Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.648
Filtrar
1.
Sudan J Paediatr ; 24(1): 73-79, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38952618

RESUMO

Multisystem inflammatory syndrome of childhood (MIS-C) is a recently described entity in pediatrics post-COVID-19 pandemic. Hemophagocytic lymphohistiocytosis (HLH) is a clinical syndrome caused by an unregulated proliferation of macrophages as well as T lymphocytes. Both entities can be considered overlapping, although distinct criteria for each can be found in the literature. Herein, we report a patient with MIS-C post-COVID-19 infection, complicated with HLH secondary to Plasmodium falciparum malaria from a blood transfusion.

2.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38959328

RESUMO

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Assuntos
COVID-19 , Classe Ib de Fosfatidilinositol 3-Quinase , Inflamação , SARS-CoV-2 , COVID-19/patologia , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Animais , Inflamação/patologia , Humanos , Tratamento Farmacológico da COVID-19 , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Pulmão/patologia , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Síndrome da Liberação de Citocina/tratamento farmacológico , Permeabilidade Capilar/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia
3.
Cell Commun Signal ; 22(1): 349, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965547

RESUMO

T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.


Assuntos
Basigina , COVID-19 , Linfopenia , SARS-CoV-2 , Humanos , Linfopenia/imunologia , Linfopenia/virologia , COVID-19/imunologia , COVID-19/virologia , COVID-19/patologia , SARS-CoV-2/metabolismo , Basigina/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Síndrome da Liberação de Citocina/imunologia , Animais
4.
J Indian Soc Periodontol ; 28(1): 113-121, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38988958

RESUMO

Background: Periodontal disease is associated with immune dysregulation, and cytokines released can add on to the coronavirus disease 2019 (COVID-19)-associated cytokine storm, further worsening the related adverse outcomes. Specific studies investigating cytokine levels in COVID-19 patients with periodontal disease are lacking. Examining the correlation between these conditions could aid in categorizing risk categories, determining referrals, and strengthening oral hygiene protocols. The current study sought to evaluate cytokine levels in the saliva of COVID-19-positive patients with and without periodontal disease. Materials and Methods: Twenty-six COVID-19-positive patients were subjected to periodontal examination, saliva collection, and assessment of cytokine levels through cytokine bead-based multiplex assay, using fluorescence-encoded beads with flow cytometry (BD FACS LSRFortessa). Eleven cytokines were assessed (interleukin [IL] 2, 4, 6, 10, 17A, and interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-α), chemokine ligand 2 (CCL2/monocyte chemoattractant protein-1), C-X-C motif chemokine ligand (CXCL) 8/IL 8, CXCL 9/monokine-induced gamma interferon [MIG]), and CXCL 10 (chemokine IFN-gamma inducible protein 10 kDa). The cytokine levels of the recruited subjects were also compared graphically with the salivary cytokine levels reported in the literature for health, COVID-19, and periodontal disease alone. Results: Out of 26 COVID-19-positive patients, 17 had periodontal disease. Levels of all cytokines were raised in patients with both diseases when compared to values reported in literature for health, periodontal disease alone, or COVID-19 alone. However, there was no statistical difference among the recruited subjects for IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-gamma, TNF-α, CCL2, CXCL 8, and CXCL 10. MIG levels were found to be higher in periodontally healthy, COVID-19-positive subjects (P = 0.01). Conclusions: Periodontal disease might contribute to the COVID-19-induced cytokine storm, potentially amplifying its impact.

5.
Int J Emerg Med ; 17(1): 85, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992604

RESUMO

INTRODUCTION: Severe multisystem inflammatory syndrome in children (MIS-C) and severe dengue are challenging to identify during the COVID-19 pandemic in dengue-endemic areas. Fever, multiorgan involvement, and shock characterize both severe MIS-C and severe dengue. Distinguishing between the two diseases is beneficial in initiating proper management. METHODS: Medical records of children < 18 years old who were hospitalized at Hasan Sadikin General Hospital's PICU between December 2020 and July 2022 with severe MIS-C or severe dengue were recorded. Differences were assessed using comparative and descriptive analyses. RESULTS: Seventeen severe dengue patients and 4 severe MIS-C were included. The average age of severe MIS-C was 11.5 years (SD ± 2.9, 95% CI), and that of severe dengue patients was 6.2 years (SD ± 4.4, 95% CI) (p value = 0.034, 95%). Fever and abdominal pain were the most common symptoms in both groups (p = 0.471, 95% CI). Rash (p = 0.049) and nonpurulent conjunctivitis (p = 0.035) were two symptoms with significant differences. The highest platelet count (p-value = 0.006, 95% CI), AST (p-value = 0.026, 95% CI), and D-dimer level (p-value = 0.025, 95% CI) were significantly different between the two cohorts. Cardiac abnormalities were found in all (100%) severe MIS-C patients, but only one (5.9%) in severe dengue patients. CONCLUSION: Age, rash, nonpurulent conjunctivitis, platelet count, AST and D-dimer level may distinguish severe MIS-C from severe dengue fever.

6.
Int J Mol Sci ; 25(13)2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-39000379

RESUMO

Hyperinflammatory Coronavirus disease 2019 (COVID-19) and rapidly-progressive interstitial lung diseases (RP-ILD) secondary to inflammatory myopathies (IIM) present important similarities. These data support the use of anti-rheumatic drugs for the treatment of COVID-19. The aim of this study was to compare the efficacy of combining baricitinib and pulse steroids with the Standard of Care (SoC) for the treatment of critically ill COVID-19 patients. We retrospectively enrolled consecutive patients admitted to the Intensive Care Unit (ICU) with COVID-19-pneumonia. Patients treated with SoC (dexamethasone plus remdesivir) were compared to patients treated with baricitinib plus 6-methylprednisolone pulses (Rheuma-group). We enrolled 246 patients: 104/246 in the SoC and 142/246 in the Rheuma-group. All patients presented laboratory findings suggestive of hyperinflammatory response. Sixty-four patients (26.1%) died during ICU hospitalization. The mortality rate in the Rheuma-group was significantly lower than in the SoC-group (15.5 vs. 40.4%, p < 0.001). Compared to the SoC-group, patients in the Rheuma-group presented significantly lower inflammatory biomarker levels after one week of treatment. Higher ferritin levels after one week of treatment were strongly associated with mortality (p < 0.001). In this large real-life COVID-19 cohort, baricitinib and pulse steroids led to a significant reduction in mortality, paralleled by a prompt reduction in inflammatory biomarkers. Our experience supports the similarities between hyperinflammatory COVID-19 and the IIM-associated RP-ILD.


Assuntos
Azetidinas , Tratamento Farmacológico da COVID-19 , COVID-19 , Quimioterapia Combinada , Unidades de Terapia Intensiva , Metilprednisolona , Purinas , Pirazóis , SARS-CoV-2 , Sulfonamidas , Humanos , Purinas/uso terapêutico , Purinas/administração & dosagem , Masculino , Feminino , Azetidinas/uso terapêutico , Azetidinas/administração & dosagem , Sulfonamidas/uso terapêutico , Sulfonamidas/administração & dosagem , Pirazóis/uso terapêutico , Pirazóis/administração & dosagem , Pessoa de Meia-Idade , Idoso , Estudos Retrospectivos , Metilprednisolona/uso terapêutico , Metilprednisolona/administração & dosagem , COVID-19/mortalidade , COVID-19/complicações , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Monofosfato de Adenosina/administração & dosagem , Resultado do Tratamento , Alanina/análogos & derivados , Alanina/uso terapêutico , Alanina/administração & dosagem
7.
Cureus ; 16(6): e62731, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-39036184

RESUMO

Ileocecal intussusception (ICI) is the most common abdominal emergency and cause of intestinal obstruction in young children, carrying a high risk of mortality and morbidity. Enteric viral infectious and inflammatory syndromes are known triggers for intussusception (ileoileal and ileocolic) by causing mesenteric lymphoid hyperplasia that may act as a leading point allowing the bowel to invaginate into itself. Gastrointestinal (GI) symptoms are common in children with coronavirus disease 2019 (COVID-19) infection, with a subset of patients solely having GI complaints at the time of presentation.  COVID-19 as a trigger for intussusception in children has been hypothesized and suggested in multiple cases reported to date, both during the acute phase of illness and as a part of multisystem inflammatory syndrome in children (MIS-C). We present a seven-month-old male who developed ICI and became a diagnostic dilemma due to viral co-infections and the gradual emergence of MIS-C during the hospital stay. We are describing this presentation in an attempt to expand the understanding of the implications of COVID-19 and MIS-C in this young and unique age group.

8.
Asian J Transfus Sci ; 18(1): 138-140, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39036681

RESUMO

Multi-system inflammatory syndrome in children associated with COVID19 (MIS-C) is a unique clinical syndrome characterised by fever, gastrointestinal symptoms, skin and oral rash and or neurological symptoms in the presence of raised acute phase reactants and coagulopathy. Ferritin is an acute phase reactant which is used as a marker of inflammation. Diagnosis of MIS-C in the background of transfusion dependent thalassemia with iron overload needs a strong clinical suspicion. Early diagnosis and prompt treatment are necessary to ensure a rapid uneventful recovery. A three-year-old male child born to non-consanguineously related parents reported to pediatric emergency with difficulty breathing and pain abdomen for one day. The child was a diagnosed case of Beta thalassemia major since the age of one year and was on regular transfusions and was on iron chelation for past eleven months with deferrasirox. Initial clinical examination showed a sick and irritable child with tachypnea tachycardia and hypoxia. Initial investigations showed raised acute phase reactants along with severe anemia. The child was investigated for MIS-C because of unexpected rise of serum ferritin from 1980 ng/mL (October 2020) to 6686 ng/mL (in January 2021) despite being on regular chelation. Antibody titre for SARS COVID-19 was positive. The patient was treated with intravenous corticosteroids and improved with the same. The advent of COVID19 pandemic saw most children having a mild disease with no or minimal symptoms. Some kids however presented with more serious delayed symptoms of MIS-C. To diagnose same in multi transfused patients a strong clinical suspicion and just judgement based on the clinical and laboratory findings should be done. Unexplained rise in ferritin levels, typical symptoms and high probability of exposure to COVID19 helped in clinching diagnosis.

9.
medRxiv ; 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39040211

RESUMO

Background: Severe COVID and multisystem inflammatory syndrome (MIS-C) are characterized by excessive inflammatory cytokines/chemokines. In adults, disease severity is associated with SARS-CoV-2-specific IgG Fc afucosylation, which induces pro-inflammatory cytokine secretion from innate immune cells. This study aimed to define spike IgG Fc glycosylation following SARS-CoV-2 infection in adults and children and following SARS-CoV-2 vaccination in adults and the relationships between glycan modifications and cytokine/chemokine levels. Methods: We analyzed longitudinal (n=146) and cross-sectional (n=49) serum/plasma samples from adult and pediatric COVID patients, MIS-C patients, adult vaccinees, and adult and pediatric healthy controls. We developed methods for characterizing bulk and spike IgG Fc glycosylation by capillary electrophoresis (CE) and measured levels of ten inflammatory cytokines/chemokines by multiplexed ELISA. Results: Spike IgG were more afucosylated than bulk IgG during acute adult COVID and MIS-C. We observed an opposite trend following vaccination, but it was not significant. Spike IgG were more galactosylated and sialylated and less bisected than bulk IgG during adult COVID, with similar trends observed during pediatric COVID/MIS-C and following SARS-CoV-2 vaccination. Spike IgG glycosylation changed with time following adult COVID or vaccination. Afucosylated spike IgG exhibited inverse and positive correlations with inflammatory markers in MIS-C and following vaccination, respectively; galactosylated and sialylated spike IgG inversely correlated with pro-inflammatory cytokines in adult COVID and MIS-C; and bisected spike IgG positively correlated with inflammatory cytokines/chemokines in multiple groups. Conclusions: We identified previously undescribed relationships between spike IgG glycan modifications and inflammatory cytokines/chemokines that expand our understanding of IgG glycosylation changes that may impact COVID and MIS-C immunopathology.

10.
J Pak Med Assoc ; 74(7): 1345-1350, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39028068

RESUMO

The current study was planned to explore the potential synergistic role of the co-administration of sarilumab and dexamethasone in reducing blood biomarkers associated with cytokine release syndrome in hospitalised patients of coronavirus disease-2019. The sample comprised 22 patients hospitalised with severe and critical severity levels and who were treated with sarilumab and dexamethasone. Positive responses were seen in blood biomarkers, including decreased interleukin-6 alpha levels and improved oxygen saturation. Tumour necrosis factor, Ddimer, C-reactive protein, ferritin and lymphocyte count also showed positive responses in patients who survived than those who died. Lactate dehydrogenase levels fluctuated with improvement among the survivors, but had limited effectiveness in those who died. The findings suggested promising avenues for future treatment strategies in patients with severe coronavirus disease-2019 and cytokine release syndrome.


Assuntos
Anticorpos Monoclonais Humanizados , Biomarcadores , Proteína C-Reativa , Tratamento Farmacológico da COVID-19 , COVID-19 , Síndrome da Liberação de Citocina , Dexametasona , Ferritinas , SARS-CoV-2 , Humanos , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Síndrome da Liberação de Citocina/tratamento farmacológico , Síndrome da Liberação de Citocina/sangue , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Masculino , Feminino , Pessoa de Meia-Idade , Biomarcadores/sangue , COVID-19/sangue , COVID-19/complicações , Ferritinas/sangue , Proteína C-Reativa/metabolismo , Proteína C-Reativa/análise , Interleucina-6/sangue , Quimioterapia Combinada , Fator de Necrose Tumoral alfa/sangue , Contagem de Linfócitos , L-Lactato Desidrogenase/sangue , Adulto , Idoso , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Hospitalização
11.
Hepatol Forum ; 5(3): 139-149, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39006140

RESUMO

COVID-19 has affected millions worldwide, causing significant morbidity and mortality. While predominantly involving the respiratory tract, SARS-CoV-2 has also caused systemic illnesses involving other sites. Liver injury due to COVID-19 has been variably reported in observational studies. It has been postulated that liver damage may be due to direct damage by the SARS-CoV-2 virus or multifactorial secondary to hepatotoxic therapeutic options, as well as cytokine release syndrome and sepsis-induced multiorgan dysfunction. The approach to a COVID-19 patient with liver injury requires a thorough evaluation of the pattern of hepatocellular injury, along with the presence of underlying chronic liver disease and concurrent medications which may cause drug-induced liver injury. While studies have shown uneventful recovery in the majority of mildly affected patients, severe COVID-19 associated liver injury has been associated with higher mortality, prolonged hospitalization, and greater morbidity in survivors. Furthermore, its impact on long-term outcomes remains to be ascertained as recent studies report an association with metabolic-fatty liver disease. This present review provides insight into the subject by describing the postulated mechanism of liver injury, its impact in the presence of pre-existing liver disease, and its short- and long-term clinical implications.

12.
PLoS One ; 19(7): e0299965, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39018291

RESUMO

The challenges of the COVID-19 pandemic have highlighted an increasing clinical demand for safe and effective treatment options against an overzealous immune defence response, also known as the "cytokine storm". Andrographolide is a naturally derived bioactive compound with promising anti-inflammatory activity in many clinical studies. However, its cytokine-inhibiting activity, in direct comparison to commonly used nonsteroidal anti-inflammatory drugs (NSAIDs), has not been extensively investigated in existing literature. The anti-inflammatory activities of andrographolide and common NSAIDs, such as diclofenac, aspirin, paracetamol and ibuprofen were measured on lipopolysaccharide (LPS) and interferon-γ induced RAW264.7 cells. The levels of PGE2, nitric oxide (NO), TNF-α & LPS-induced release of pro-inflammatory cytokines on differentiated human macrophage THP-1 cells were measured against increasing concentrations of andrographolide and aforementioned NSAIDs. The associated mechanistic pathway was examined on NFκB using flow cytometry on the human endothelial-leukocyte adhesion molecule (ELAM9) (E-selectin) transfected RAW264.7 cells with green fluorescent protein (GFP). Andrographolide exhibited broad and potent anti-inflammatory and cytokine-inhibiting activity in both cell lines by inhibiting the release of IL-6, TNF-α and IFN-γ, which are known to play a key role in the etiology of cytokine storm and the pathogenesis of inflammation. In comparison, the tested NSAIDs demonstrated weak or no activity against proinflammatory mediators except for PGE2, where the activity of andrographolide (IC50 = 8.8 µM, 95% CI = 7.4 to 10.4 µM) was comparable to that of paracetamol (IC50 = 7.73 µM, 95% CI = 6.14 to 9.73 µM). The anti-inflammatory action of andrographolide was associated with its potent downregulation of NFκB. The wide-spectrum anti-inflammatory activity of andrographolide demonstrates its therapeutic potential against cytokine storms as an alternative to NSAIDs.


Assuntos
Anti-Inflamatórios não Esteroides , Citocinas , Diterpenos , Diterpenos/farmacologia , Animais , Camundongos , Humanos , Anti-Inflamatórios não Esteroides/farmacologia , Células RAW 264.7 , Citocinas/metabolismo , Óxido Nítrico/metabolismo , Anti-Inflamatórios/farmacologia , Síndrome da Liberação de Citocina/tratamento farmacológico , NF-kappa B/metabolismo , Dinoprostona/metabolismo , Lipopolissacarídeos/farmacologia , Células THP-1 , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Interferon gama/metabolismo , Selectina E/metabolismo
13.
Front Pediatr ; 12: 1382133, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39026941

RESUMO

Introduction: The aim of the study was to summarize and update clinical features and outcomes of multisystem inflammatory syndrome in neonates (MIS-N). Methods: A systematic literature search was conducted of studies on MIS-N published in PubMed, MEDLINE, EMBASE, CNKI, and WHO COVID-19 databases between 1 December 2019 and 30 June 2023. Reference lists of selected articles, Google Scholar, and pre-print servers were searched for additional studies. The methodological quality of included studies was assessed. Results: Of 1,572 records screened after the initial search, 35 studies involving a total of 201 neonates with MIS-N were included. One study was retrieved from a pre-print server. For those with available data, 34/47 (78.7%) mothers were infected in the third trimester. Of the 199 mothers (two with twin pregnancies), 183 (92.0%) were from India. The median age of neonates at presentation was 2.0 days (interquartile range 1.0-9.5). Over two-thirds (144/201, 71.6%) presented with respiratory distress, while 112 (55.7%) had cardiac involvement, such as ventricular dysfunctions, involvement of coronary arteries, and atrioventricular blocks. Arrhythmias and thrombosis were reported in 15/201 (7.5%) and 2/201 (3.0%) neonates, respectively. All neonates, except one, required critical care; 64/160 (40.0%) required inotropic support and 105/187 (56.1%) required respiratory support, of whom 59/105 (56.2%) were specified to require intubation. The mortality rate was 5.0% (10/201). Discussion/Conclusion: MIS-N should be considered in ill neonates presenting with involvement of two or more organ systems, especially among those neonates with cardiorespiratory dysfunctions, in the presence of proven or suspected maternal COVID-19 infection during pregnancy. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021278717, PROSPERO, identifier CRD42021278717.

14.
Clin Exp Immunol ; 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39028583

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces pneumonia and acute respiratory failure in Coronavirus Disease 2019 (COVID-19) patients with inborn errors of immunity to type I interferon (IFN-I). The impact of SARS-CoV-2 infection varies widely, ranging from mild respiratory symptoms to life-threatening illness and organ failure, with a higher incidence in men than in women. Approximately 3 to 5% of critical COVID-19 patients under 60 and a smaller percentage of elderly patients exhibit genetic defects in IFN-I production, including X-chromosome-linked TLR7 and autosomal TLR3 deficiencies. Around 15 to 20% of cases over 70 years old, and a smaller percentage of younger patients, present with preexisting autoantibodies neutralizing type I interferons. Additionally, innate errors affecting the control of the response to type I interferon have been associated with pediatric multisystem inflammatory syndrome (MIS-C). Several studies have described rare errors of immunity, such as XIAP deficiency, CYBB, SOCS1, OAS1/2, and RNASEL, as underlying factors in MIS-C susceptibility. However, further investigations in expanded patient cohorts are needed to validate these findings and pave the way for new genetic approaches to MIS-C. This review aims to present recent evidence from the scientific literature on genetic and immunological abnormalities predisposing individuals to critical SARS-CoV-2 infection through IFN-I. We will also discuss multisystem inflammatory syndrome in children (MIS-C). Understanding the immunological mechanisms and pathogenesis of severe COVID-19 may inform personalized patient care and population protection strategies against future serious viral infections.

15.
Viruses ; 16(6)2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38932242

RESUMO

Multisystem Inflammatory Syndrome in Children (MIS-C) is a potentially life-threatening complication of COVID-19. The pathophysiological mechanisms leading to severe disease are poorly understood. This study leveraged clinical samples from a well-characterized cohort of children hospitalized with COVID-19 or MIS-C to compare immune-mediated biomarkers. Our objective was to identify selected immune molecules that could explain, in part, why certain SARS-CoV-2-infected children developed MIS-C. We hypothesized that type-2 helper T cell-mediated inflammation can elicit autoantibodies, which may account for some of the differences observed between the moderate-severe COVID-19 (COVID+) and MIS-C cohort. We enumerated blood leukocytes and measured levels of selected serum cytokines, chemokines, antibodies to COVID-19 antigens, and autoantibodies in children presenting to an academic medical center in Connecticut, United States. The neutrophil/lymphocyte and eosinophil/lymphocyte ratios were significantly higher in those in the MIS-C versus COVID+ cohort. IgM and IgA, but not IgG antibodies to SARS-CoV-2 receptor binding domain were significantly higher in the MIS-C cohort than the COVID+ cohort. The serum levels of certain type-2 cytokines (interleukin (IL)-4, IL-5, IL-6, IL-8, IL-10, IL-13, and IL-33) were significantly higher in children with MIS-C compared to the COVID+ and SARS-CoV-2-negative cohorts. IgG autoantibodies to brain antigens and pentraxin were higher in children with MIS-C compared to SARS-CoV-19-negative controls, and children with MIS-C had higher levels of IgG anti-contactin-associated protein-like 2 (caspr2) compared to the COVID+ and SARS-CoV-19-negative controls. We speculate that autoimmune responses in certain COVID-19 patients may induce pathophysiological changes that lead to MIS-C. The triggers of autoimmunity and factors accounting for type-2 inflammation require further investigation.


Assuntos
Autoanticorpos , COVID-19 , Citocinas , SARS-CoV-2 , Síndrome de Resposta Inflamatória Sistêmica , Humanos , COVID-19/imunologia , COVID-19/sangue , COVID-19/complicações , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Síndrome de Resposta Inflamatória Sistêmica/sangue , Criança , Feminino , Masculino , Estudos Prospectivos , SARS-CoV-2/imunologia , Pré-Escolar , Autoanticorpos/sangue , Autoanticorpos/imunologia , Citocinas/sangue , Adolescente , Lactente , Biomarcadores/sangue , Anticorpos Antivirais/sangue , Inflamação/imunologia , Inflamação/sangue
16.
Int J Mol Sci ; 25(11)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38892153

RESUMO

The role of programmed death cell protein 1 (PD-1) has already been described in a range of various diseases, including COVID-19. This study provides new, innovative data, related to the expression of PD-1 and the risk of Paediatric Inflammatory Multisystem Syndrome, temporally associated with SARS-CoV-2 infection (PIMS-TS)-a rare, but potentially life-threatening complication of COVID-19. In this study, we evaluated the expression of PD-1 protein in patients with PIMS. Blood samples were taken from patients at the time of diagnosis (n = 33), after 6 weeks (n = 33), 3 months (n = 24), 6 months (n = 24) and 12 months (n = 8). The immunophenotypes were evaluated in flow cytometry. The control group consisted of 35 healthy children with negative SARS-CoV-2 antigen/PCR test, who were asymptomatic and had no history of allergic, autoimmune or oncological diseases. The associations between immunophenotypes, biochemical findings and clinical data were analysed. Significant increases in the expression of PD-1 for CD4+ and CD8+ T cells, compared to the control group, were observed in the day of admission, with a gradual decrease during the first weeks from initiation of treatment. This study sheds new light on the pathogenesis of PIMS-TS, emphasizing the role of PD-1 protein. Future research is essential for early risk prediction in SARS-CoV-2 patients and for devising effective clinical prevention and management strategies.


Assuntos
COVID-19 , Receptor de Morte Celular Programada 1 , SARS-CoV-2 , Síndrome de Resposta Inflamatória Sistêmica , Humanos , COVID-19/complicações , COVID-19/imunologia , COVID-19/sangue , COVID-19/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/sangue , Síndrome de Resposta Inflamatória Sistêmica/diagnóstico , Masculino , Criança , Feminino , Pré-Escolar , Estudos Prospectivos , Adolescente , Lactente , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Imunofenotipagem
17.
J Infect Dis ; 229(6): 1740-1749, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38871359

RESUMO

BACKGROUND: We examined effects of single-nucleotide variants (SNVs) of IL1RN, the gene encoding the anti-inflammatory interleukin 1 receptor antagonist (IL-1Ra), on the cytokine release syndrome (CRS) and mortality in patients with acute severe respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. METHODS: IL1RN CTA haplotypes formed from 3 SNVs (rs419598, rs315952, rs9005) and the individual SNVs were assessed for association with laboratory markers of inflammation and mortality. We studied 2589 patients hospitalized with SARS-CoV-2 between March 2020 and March 2021. RESULTS: Mortality was 15.3% and lower in women than men (13.1% vs 17.3%, P = .0003). Carriers of the CTA-1/2 IL1RN haplotypes exhibited decreased inflammatory markers and increased plasma IL-1Ra. Evaluation of the individual SNVs of the IL1RN, carriers of the rs419598 C/C SNV exhibited significantly reduced inflammatory biomarker levels and numerically lower mortality compared to the C/T-T/T genotype (10.0% vs 17.8%, P = .052) in men, with the most pronounced association observed in male patients ≤74 years old, whose mortality was reduced by 80% (3.1% vs 14.0%, P = .030). CONCLUSIONS: The IL1RN haplotype CTA and C/C variant of rs419598 are associated with attenuation of the CRS and decreased mortality in men with acute SARS-CoV-2 infection. The data suggest that the IL1RN pathway modulates the severity of coronavirus disease 2019 (COVID-19) via endogenous anti-inflammatory mechanisms.


Assuntos
COVID-19 , Síndrome da Liberação de Citocina , Haplótipos , Proteína Antagonista do Receptor de Interleucina 1 , Polimorfismo de Nucleotídeo Único , SARS-CoV-2 , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Antagonista do Receptor de Interleucina 1/sangue , COVID-19/mortalidade , COVID-19/genética , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , SARS-CoV-2/genética , Síndrome da Liberação de Citocina/genética , Síndrome da Liberação de Citocina/mortalidade , Adulto , Genótipo , Biomarcadores/sangue
18.
Int J Immunopathol Pharmacol ; 38: 3946320241265265, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38889772

RESUMO

Introduction: Exceedingly high levels of the chemokine CCL5/RANTES have been found in fatty degenerated osteonecrotic alveolar bone cavities (FDOJ) and aseptic ischemic osteolysis of the jaw (AIOJ) from toothless regions. Because CCL5/RANTES seems to have a prominent role in creating the COVID-19 "cytokine storm", some researchers have used the monoclonal antibody Leronlimab to block the CCR5 on inflammatory cells.Objective: Is preexisting FDOJ/AIOJ jaw marrow pathology a "hidden" co-morbidity affecting some COVID-19 infections? To what extent does the chronic CCL5/RANTES expression from preexisting FDOJ/AIOJ areas contribute to the progression of the acute cytokine storm in COVID-19 patients?Methods: Authors report on reducing the COVID-19 "cytokine storm" by treating infected patients through targeting the chemokine receptor 5 (CCR5) with Leronlimab and interrupting the activation of CCR5 by high CCL5/RANTES signaling, thus dysregulating the inflammatory phase of the viremia. Surgical removal of FDOJ/AIOJ lesions with high CCL5/RANTES from patients with inflammatory diseases may be classified as a co-morbid disease.Results: Both multiplex analysis of 249 FDOJ/AIOJ bone tissue samples as well as serum levels of CCL5/RANTES displayed exceedingly high levels in both specimens.Discussion: By the results the authors hypothesize that chronic CCL5/RANTES induction from FDOJ/AIOJ areas may sensitize CCR5 throughout the immune system, thus, enabling it to amplify its response when confronted with the virus. As conventional intraoral radiography does little to assess the quality of the alveolar bone, ultrasonography units are available to help dentists locate the FDOJ/AIOJ lesions in an office setting.Conclusion: The authors propose a new approach to containment of the COVID-19 cytokine storm by a prophylactic focus for future viral-related pandemics, which may be early surgical clean-up of CCL5/RANTES expression sources in the FDOJ/AIOJ areas, thus diminishing a possible pre-sensitization of CCR5. A more complete dental examination includes trans-alveolar ultrasono-graphy (TAU) for hidden FDOJ/AIOJ lesions.


Assuntos
COVID-19 , Quimiocina CCL5 , Humanos , COVID-19/imunologia , COVID-19/epidemiologia , Comorbidade , Masculino , Feminino , Pessoa de Meia-Idade , Receptores CCR5/metabolismo , Idoso , Doenças Maxilomandibulares/epidemiologia , Doenças Maxilomandibulares/imunologia , SARS-CoV-2 , Síndrome da Liberação de Citocina , Anticorpos Monoclonais Humanizados/uso terapêutico , Adulto
19.
Cytotherapy ; 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38864802

RESUMO

BACKGROUND: Post-transplant or hematological cancer patients have a higher risk of mortality after infection with ancestral and early variants of severe acute respiratory syndrome (SARS)-CoV-2. Adoptive cell therapy (ACT) with virus-specific T cells (VSTs) could augment endogenous T cell immunity to avoid disease deterioration before viral clearance. METHODS: We established a third-party SARS-CoV-2-specific T cell (COVID-T) bank in 2020 (NCT04351659) using convalescent and/or vaccinated donors. In a phase I/II study (NCT04457726), 13 adult and pediatric patients, acutely positive for SARS-CoV-2 and predicted to have a high chance of mortality, were recruited from September 2021 to February 2022. Twelve patients received a single dose of COVID-T cells, matched on at least 1 HLA. RESULTS: A dose of either 75,000 or 150,000 IFN-γ+CD3+ cells/m2 SARS-COV-2-specific T cells did not cause cytokine release syndrome, acute respiratory distress syndrome, or graft-versus-host disease. In the 8 patients who had detectable donor SARS-COV-2-specific T cells after ACT, none progressed to severe disease or died with COVID-19. In contrast, among the other four patients without evidence of donor micro-chimerism, two died of COVID-19. CONCLUSIONS: Long-acting third-party VSTs from convalescent or vaccinated donors could be expediently produced and might be clinically useful in future pandemics, particularly before global vaccination is implemented.

20.
Mol Med ; 30(1): 81, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38862942

RESUMO

BACKGROUND: Studies have highlighted a possible crosstalk between the pathogeneses of COVID-19 and systemic lupus erythematosus (SLE); however, the interactive mechanisms remain unclear. We aimed to elucidate the impact of COVID-19 on SLE using clinical information and the underlying mechanisms of both diseases. METHODS: RNA-seq datasets were used to identify shared hub gene signatures between COVID-19 and SLE, while genome-wide association study datasets were used to delineate the interaction mechanisms of the key signaling pathways. Finally, single-cell RNA-seq datasets were used to determine the primary target cells expressing the shared hub genes and key signaling pathways. RESULTS: COVID-19 may affect patients with SLE through hematologic involvement and exacerbated inflammatory responses. We identified 14 shared hub genes between COVID-19 and SLE that were significantly associated with interferon (IFN)-I/II. We also screened and obtained four core transcription factors related to these hub genes, confirming the regulatory role of the IFN-I/II-mediated Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling pathway on these hub genes. Further, SLE and COVID-19 can interact via IFN-I/II and IFN-I/II receptors, promoting the levels of monokines, including interleukin (IL)-6/10, tumor necrosis factor-α, and IFN-γ, and elevating the incidence rate and risk of cytokine release syndrome. Therefore, in SLE and COVID-19, both hub genes and core TFs are enriched within monocytes/macrophages. CONCLUSIONS: The interaction between SLE and COVID-19 promotes the activation of the IFN-I/II-triggered JAK-STAT signaling pathway in monocytes/macrophages. These findings provide a new direction and rationale for diagnosing and treating patients with SLE-COVID-19 comorbidity.


Assuntos
COVID-19 , Estudo de Associação Genômica Ampla , Lúpus Eritematoso Sistêmico , SARS-CoV-2 , Transdução de Sinais , Humanos , COVID-19/genética , Lúpus Eritematoso Sistêmico/genética , SARS-CoV-2/fisiologia , Feminino , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição STAT/genética , Masculino , Transcriptoma , Perfilação da Expressão Gênica , Multiômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA