Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-38819519

ABSTRACT

PURPOSE: Since the onset of the COVID-19 pandemic, most research has focused on the management of the acute symptoms of the disease. Yet some people tend to experience symptoms beyond the acute phase, defined as Post-COVID-19 Condition (PCC). This study aims to assess the impact of COVID-19 and PCC on anxiety and depression. METHODS: This is a prospective longitudinal cohort study among the Belgian adult population with recent SARS-CoV-2 infection for which contact tracing was initiated. A total of 3127 people were followed-up just after their infection and three months later (from April 2021 to January 2022). Anxiety and depression were assessed at the two stages using the GAD-7 (Generalized Anxiety Disorder) and the PHQ-9 (Patient Health Questionnaire). RESULTS: Three months after infection, participants with PCC (50%) had an increased probability of having both anxiety and depressive symptoms (p < 0.001). The proportion with anxiety and depressive symptoms at three months were significantly higher in people with PCC (11% and 19%) compared to people without persistent COVID symptoms (3.8% and 4.2%) and to a matched sub-sample not infected with SARS-CoV-2 (6.5% and 4.3%). Having at least one acute COVID-19 symptom (p < 0.001), experiencing financial loss following the infection (p < 0.001), and different PCC symptoms were associated with anxiety and depressive symptoms worsening over time. CONCLUSIONS: This study showed that three months after a SARS-CoV-2 infection, one in two people suffer from PCC with significant consequences for their mental health. Follow-up on mental health must therefore have an important place in people suffering from PCC.

2.
J Clin Immunol ; 39(5): 486-493, 2019 07.
Article in English | MEDLINE | ID: mdl-31154555

ABSTRACT

PURPOSE: The ubiquitous calcium-independent phospholipase A2 enzyme (iPLA2) is inhibited by calmodulin binding and known to be responsible for phospholipid remodeling housekeeping functions including granule exocytosis-associated membrane fusion in normal human neutrophils. We evaluate in human neutrophils the iPLA2 secretagogue effects using normal neutrophils, where reactive oxygen species (ROS) generation has been blocked by diphenyleneiodonium, as well as in neutrophils from chronic granulomatous disease (CGD) patients. METHODS: Neutrophils were pretreated with W7, a calmodulin inhibitor known to activate iPLA2 and exocytosis of granules, and vesicles as well as intra- and extra-microbicidal activity against Staphylococcus aureus and Aspergillus fumigatus were evaluated. RESULTS: W7 increases exocytosis of primary, secondary, and tertiary granules and vesicles and improves neutrophil microbicidal activity against S. aureus and A. fumigatus. CONCLUSIONS: In neutrophils, calmodulin-mediated iPLA2 inhibition controls granule and vesicle exocytosis in the phagosome and in the extracellular microenvironment. Relieving iPLA2 inhibition results in increased exocytosis of primary, secondary, and tertiary granules and secretory vesicles with correction of defective intracellular and extracellular microbicidal activity. In CGD patients presenting ROS defective production, this increase in the non-oxidative killing pathway partially corrects their microbicidal defects.


Subject(s)
Neutrophils/physiology , Phospholipases A2, Calcium-Independent/metabolism , Reactive Oxygen Species/metabolism , Aspergillus fumigatus , Exocytosis/drug effects , Female , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/metabolism , Humans , Male , Onium Compounds/pharmacology , Phagocytosis , Staphylococcus aureus , Sulfonamides/pharmacology
3.
Oncoimmunology ; 6(5): e1314425, 2017.
Article in English | MEDLINE | ID: mdl-28638744

ABSTRACT

The demethylating agent 5-azacytidine (AZA) has proven its efficacy in the treatment of myelodysplastic syndrome and acute myeloid leukemia. In addition, AZA can demethylate FOXP3 intron 1 (FOXP3i1) leading to the generation of regulatory T cells (Treg). Here, we investigated the impact of AZA on xenogeneic graft-vs.-host disease (xGVHD) and graft-vs.-leukemia effects in a humanized murine model of transplantation (human PBMCs-infused NSG mice), and described the impact of the drug on human T cells in vivo. We observed that AZA improved both survival and xGVHD scores. Further, AZA significantly decreased human T-cell proliferation as well as IFNγ and TNF-α serum levels, and reduced the expression of GRANZYME B and PERFORIN 1 by cytotoxic T cells. In addition, AZA significantly increased Treg frequency through hypomethylation of FOXP3i1 as well as increased Treg proliferation. The latter was subsequent to higher STAT5 signaling in Treg from AZA-treated mice, which resulted from higher IL-2 secretion by conventional T cells from AZA-treated mice itself secondary to demethylation of the IL-2 gene promoter by AZA. Importantly, Tregs harvested from AZA-treated mice were suppressive and stable over time since they persisted at high frequency in secondary transplant experiments. Finally, graft-vs.-leukemia effects (assessed by growth inhibition of THP-1 cells, transfected to express the luciferase gene) were not abrogated by AZA. In summary, our data demonstrate that AZA prevents xGVHD without abrogating graft-vs.-leukemia effects. These findings could serve as basis for further studies of GVHD prevention by AZA in acute myeloid leukemia patients offered an allogeneic transplantation.

4.
J Commun Disord ; 45(2): 111-20, 2012.
Article in English | MEDLINE | ID: mdl-22192635

ABSTRACT

UNLABELLED: Several authors mentioned the occurrence of articulation problems in the neurofibromatosis type 1 (NF1) population. However, few studies have undertaken a detailed analysis of the articulation skills of NF1 patients, especially in schoolchildren and adults. Therefore, the aim of the present study was to examine in depth the articulation skills of NF1 schoolchildren and adults, both phonetically and phonologically. Speech samples were collected from 43 Flemish NF1 patients (14 children and 29 adults), ranging in age between 7 and 53 years, using a standardized speech test in which all Flemish single speech sounds and most clusters occur in all their permissible syllable positions. Analyses concentrated on consonants only and included a phonetic inventory, a phonetic, and a phonological analysis. It was shown that phonetic inventories were incomplete in 16.28% (7/43) of participants, in which totally correct realizations of the sibilants /ʃ/ and/or /ʒ/ were missing. Phonetic analysis revealed that distortions were the predominant phonetic error type. Sigmatismus stridens, multiple ad- or interdentality, and, in children, rhotacismus non vibrans were frequently observed. From a phonological perspective, the most common error types were substitution and syllable structure errors. Particularly, devoicing, cluster simplification, and, in children, deletion of the final consonant of words were perceived. Further, it was demonstrated that significantly more men than women presented with an incomplete phonetic inventory, and that girls tended to display more articulation errors than boys. Additionally, children exhibited significantly more articulation errors than adults, suggesting that although the articulation skills of NF1 patients evolve positively with age, articulation problems do not resolve completely from childhood to adulthood. As such, the articulation errors made by NF1 adults may be regarded as residual articulation disorders. It can be concluded that the speech of NF1 patients is characterized by mild articulation disorders at an age where this is no longer expected. LEARNING OUTCOMES: Readers will be able to describe neurofibromatosis type 1 (NF1) and explain the articulation errors displayed by schoolchildren and adults with this genetic syndrome.


Subject(s)
Articulation Disorders/etiology , Neurofibromatosis 1/complications , Adolescent , Adult , Age Factors , Child , Female , Humans , Male , Middle Aged , Phonetics , Speech , Young Adult
5.
Cell Signal ; 22(10): 1437-47, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20566383

ABSTRACT

The G-protein coupled receptor (GPCR) fMLP receptor (FPR) and the two receptors tyrosine kinase (RTK), the nerve growth factor (NGF) receptor TrkA and the epidermal growth factor (EGF) receptor (EGFR) are involved in reactive oxygen species (ROS), matrix metalloproteinase-9 (MMP-9) production and CD11b membrane integrin upregulation. We show that in monocytes the three receptors crosstalk each other to modulate these pro-inflammatory mediators. Tyrphostin AG1478, the EGFR inhibitor, inhibits fMLP and NGF-associated ROS production, fMLP-associated CD11b upregulation and NGF-induced TrkA phosphorylation; K252a, the NGF receptor inhibitor, inhibits fMLP or EGF-associated ROS production, CD11b expression and EGF-induced EGFR phosphorylation; cyclosporine H, the FPR inhibitor inhibits EGF or NGF-associated ROS production, EGF-associated CD11b upregulation and prevents EGFR and TrkA phosphorylation by their respective ligand EGF and NGF. In response to fMLP, TrkA phosphorylation is inhibited by the EGFR inhibitor while EGFR phosphorylation is inhibited by the TrkA inhibitor. Receptor crosstalks are Src and ERK dependent. Down-regulation of each receptor by specific siRNA suppresses the ability of the two other receptors to promote ligand-mediated ERK phosphorylation and pro-inflammatory activities including ROS, MMP-9 production and CD11b upregulation. Thus, in monocytes GPCR ligands' activity involves activation of RTK while RTK-ligands activity engages GPCR-signalling molecules.


Subject(s)
ErbB Receptors/metabolism , Monocytes/metabolism , Receptor Cross-Talk , Receptor, trkA/metabolism , Receptors, Formyl Peptide/metabolism , CD11b Antigen/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Humans , Hydrogen Peroxide/metabolism , Monocytes/enzymology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Nerve Growth Factor/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/genetics , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/genetics
6.
Blood ; 107(6): 2525-30, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16317100

ABSTRACT

Thrombopoietin (TPO), the major growth factor for cells of the megakaryocytic lineage, is removed from circulation by binding to c-mpl receptors present on platelets and megakaryocytes. We studied patients with acute lymphoblastic leukemia (ALL) or acute myeloblastic leukemia (AML) and used TPO-induced c-fos protein up-regulation as a marker of c-mpl functionality and observed that c-mpl-presenting blast cells were present in 62% (37 of 60) of patients with ALL but that c-mpl was nonfunctional in 0 of 28 patients and that they were present in 56% (22 of 39) of patients with AML and were functional in 43% (12 of 28). Adequate increases in serum TPO level in response to thrombocytopenia were seen in patients with ALL and with c-mpl-deficient (c-mpl-) AML. In contrast, in patients with c-mpl-proficient (c-mpl+) AML, TPO levels were found to be inappropriately low but increased to expected values during induction chemotherapy as blasts disappeared. In vitro significant TPO-associated blast cell proliferation or decreased apoptosis was observed only in patients with c-mpl+ AML compared with ALL or c-mpl- AML and was highly correlated with low in vivo TPO levels (P < .001). These data suggest that, in patients with AML, inadequate TPO levels are secondary to TPO clearing by functional c-mpl receptor myeloid blast cells and that TPO may serve as an in vivo myeloid leukemic growth factor in a significant number of patients.


Subject(s)
Blast Crisis/pathology , Leukemia, Myeloid, Acute/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thrombopoietin/blood , Acute Disease , Apoptosis , Blast Crisis/blood , Cell Proliferation , Growth Substances/blood , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/physiology , Receptors, Cytokine/analysis , Receptors, Cytokine/physiology , Receptors, Thrombopoietin , Thrombocytopenia/blood
7.
J Immunol ; 175(6): 4091-102, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148159

ABSTRACT

The neuropeptide pituitary adenylate cyclase-activating protein (PACAP) acts via the G protein-coupled receptor vasoactive intestinal peptide/PACAP receptor-1 to induce phospholipase C/calcium and MAPK-dependent proinflammatory activities in human polymorphonuclear neutrophils (PMNs). In this study, we evaluate other mechanisms that regulate PACAP-evoked calcium transients, the nature of the calcium sources, and the role of calcium in proinflammatory activities. Reduction in the activity of PMNs to respond to PACAP was observed after cell exposure to inhibitors of the cAMP/protein kinase A, protein kinase C, and PI3K pathways, to pertussis toxin, genistein, and after chelation of intracellular calcium or after extracellular calcium depletion. Mobilization of intracellular calcium stores was based on the fact that PACAP-associated calcium transient was decreased after exposure to 1) thapsigargin, 2) Xestospongin C, and 3) the protonophore carbonyl cyanide 4-(trifluoromethoxy) phenyl hydrazone; inhibition of calcium increase by calcium channel blockers, by nifedipine and verapamil, indicated that PACAP was also acting on calcium influx. Such mobilization was not dependent on a functional actin cytoskeleton. Homologous desensitization with nanomoles of PACAP concentration and heterologous receptors desensibilization by G protein-coupled receptor agonists were observed. Intracellular calcium depletion modulated PACAP-associated ERK but not p38 phosphorylation; in contrast, extracellular calcium depletion modulated PACAP-associated p38 but not ERK phosphorylation. In PACAP-treated PMNs, reactive oxygen species production and CD11b membrane up-regulation in contrast to lactoferrin release were dependent on both intra- and extracellular calcium, whereas matrix metalloproteinase-9 release was unaffected by extracellular calcium depletion. These data indicate that both extracellular and intracellular calcium play key roles in PACAP proinflammatory activities.


Subject(s)
Calcium/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Signal Transduction/immunology , CD11b Antigen/biosynthesis , Calcium Signaling , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Neuropeptides/pharmacology , Reactive Oxygen Species/metabolism
8.
J Immunol ; 173(6): 4154-63, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15356166

ABSTRACT

Anti-inflammatory activities of pituitary adenylate cyclase-activating protein (PACAP) are mediated in part through specific effects on lymphocytes and macrophages. This study shows that in human polymorphonuclear neutrophils (PMNs), PACAP acts as a proinflammatory molecule. In PMNs, vaso-intestinal peptide/PACAP receptor 1 (VPAC-1) was the only receptor found to be expressed by RT-PCR. Using VPAC-1 Ab, we found that VPAC-1 mRNA was translated into proteins. In PMNs, PACAP increases cAMP, inositol triphosphate metabolites, and calcium. It activates two of the three members of the MAPK superfamily, the ERK and the stress-activated MAPK p38. U73122, an inhibitor of phospholipase C (PLC), inhibits PACAP-induced ERK activation, whereas p38 MAPK phosphorylation was unaffected. Using specific pharmalogical inhibitors of ERK (PD098059) and p38 MAPK (SB203580), we found that PACAP-mediated calcium increase was ERK and PLC dependent and p38 independent. PACAP primes fMLP-associated calcium increase; it also primes fMLP activation of the respiratory burst as well as elastase release, these last two processes being ERK and PLC dependent and p38 MAPK independent. PACAP also increases membrane expression of CD11b and release of lactoferrin and metallo proteinase-9 (MMP-9). These effects were PLC dependent (CD 11b, lactoferrin, MMP-9), ERK dependent (CD 11b, lactoferrin, MMP-9), and p38 dependent (CD11b, lactoferrin). We conclude that PACAP is a direct PMN activator as well as an effective PMN priming agent that requires PLC, ERK, and p38 MAPK activities.


Subject(s)
Inflammation Mediators/physiology , Mitogen-Activated Protein Kinases/metabolism , Neuropeptides/physiology , Neutrophil Activation/immunology , Neutrophils/enzymology , Neutrophils/immunology , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/physiology , CD11b Antigen/biosynthesis , Calcium/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Cyclic AMP/metabolism , Enzyme Activation/immunology , Humans , Inflammation Mediators/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Lactoferrin/metabolism , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinases/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neuropeptides/metabolism , Neutrophils/metabolism , Pancreatic Elastase/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide/biosynthesis , Receptors, Vasoactive Intestinal Polypeptide, Type I , Respiratory Burst/immunology , Signal Transduction/immunology , Type C Phospholipases/physiology , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...