi-component model of COVID-19 pathophysiology, in which at least two distinct mechanisms can predispose to life-threatening disease: failure to control viral replication; or an enhanced tendency towards pulmonary inflammation and intravascular coagulation. We show that comparison between cases of critical illness and population controls is highly efficient for the detection of therapeutically relevant mechanisms of disease.


Asunto(s)
COVID-19 , Enfermedad Crítica , Genoma Humano , Interacciones Huésped-Patógeno , Secuenciación Completa del Genoma , Transportadoras de Casetes de Unión a ATP , COVID-19/genética , COVID-19/mortalidad , COVID-19/patología , COVID-19/virología , Moléculas de Adhesión Celular , Cuidados Críticos , Enfermedad Crítica/mortalidad , Selectina E , Factor VIII , Fucosiltransferasas , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno/genética , Humanos , Subunidad beta del Receptor de Interleucina-10 , Lectinas Tipo C , Mucina-1 , Proteínas del Tejido Nervioso , Proteínas de Transferencia de Fosfolípidos , Receptores de Superficie Celular , Proteínas Represoras , SARS-CoV-2/patogenicidad
10.
Oxid Med Cell Longev ; 2022: 1630918, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-1714452

RESUMEN

BACKGROUND: The impairment of microcirculation is associated with the unfavorable outcome for extracorporeal membrane oxygenation (ECMO) patients. Studies revealed that pulsatile modification improves hemodynamics and attenuates inflammation during ECMO support. However, whether flow pattern impacts microcirculation and endothelial integrity is rarely documented. The objective of this work was to explore how pulsatility affects microcirculation during ECMO. METHODS: Canine animal models with cardiac arrest were supported by ECMO, with the i-Cor system used to generate nonpulsatile or pulsatile flow. The sublingual microcirculation parameters were examined using the CytoCam microscope system. The expression of hsa_circ_0007367, a circular RNA, was measured during ECMO support. In vitro validation was performed in pulmonary vascular endothelial cells (PMVECs) exposed to pulsatile or nonpulsatile flow, and the expressions of hsa_circ_0007367, endothelial tight junction markers, endothelial adhesive molecules, endothelial nitric oxide synthases (eNOS), and NF-κB signaling activity were analyzed. RESULTS: The pulsatile modification of ECMO enhanced microcirculatory perfusion, attenuated pulmonary inflammation, and stabilized endothelial integrity in animal models; meanwhile, the expression of hsa_circ_0007367 was significantly upregulated both in animals and PMVECs exposed to pulsatile flow. In particular, upregulation of hsa_circ_0007367 stabilized the expressions of endothelial tight junction markers zonula occludens- (ZO-) 1 and occludin, followed by modulating the endothelial nitric oxide synthases (eNOS) activity and inhibiting the NF-κB signaling pathway. CONCLUSION: The modification of pulsatility contributes to microcirculatory perfusion and endothelial integrity during ECMO. The expression of hsa_circ_0007367 plays a pivotal role in this protective mechanism.


Asunto(s)
Ácidos Nucleicos Libres de Células/genética , Células Endoteliales/fisiología , Oxigenación por Membrana Extracorpórea/métodos , Paro Cardíaco/terapia , Animales , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Perros , Células Endoteliales/metabolismo , Paro Cardíaco/genética , Paro Cardíaco/patología , Paro Cardíaco/fisiopatología , Inflamación , Pulmón/irrigación sanguínea , Pulmón/patología , Microcirculación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ocludina/genética , Ocludina/metabolismo , Flujo Pulsátil , Ratas , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
11.
Arthritis Rheumatol ; 74(7): 1132-1138, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1694821

RESUMEN

OBJECTIVE: While endothelial dysfunction has been implicated in the widespread thromboinflammatory complications of COVID-19, the upstream mediators of endotheliopathy remain, for the most part, unknown. This study was undertaken to identify circulating factors contributing to endothelial cell activation and dysfunction in COVID-19. METHODS: Human endothelial cells were cultured in the presence of serum or plasma from 244 patients hospitalized with COVID-19 and plasma from 100 patients with non-COVID-19-related sepsis. Cell adhesion molecules (E-selectin, vascular cell adhesion molecule 1, and intercellular adhesion molecule 1 [ICAM-1]) were quantified using in-cell enzyme-linked immunosorbent assay. RESULTS: Serum and plasma from COVID-19 patients increased surface expression of cell adhesion molecules. Furthermore, levels of soluble ICAM-1 and E-selectin were elevated in patient serum and correlated with disease severity. The presence of circulating antiphospholipid antibodies was a strong marker of the ability of COVID-19 serum to activate endothelium. Depletion of total IgG from antiphospholipid antibody-positive serum markedly reduced the up-regulation of cell adhesion molecules. Conversely, supplementation of control serum with patient IgG was sufficient to trigger endothelial activation. CONCLUSION: These data are the first to indicate that some COVID-19 patients have potentially diverse antibodies that drive endotheliopathy, providing important context regarding thromboinflammatory effects of autoantibodies in severe COVID-19.


Asunto(s)
Anticuerpos Antifosfolípidos , COVID-19 , Células Endoteliales , Anticuerpos Antifosfolípidos/inmunología , COVID-19/inmunología , Moléculas de Adhesión Celular/metabolismo , Selectina E , Células Endoteliales/metabolismo , Endotelio Vascular , Humanos , Inmunoglobulina G/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Aging (Albany NY) ; 14(4): 1597-1610, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: covidwho-1689674

RESUMEN

BACKGROUND: COVID-19 survivors report residual lung abnormalities after discharge from the hospital. The aim of this study was to identify biomarkers in serum and induced sputum samples from patients after hospitalization for COVID-19. METHODS: Patients admitted to hospitals in Spain with laboratory-confirmed COVID-19 were recruited for this study. SARS-CoV-2-infected patients were divided into groups with mild/moderate and severe disease according to the severity of their symptoms during hospitalization. Levels of 92 biomarkers were measured in serum and induced sputum samples. RESULTS: A total of 108 patients (46.2% severe cases) were included in this study. The median number of days after the onset of symptoms was 104. A significant difference was observed in diffusing capacity for carbon monoxide (DLCO), an indicator of lung function, whereby DLCO <80% was significantly lower in severe cases (p <0.001). Differences in inflammatory biomarkers were observed between patients with mild/moderate and severe disease. For some biomarkers, correlations in serum and induced sputum levels were detected. Independent predictors of severe disease were DLCO <80% and the serum CDCP1 value. CONCLUSIONS: Higher levels of CDCP1 remain after hospital discharge and are associated with the severity of COVID-19. The possible prognostic implications warrant further investigation.


Asunto(s)
Antígenos de Neoplasias/sangre , COVID-19/sangre , Moléculas de Adhesión Celular/sangre , Antígenos de Neoplasias/análisis , Biomarcadores/sangre , Moléculas de Adhesión Celular/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Esputo/química
13.
Elife ; 112022 01 25.
Artículo en Inglés | MEDLINE | ID: covidwho-1662829

RESUMEN

The human proteome is replete with short linear motifs (SLiMs) of four to six residues that are critical for protein-protein interactions, yet the importance of the sequence surrounding such motifs is underexplored. We devised a proteomic screen to examine the influence of SLiM sequence context on protein-protein interactions. Focusing on the EVH1 domain of human ENAH, an actin regulator that is highly expressed in invasive cancers, we screened 36-residue proteome-derived peptides and discovered new interaction partners of ENAH and diverse mechanisms by which context influences binding. A pocket on the ENAH EVH1 domain that has diverged from other Ena/VASP paralogs recognizes extended SLiMs and favors motif-flanking proline residues. Many high-affinity ENAH binders that contain two proline-rich SLiMs use a noncanonical site on the EVH1 domain for binding and display a thermodynamic signature consistent with the two-motif chain engaging a single domain. We also found that photoreceptor cilium actin regulator (PCARE) uses an extended 23-residue region to obtain a higher affinity than any known ENAH EVH1-binding motif. Our screen provides a way to uncover the effects of proteomic context on motif-mediated binding, revealing diverse mechanisms of control over EVH1 interactions and establishing that SLiMs can't be fully understood outside of their native context.


Asunto(s)
Actinas/metabolismo , Sitios de Unión , Proteínas de Unión al ADN/metabolismo , Proteínas de Microfilamentos/metabolismo , Prolina/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células HEK293 , Humanos , Proteómica
14.
J Leukoc Biol ; 111(6): 1287-1295, 2022 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1650087

RESUMEN

Immune cell dysregulation and lymphopenia characterize COVID-19 pathology in moderate to severe disease. While underlying inflammatory factors have been extensively studied, homeostatic and mucosal migratory signatures remain largely unexplored as causative factors. In this study, we evaluated the association of circulating IL-6, soluble mucosal addressin cell adhesion molecule (sMAdCAM), and IL-15 with cellular dysfunction characterizing mild and hypoxemic stages of COVID-19. A cohort of SARS-CoV-2 infected individuals (n = 130) at various stages of disease progression together with healthy controls (n = 16) were recruited from COVID Care Centres (CCCs) across Mumbai, India. Multiparametric flow cytometry was used to perform in-depth immune subset characterization and to measure plasma IL-6 levels. sMAdCAM, IL-15 levels were quantified using ELISA. Distinct depletion profiles, with relative sparing of CD8 effector memory and CD4+ regulatory T cells, were observed in hypoxemic disease within the lymphocyte compartment. An apparent increase in the frequency of intermediate monocytes characterized both mild as well as hypoxemic disease. IL-6 levels inversely correlated with those of sMAdCAM and both markers showed converse associations with observed lympho-depletion suggesting opposing roles in pathogenesis. Interestingly, IL-15, a key cytokine involved in lymphocyte activation and homeostasis, was detected in symptomatic individuals but not in healthy controls or asymptomatic cases. Further, plasma IL-15 levels negatively correlated with T, B, and NK count suggesting a compensatory production of this cytokine in response to the profound lymphopenia. Finally, higher levels of plasma IL-15 and IL-6, but not sMAdCAM, were associated with a longer duration of hospitalization.


Asunto(s)
COVID-19 , Interleucina-15/sangre , Linfopenia , Linfocitos T CD8-positivos , Moléculas de Adhesión Celular , Citocinas , Humanos , Interleucina-6 , Linfopenia/etiología , SARS-CoV-2
15.
J Med Chem ; 64(19): 14332-14343, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1621195

RESUMEN

In addition to a variety of viral-glycoprotein receptors (e.g., heparan sulfate, Niemann-Pick C1, etc.), dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), from the C-type lectin receptor family, plays one of the most important pathogenic functions for a wide range of viruses (e.g., Ebola, human cytomegalovirus (HCMV), HIV-1, severe acute respiratory syndrome coronavirus 2, etc.) that invade host cells before replication; thus, its inhibition represents a relevant extracellular antiviral therapy. We report two novel p-tBu-calixarene glycoclusters 1 and 2, bearing tetrahydroxamic acid groups, which exhibit micromolar inhibition of soluble DC-SIGN binding and provide nanomolar IC50 inhibition of both DC-SIGN-dependent Jurkat cis-cell infection by viral particle pseudotyped with Ebola virus glycoprotein and the HCMV-gB-recombinant glycoprotein interaction with monocyte-derived dendritic cells expressing DC-SIGN. A unique cooperative involvement of sugar, linker, and calixarene core is likely behind the strong avidity of DC-SIGN for these low-valent systems. We claim herein new promising candidates for the rational development of a large spectrum of antiviral therapeutics.


Asunto(s)
Calixarenos/química , Moléculas de Adhesión Celular/antagonistas & inhibidores , Glicoconjugados/metabolismo , Glicoproteínas/antagonistas & inhibidores , Ácidos Hidroxámicos/química , Lectinas Tipo C/antagonistas & inhibidores , Fenoles/química , Receptores de Superficie Celular/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Citomegalovirus/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Ebolavirus/fisiología , Glicoconjugados/química , Glicoconjugados/farmacología , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Células Jurkat , Lectinas Tipo C/metabolismo , Modelos Biológicos , Unión Proteica , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Virales/genética , Proteínas Virales/metabolismo
16.
Cells ; 10(12)2021 11 23.
Artículo en Inglés | MEDLINE | ID: covidwho-1538383

RESUMEN

Dendritic cells (DCs) are the most potent antigen-presenting cells, and their function is essential to configure adaptative immunity and avoid excessive inflammation. DCs are predicted to play a crucial role in the clinical evolution of the infection by the severe acute respiratory syndrome (SARS) coronavirus (CoV)-2. DCs interaction with the SARS-CoV-2 Spike protein, which mediates cell receptor binding and subsequent fusion of the viral particle with host cell, is a key step to induce effective immunity against this virus and in the S protein-based vaccination protocols. Here we evaluated human DCs in response to SARS-CoV-2 S protein, or to a fragment encompassing the receptor binding domain (RBD) challenge. Both proteins increased the expression of maturation markers, including MHC molecules and costimulatory receptors. DCs interaction with the SARS-CoV-2 S protein promotes activation of key signaling molecules involved in inflammation, including MAPK, AKT, STAT1, and NFκB, which correlates with the expression and secretion of distinctive proinflammatory cytokines. Differences in the expression of ACE2 along the differentiation of human monocytes to mature DCs and inter-donor were found. Our results show that SARS-CoV-2 S protein promotes inflammatory response and provides molecular links between individual variations and the degree of response against this virus.


Asunto(s)
Células Dendríticas/patología , Células Dendríticas/virología , Receptores Virales/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Citocinas/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inflamación/patología , Lectinas Tipo C/metabolismo , Dominios Proteicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Donantes de Tejidos
17.
Viruses ; 13(11)2021 11 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1502528

RESUMEN

Men are disproportionately affected by the coronavirus disease-2019 (COVID-19), and face higher odds of severe illness and death compared to women. The vascular effects of androgen signaling and inflammatory cytokines in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-mediated endothelial injury are not defined. We determined the effects of SARS-CoV-2 spike protein-mediated endothelial injury under conditions of exposure to androgen dihydrotestosterone (DHT) and tumor necrosis factor-a (TNF-α) and tested potentially therapeutic effects of mineralocorticoid receptor antagonism by spironolactone. Circulating endothelial injury markers VCAM-1 and E-selectin were measured in men and women diagnosed with COVID-19. Exposure of endothelial cells (ECs) in vitro to DHT exacerbated spike protein S1-mediated endothelial injury transcripts for the cell adhesion molecules E-selectin, VCAM-1 and ICAM-1 and anti-fibrinolytic PAI-1 (p < 0.05), and increased THP-1 monocyte adhesion to ECs (p = 0.032). Spironolactone dramatically reduced DHT+S1-induced endothelial activation. TNF-α exacerbated S1-induced EC activation, which was abrogated by pretreatment with spironolactone. Analysis from patients hospitalized with COVID-19 showed concordant higher circulating VCAM-1 and E-Selectin levels in men, compared to women. A beneficial effect of the FDA-approved drug spironolactone was observed on endothelial cells in vitro, supporting a rationale for further evaluation of mineralocorticoid antagonism as an adjunct treatment in COVID-19.


Asunto(s)
COVID-19/patología , Dihidrotestosterona/farmacología , Endotelio Vascular/patología , Inflamación , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/fisiología , Espironolactona/farmacología , Antagonistas de Receptores de Angiotensina/farmacología , COVID-19/fisiopatología , COVID-19/virología , Moléculas de Adhesión Celular/sangre , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Caracteres Sexuales , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología , Valsartán/farmacología
18.
J Clin Invest ; 131(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1495789

RESUMEN

To explore how the immune system controls clearance of SARS-CoV-2, we used a single-cell, mass cytometry-based proteomics platform to profile the immune systems of 21 patients who had recovered from SARS-CoV-2 infection without need for admission to an intensive care unit or for mechanical ventilation. We focused on receptors involved in interactions between immune cells and virus-infected cells. We found that the diversity of receptor repertoires on natural killer (NK) cells was negatively correlated with the viral clearance rate. In addition, NK subsets expressing the receptor DNAM1 were increased in patients who more rapidly recovered from infection. Ex vivo functional studies revealed that NK subpopulations with high DNAM1 expression had cytolytic activities in response to target cell stimulation. We also found that SARS-CoV-2 infection induced the expression of CD155 and nectin-4, ligands of DNAM1 and its paired coinhibitory receptor TIGIT, which counterbalanced the cytolytic activities of NK cells. Collectively, our results link the cytolytic immune responses of NK cells to the clearance of SARS-CoV-2 and show that the DNAM1 pathway modulates host-pathogen interactions during SARS-CoV-2 infection.


Asunto(s)
COVID-19/inmunología , COVID-19/virología , Células Asesinas Naturales/inmunología , Receptores de Células Asesinas Naturales/inmunología , SARS-CoV-2/inmunología , Adolescente , Adulto , Anciano , Animales , Antígenos de Diferenciación de Linfocitos T/inmunología , Moléculas de Adhesión Celular/inmunología , Estudios de Cohortes , Citotoxicidad Inmunológica , Femenino , Xenoinjertos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunofenotipificación , Técnicas In Vitro , Ligandos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Subfamília D de Receptores Similares a Lectina de las Células NK/inmunología , Pandemias , Receptores Inmunológicos/inmunología , Receptores Virales/inmunología , Carga Viral , Adulto Joven
19.
Biomolecules ; 11(11)2021 10 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1488476

RESUMEN

Glycosylation is an important post-translational modification that affects a wide variety of physiological functions. DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) is a protein expressed in antigen-presenting cells that recognizes a variety of glycan epitopes. Until now, the binding of DC-SIGN to SARS-CoV-2 Spike glycoprotein has been reported in various articles and is regarded to be a factor in systemic infection and cytokine storm. The mechanism of DC-SIGN recognition offers an alternative method for discovering new medication for COVID-19 treatment. Here, we discovered three potential pockets that hold different glycan epitopes by performing molecular dynamics simulations of previously reported oligosaccharides. The "EPN" motif, "NDD" motif, and Glu354 form the most critical pocket, which is known as the Core site. We proposed that the type of glycan epitopes, rather than the precise amino acid sequence, determines the recognition. Furthermore, we deduced that oligosaccharides could occupy an additional site, which adds to their higher affinity than monosaccharides. Based on our findings and previously described glycoforms on the SARS-CoV-2 Spike, we predicted the potential glycan epitopes for DC-SIGN. It suggested that glycan epitopes could be recognized at multiple sites, not just Asn234, Asn149 and Asn343. Subsequently, we found that Saikosaponin A and Liquiritin, two plant glycosides, were promising DC-SIGN antagonists in silico.


Asunto(s)
COVID-19/inmunología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Epítopos/química , Glicósidos/química , Lectinas Tipo C/antagonistas & inhibidores , Polisacáridos/química , Receptores de Superficie Celular/antagonistas & inhibidores , Secuencias de Aminoácidos , Sitios de Unión , COVID-19/metabolismo , Simulación por Computador , Citocinas/metabolismo , Flavanonas/química , Glucósidos/química , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Monosacáridos/química , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/química , Saponinas/química , Glicoproteína de la Espiga del Coronavirus/química
20.
Biol Direct ; 16(1): 18, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: covidwho-1477451

RESUMEN

Skeletal muscle has an extraordinary regenerative capacity reflecting the rapid activation and effective differentiation of muscle stem cells (MuSCs). In the course of muscle regeneration, MuSCs are reprogrammed by immune cells. In turn, MuSCs confer immune cells anti-inflammatory properties to resolve inflammation and facilitate tissue repair. Indeed, MuSCs can exert therapeutic effects on various degenerative and inflammatory disorders based on their immunoregulatory ability, including effects primed by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). At the molecular level, the tryptophan metabolites, kynurenine or kynurenic acid, produced by indoleamine 2,3-dioxygenase (IDO), augment the expression of TNF-stimulated gene 6 (TSG6) through the activation of the aryl hydrocarbon receptor (AHR). In addition, insulin growth factor 2 (IGF2) produced by MuSCs can endow maturing macrophages oxidative phosphorylation (OXPHOS)-dependent anti-inflammatory functions. Herein, we summarize the current understanding of the immunomodulatory characteristics of MuSCs and the issues related to their potential applications in pathological conditions, including COVID-19.


Asunto(s)
COVID-19/terapia , Sistema Inmunológico/fisiología , Músculos/fisiología , Regeneración/fisiología , Células Madre/citología , Animales , COVID-19/inmunología , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Inflamación , Factor II del Crecimiento Similar a la Insulina/metabolismo , Interferón gamma/metabolismo , Ácido Quinurénico/metabolismo , Quinurenina/metabolismo , Macrófagos/metabolismo , Ratones , Músculos/metabolismo , Fosforilación Oxidativa , Receptores de Hidrocarburo de Aril/metabolismo , Triptófano/química , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA