Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Clin Exp Immunol ; 212(1): 52-60, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: covidwho-2308658

RESUMEN

Inflammation plays a fundamental role in the development of several metabolic diseases, including obesity and type 2 diabetes (T2D); the complement system has been implicated in their development. People of Black African (BA) ethnicity are disproportionately affected by T2D and other metabolic diseases but the impact of ethnicity on the complement system has not been explored. We investigated ethnic differences in complement biomarkers and activation status between men of BA and White European (WE) ethnicity and explored their association with parameters of metabolic health. We measured a panel of 15 complement components, regulators, and activation products in fasting plasma from 89 BA and 96 WE men. Ethnic differences were statistically validated. Association of complement biomarkers with metabolic health indices (BMI, waist circumference, insulin resistance, and HbA1c) were assessed in the groups. Plasma levels of the key complement components C3 and C4, the regulators clusterin and properdin and the activation marker iC3b were significantly higher in BA compared to WE men after age adjustment, while FD levels were significantly lower. C3 and C4 levels positively correlated with some or all markers of metabolic dysfunction in both ethnic groups while FD was inversely associated with HbA1c in both groups, and clusterin and properdin were inversely associated with some markers of metabolic dysfunction only in the WE group. Our findings of increased levels of complement components and activation products in BA compared to WE men suggest differences in complement regulation that may impact susceptibility to poor metabolic health.


Asunto(s)
Clusterina , Resistencia a la Insulina , Enfermedades Metabólicas , Properdina , Humanos , Masculino , Biomarcadores , Diabetes Mellitus Tipo 2 , Etnicidad , Hemoglobina Glucada , Población Blanca , Población Negra , Enfermedades Metabólicas/etnología , Complemento C4 , Complemento C3
2.
Immunobiology ; 228(2): 152351, 2023 03.
Artículo en Inglés | MEDLINE | ID: covidwho-2245868

RESUMEN

We have attempted to explore further the involvement of complement components in the host COVID-19 (Coronavirus disease-19) immune responses by targeted genotyping of COVID-19 adult patients and analysis for missense coding Single Nucleotide Polymorphisms (coding SNPs) of genes encoding Alternative pathway (AP) components. We have identified a small group of common coding SNPs in Survivors and Deceased individuals, present in either relatively similar frequencies (CFH and CFI SNPs) or with stark differences in their relative abundance (C3 and CFB SNPs). In addition, we have identified several sporadic, potentially protective, coding SNPs of C3, CFB, CFD, CFH, CFHR1 and CFI in Survivors. No coding SNPs were detected for CD46 and CD55. Our demographic analysis indicated that the C3 rs1047286 or rs2230199 coding SNPs were present in 60 % of all the Deceased patients (n = 25) (the rs2230199 in 67 % of all Deceased Males) and in 31 % of all the Survivors (n = 105, p = 0.012) (the rs2230199 in 25 % of all Survivor Males). When we analysed these two major study groups using the presence of the C3 rs1047286 or rs2230199 SNPs as potential biomarkers, we noticed the complete absence of the protective CFB rs12614 and rs641153 coding SNPs from Deceased Males compared to Females (p = 0.0023). We propose that in these individuals, C3 carrying the R102G and CFB lacking the R32W or the R32Q amino acid substitutions, may contribute to enhanced association dynamics of the C3bBb AP pre-convertase complex assembly, thus enabling the exploitation of the activation of the Complement Alternative pathway (AP) by SARS-CoV-2.


Asunto(s)
COVID-19 , Degeneración Macular , Masculino , Femenino , Humanos , Factor B del Complemento/genética , Complemento C3/genética , Polimorfismo de Nucleótido Simple , Genotipo , Degeneración Macular/genética , Factor H de Complemento/genética , SARS-CoV-2 , Complemento C2/genética
3.
Immunobiology ; 228(1): 152316, 2023 01.
Artículo en Inglés | MEDLINE | ID: covidwho-2165411

RESUMEN

We studied the role of cytotoxic components (DAMPs) formed in the body of patients with COVID-19 in ensuring the long-term preservation of post-COVID-19 manifestations and the possibility of creating an experimental model by transferring DAMPs to rats. In patients with post-COVID-19 syndrome (PCS) 2 months after SARS-CoV-2 infection we determined the presence of cytotoxic components in the blood serum (Terasaki test, Dunaliella viridis test and content of DAMPs). In post-COVID-19 syndrome patients with a high content of serum cytotoxic oligopeptide fraction (selective group, n = 16) we determined the number of leukocytes, lymphocytes, neutrophil granulocytes and monocytes in the blood, the content of C-reactive protein (CRP), the concentration of C3 and C4 complement components and circulating immune complexes, the serum content of IL-6, IL -10, IL-18, TNF-α, phagocytic activity of neutrophils, presence of neutrophil traps and autoantibodies ANA. It has been shown that in patients with PCS, there are components with cytotoxicity in the blood serum, form specific immunopathological patterns, which are characterized by: an increased content of CRP, complement system components C3 and C4 and cytokines (TNF-α, IL-6, IL-10, IL-18) activation, the formation of a wide range of autoantibodies ANA, the low efficiency of endocytosis in oxygen-independent phagocytosis; their phagocytic activity reaches its functional limit, and against this background, activation of neutrophil traps occurs, which can contribute to further induction of DAMPs. This self-sustaining cell-killing activation provided long-term preservation of PCS symptoms. The transfer of blood serum components from selective group patients with PCS to rats was accompanied by the appearance of cytotoxic components in them which induced sensitization and immunopathological reactions. Preventive administration of a biologically active substance with polyfunctional properties MF to experimental animals "corrected" the initial functional state of the body's immune-metabolic system and eliminated or facilitated immuno-inflammatory reactions.


Asunto(s)
COVID-19 , Humanos , Ratas , Animales , Interleucina-18 , Síndrome Post Agudo de COVID-19 , Interleucina-6 , Factor de Necrosis Tumoral alfa , Peso Molecular , SARS-CoV-2 , Proteína C-Reactiva/metabolismo , Complemento C3 , Autoanticuerpos
4.
Front Immunol ; 13: 1007102, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-2163013

RESUMEN

Background: The complement system is an essential component of our innate defense and plays a vital role in the pathogenesis of many diseases. Assessment of complement activation is critical in monitoring both disease progression and response to therapy. Complement analysis requires accurate and standardized sampling and assay procedures, which has proven to be challenging. Objective: We performed a systematic analysis of the current methods used to assess complement components and reviewed whether the identified studies performed their complement measurements according to the recommended practice regarding pre-analytical sample handling and assay technique. Results are supplemented with own data regarding the assessment of key complement biomarkers to illustrate the importance of accurate sampling and measuring of complement components. Methods: A literature search using the Pubmed/MEDLINE database was performed focusing on studies measuring the key complement components C3, C5 and/or their split products and/or the soluble variant of the terminal C5b-9 complement complex (sTCC) in human blood samples that were published between February 2017 and February 2022. The identified studies were reviewed whether they had used the correct sample type and techniques for their analyses. Results: A total of 92 out of 376 studies were selected for full-text analysis. Forty-five studies (49%) were identified as using the correct sample type and techniques for their complement analyses, while 25 studies (27%) did not use the correct sample type or technique. For 22 studies (24%), it was not specified which sample type was used. Conclusion: A substantial part of the reviewed studies did not use the appropriate sample type for assessing complement activation or did not mention which sample type was used. This deviation from the standardized procedure can lead to misinterpretation of complement biomarker levels and hampers proper comparison of complement measurements between studies. Therefore, this study underlines the necessity of general guidelines for accurate and standardized complement analysis.


Asunto(s)
Activación de Complemento , Complemento C5 , Humanos , Complemento C3 , Complejo de Ataque a Membrana del Sistema Complemento , Biomarcadores
5.
J Virol ; 96(18): e0102422, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: covidwho-2008764

RESUMEN

Zoonotic coronaviruses represent an ongoing threat to public health. The classical porcine epidemic diarrhea virus (PEDV) first appeared in the early 1970s. Since 2010, outbreaks of highly virulent PEDV variants have caused great economic losses to the swine industry worldwide. However, the strategies by which PEDV variants escape host immune responses are not fully understood. Complement component 3 (C3) is considered a central component of the three complement activation pathways and plays a crucial role in preventing viral infection. In this study, we found that C3 significantly inhibited PEDV replication in vitro, and both variant and classical PEDV strains induced high levels of interleukin-1ß (IL-1ß) in Huh7 cells. However, the PEDV variant strain reduces C3 transcript and protein levels induced by IL-1ß compared with the PEDV classical strain. Examination of key molecules of the C3 transcriptional signaling pathway revealed that variant PEDV reduced C3 by inhibiting CCAAT/enhancer-binding protein ß (C/EBP-ß) phosphorylation. Mechanistically, PEDV nonstructural protein 1 (NSP1) inhibited C/EBP-ß phosphorylation via amino acid residue 50. Finally, we constructed recombinant PEDVs to verify the critical role of amino acid 50 of NSP1 in the regulation of C3 expression. In summary, we identified a novel antiviral role of C3 in inhibiting PEDV replication and the viral immune evasion strategies of PEDV variants. Our study reveals new information on PEDV-host interactions and furthers our understanding of the pathogenic mechanism of this virus. IMPORTANCE The complement system acts as a vital link between the innate and the adaptive immunity and has the ability to recognize and neutralize various pathogens. Activation of the complement system acts as a double-edged sword, as appropriate levels of activation protect against pathogenic infections, but excessive responses can provoke a dramatic inflammatory response and cause tissue damage, leading to pathological processes, which often appear in COVID-19 patients. However, how PEDV, as the most severe coronavirus causing diarrhea in piglets, regulates the complement system has not been previously reported. In this study, for the first time, we identified a novel mechanism of a PEDV variant in the suppression of C3 expression, showing that different coronaviruses and even different subtype strains differ in regulation of C3 expression. In addition, this study provides a deeper understanding of the mechanism of the PEDV variant in immune escape and enhanced virulence.


Asunto(s)
Complemento C3 , Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Proteínas no Estructurales Virales , Replicación Viral , Animales , Antivirales , COVID-19/inmunología , Línea Celular Tumoral , Complemento C3/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/fisiología
6.
Int Immunopharmacol ; 110: 109019, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: covidwho-1914514

RESUMEN

OBJECTIVES: COVID-19 is an immune-related disease caused by novel Coronavirus SARS-COV-2. Lung lesions persist in some recovered patients, making long-term follow-up monitoring of their health necessary. The mechanism of these abnormalities is still unclear. In this study, the immune status was observed to explore the immune mechanism of persistent lung CT abnormalities in one-year COVID-19 recovered subjects. METHODS: One-year follow-up of 73 recovered patients from COVID-19 confirmed in Taizhou City, Zhejiang Province, was conducted to collect laboratory indicators such as blood immune cells, cytokines, complement series, immunoglobulin, and lung imaging; According to the results of lung CT, 60 patients were divided into normal CT group (n = 40) and abnormal CT group (n = 20). We compared the dynamic changes of immune indexes at three timepoints namely onset (T1), discharge (T2), and 1-year follow-up (T3), and studied the relationship between immune indexes and pulmonary sequelae. RESULTS: Compared with the healthy control, there was no significant difference in immune-related indexes, and immune levels had recovered. Patients with elder age, high BMI, severe patients, and those with underlying diseases (hypertension or diabetes) had a higher CT abnormal rate after recovery. Longitudinal observation showed that immunoglobulin increased first and then decreased, immune cell TBNK decreased in the onset period and increased in the recovery period, cytokine level increased significantly in the onset period and decreased to the normal level in the recovery period, and complement series C1q, C3 and C4 increased at the onset and decreased during the one-year follow-up. Complement C3 remained at a high level in the CT abnormal group (CT normal group vs CT abnormal group; P = 0.036). Correlation analysis showed that C3 negatively correlated restrictive ventilation index (TLC-He (ratio) (r = -0.302, P = 0.017). The above results suggest that complement C3 is a negative factor correlating abnormal pulmonary function 1 year after the recovery. CONCLUSION: After one year recovering from COVID-19, the subjects were with stable immune indicators. High levels of complement C3 were associated with persistent lung abnormalities in COVID-19 recovered subjects.


Asunto(s)
COVID-19 , Anciano , Estudios de Cohortes , Complemento C3 , Humanos , Inmunoglobulinas , Estudios Longitudinales , Pulmón/diagnóstico por imagen , SARS-CoV-2 , Tomografía Computarizada por Rayos X
7.
Nat Commun ; 13(1): 317, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1671550

RESUMEN

Activation of the serum-resident complement system begins a cascade that leads to activation of membrane-resident complement receptors on immune cells, thus coordinating serum and cellular immune responses. Whilst many molecules act to control inappropriate activation, Properdin is the only known positive regulator of the human complement system. By stabilising the alternative pathway C3 convertase it promotes complement self-amplification and persistent activation boosting the magnitude of the serum complement response by all triggers. In this work, we identify a family of tick-derived alternative pathway complement inhibitors, hereafter termed CirpA. Functional and structural characterisation reveals that members of the CirpA family directly bind to properdin, inhibiting its ability to promote complement activation, and leading to potent inhibition of the complement response in a species specific manner. We provide a full functional and structural characterisation of a properdin inhibitor, opening avenues for future therapeutic approaches.


Asunto(s)
Proteínas de Artrópodos/química , Proteínas de Artrópodos/inmunología , Inactivadores del Complemento/química , Inactivadores del Complemento/inmunología , Properdina/inmunología , Rhipicephalus/inmunología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/genética , Activación de Complemento , Complemento C3/química , Complemento C3/inmunología , Vía Alternativa del Complemento , Humanos , Cinética , Properdina/química , Properdina/genética , Rhipicephalus/química , Rhipicephalus/genética , Alineación de Secuencia
8.
Thromb Haemost ; 122(2): 240-256, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: covidwho-1642057

RESUMEN

BACKGROUND: Endothelial and complement activation were both associated with immunothrombosis, a key determinant of COVID-19 severity, but their interrelation has not yet been investigated. OBJECTIVES: We aimed to determine von Willebrand factor (VWF) antigen (VWF:Ag) concentration, VWF collagen binding activity (VWF:CBA), a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) activity (ADAMTS13:Ac), and their ratios in hospitalized COVID-19 patients, and to investigate how these parameters and their constellation with complement activation relate to disease severity and in-hospital mortality in COVID-19. METHODS: Samples of 102 hospitalized patients with polymerase chain reaction-confirmed severe acute respiratory syndrome coronavirus 2 positivity were included in our observational cohort study. Patients were stratified according to the peak severity of COVID-19 disease in agreement with the World Health Organization ordinal scale. Twenty-six convalescent plasma donors with previous COVID-19 disease formed the control group. VWF:Ag concentration and VWF:CBA were determined by enzyme-linked immunosorbent assay (ELISA); ADAMTS13:Ac was determined by fluorescence resonance energy transfer. Complement C3 and C3a were measured by turbidimetry and ELISA, respectively. Clinical covariates and markers of inflammation were extracted from hospital records. RESULTS: VWF:Ag and VWF:CBA were elevated in all groups of hospitalized COVID-19 patients and increased in parallel with disease severity. ADAMTS13:Ac was decreased in patients with severe COVID-19, with the lowest values in nonsurvivors. High (> 300%) VWF:Ag concentrations or decreased (< 67%) ADAMTS13:Ac were associated with higher risk of severe COVID-19 disease or in-hospital mortality. The concomitant presence of decreased ADAMTS13:Ac and increased C3a/C3 ratio-indicating complement overactivation and consumption-was a strong independent predictor of in-hospital mortality. CONCLUSION: Our results suggest that an interaction between the VWF-ADAMTS13 axis and complement overactivation and consumption plays an important role in the pathogenesis of COVID-19.


Asunto(s)
Proteína ADAMTS13/metabolismo , COVID-19/inmunología , Complemento C3/metabolismo , SARS-CoV-2/fisiología , Factor de von Willebrand/metabolismo , Adulto , Anciano , COVID-19/epidemiología , COVID-19/mortalidad , Activación de Complemento , Convalecencia , Femenino , Hospitalización , Humanos , Hungría/epidemiología , Masculino , Persona de Mediana Edad , Nefelometría y Turbidimetría , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
9.
J Am Soc Nephrol ; 32(1): 99-114, 2021 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1496673

RESUMEN

BACKGROUND: C3 glomerulopathy (C3G) is characterized by the alternative-pathway (AP) hyperactivation induced by nephritic factors or complement gene mutations. Mice deficient in complement factor H (CFH) are a classic C3G model, with kidney disease that requires several months to progress to renal failure. Novel C3G models can further contribute to understanding the mechanism behind this disease and developing therapeutic approaches. METHODS: A novel, rapidly progressing, severe, murine model of C3G was developed by replacing the mouse C3 gene with the human C3 homolog using VelociGene technology. Functional, histologic, molecular, and pharmacologic assays characterize the presentation of renal disease and enable useful pharmacologic interventions in the humanized C3 (C3hu/hu) mice. RESULTS: The C3hu/hu mice exhibit increased morbidity early in life and die by about 5-6 months of age. The C3hu/hu mice display elevated biomarkers of kidney dysfunction, glomerulosclerosis, C3/C5b-9 deposition, and reduced circulating C3 compared with wild-type mice. Administration of a C5-blocking mAb improved survival rate and offered functional and histopathologic benefits. Blockade of AP activation by anti-C3b or CFB mAbs also extended survival and preserved kidney function. CONCLUSIONS: The C3hu/hu mice are a useful model for C3G because they share many pathologic features consistent with the human disease. The C3G phenotype in C3hu/hu mice may originate from a dysregulated interaction of human C3 protein with multiple mouse complement proteins, leading to unregulated C3 activation via AP. The accelerated disease course in C3hu/hu mice may further enable preclinical studies to assess and validate new therapeutics for C3G.


Asunto(s)
Complemento C3/genética , Modelos Animales de Enfermedad , Glomerulonefritis Membranoproliferativa/genética , Enfermedades Renales/genética , Animales , Complemento C3/metabolismo , Vía Alternativa del Complemento/genética , Exones , Regulación de la Expresión Génica , Glomerulonefritis Membranoproliferativa/metabolismo , Humanos , Enfermedades Renales/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Fenotipo , Polimorfismo de Nucleótido Simple , Insuficiencia Renal/genética , Insuficiencia Renal/metabolismo
10.
J Med Virol ; 93(9): 5515-5522, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1363690

RESUMEN

Complement system hyperactivation has been proposed as a potential driver of adverse outcomes in severe acute respiratory syndrome coronavirus 2 infected patients, given prior research of complement deposits found in tissue and blood samples, as well as evidence of clinical improvement with anticomplement therapy. Its role in augmenting thrombotic microangiopathy mediated organ damage has also been implicated in coronavirus disease 2019 (COVID-19). This study aimed to examine associations between complement parameters and progression to severe COVID-19 illness, as well as correlations with other systems. Blood samples of COVID-19 patients presenting to the emergency department (ED) were analyzed for a wide panel of complement and inflammatory biomarkers. The primary outcome was COVID-19 severity at index ED visit, while the secondary outcome was peak disease severity over the course of illness. Fifty-two COVID-19 patients were enrolled. C3a (p = 0.018), C3a/C3 ratio (p = 0.002), and sC5b-9/C3 ratio (p = 0.021) were significantly elevated in with severe disease at ED presentation. Over the course of illness, C3a (p = 0.028) and C3a/C3 ratio (p = 0.003) were highest in the moderate severity group. In multivariate regression controlled for confounders, complement hyperactivation failed to predict progression to severe disease. C3a, C3a/C3 ratio, and sC5b-9/C3 ratio were correlated positively with numerous inflammatory biomarkers, fibrinogen, and VWF:Ag, and negatively with plasminogen and ADAMTS13 activity. We found evidence of complement hyperactivation in COVID-19, associated with hyperinflammation and thrombotic microangiopathy. Complement inhibition should be further investigated for potential benefit in patients displaying a hyperinflammatory and microangiopathic phenotype.


Asunto(s)
Proteína ADAMTS13/sangre , COVID-19/sangre , Complemento C3/análisis , Complejo de Ataque a Membrana del Sistema Complemento/análisis , SARS-CoV-2 , Adulto , Anciano , Biomarcadores/sangre , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Ohio , Admisión del Paciente , Índice de Severidad de la Enfermedad
11.
Biosci Rep ; 41(8)2021 08 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1334001

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2)-induced infection, the cause of coronavirus disease 2019 (COVID-19), is characterized by unprecedented clinical pathologies. One of the most important pathologies, is hypercoagulation and microclots in the lungs of patients. Here we study the effect of isolated SARS-CoV-2 spike protein S1 subunit as potential inflammagen sui generis. Using scanning electron and fluorescence microscopy as well as mass spectrometry, we investigate the potential of this inflammagen to interact with platelets and fibrin(ogen) directly to cause blood hypercoagulation. Using platelet-poor plasma (PPP), we show that spike protein may interfere with blood flow. Mass spectrometry also showed that when spike protein S1 is added to healthy PPP, it results in structural changes to ß and γ fibrin(ogen), complement 3, and prothrombin. These proteins were substantially resistant to trypsinization, in the presence of spike protein S1. Here we suggest that, in part, the presence of spike protein in circulation may contribute to the hypercoagulation in COVID-19 positive patients and may cause substantial impairment of fibrinolysis. Such lytic impairment may result in the persistent large microclots we have noted here and previously in plasma samples of COVID-19 patients. This observation may have important clinical relevance in the treatment of hypercoagulability in COVID-19 patients.


Asunto(s)
COVID-19/patología , Fibrina/metabolismo , Fibrinólisis/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Trombosis/patología , Adulto , Anciano , Amiloide/metabolismo , Plaquetas/metabolismo , Complemento C3/metabolismo , Femenino , Fibrinógeno/metabolismo , Humanos , Pulmón/patología , Masculino , Técnicas Analíticas Microfluídicas , Persona de Mediana Edad , Protrombina/metabolismo , SARS-CoV-2/metabolismo , Trombosis/virología , Tripsina/metabolismo
12.
Int J Mol Sci ; 22(13)2021 Jul 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1304672

RESUMEN

Cardiovascular diseases have attracted our full attention not only because they are the main cause of mortality and morbidity in many countries but also because the therapy for and cure of these maladies are among the major challenges of the medicine in the 21st century [...].


Asunto(s)
Enfermedades Cardiovasculares/etiología , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Vesículas Extracelulares/metabolismo , Marcadores Genéticos , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Modelos Cardiovasculares , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Factores de Riesgo
13.
Front Immunol ; 12: 696085, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1282389

RESUMEN

Activation of the complement system has been observed in coronavirus disease 19 (COVID-19). We conducted a systematic review and meta-analysis with meta-regression to investigate possible differences in the serum concentrations of two routinely measured complement components, C3 and C4, in COVID-19 patients with different severity and survival status. We searched PubMed, Web of Science and Scopus, between January 2020 and February 2021, for studies reporting serum complement C3 and C4, measures of COVID-19 severity, and survival. Eligibility criteria were a) reporting continuous data on serum C3 and C4 concentrations in COVID-19 patients, -b) investigating COVID-19 patients with different disease severity and/or survival status, c) adult patients, d) English language, e) ≥10 patients, and f) full-text available. Using a random-effects model, standardized mean differences (SMD) with 95% confidence intervals (CIs) were calculated to evaluate differences in serum C3 and C4 concentrations between COVID-19 patients with low vs. high severity or survivor vs. non-survivor status. Risk of bias was assessed using the Newcastle-Ottawa scale whereas publication bias was assessed with the Begg's and Egger's tests. Certainty of evidence was assessed using GRADE. Nineteen studies in 3,764 COVID-19 patients were included in the meta-analysis. Both C3 and C4 concentrations were significantly lower in patients with high disease severity or non-survivor status than patients with low severity or survivor status (C3 SMD=-0.40, 95% CI -0.60 to -0.21, p<0.001; C4 SMD=-0.29, 95% CI -0.49 to -0.09, p=0.005; moderate certainty of evidence). Extreme between-study heterogeneity was observed (C3, I2 = 82.1%; C4, I2 = 84.4%). Sensitivity analysis, performed by sequentially removing each study and re-assessing the pooled estimates, showed that the magnitude and direction of the effect size was not modified. There was no publication bias. In meta-regression, the SMD of C3 was significantly associated with white blood cell count, C-reactive protein (CRP), and pro-thrombin time, whereas the SMD of C4 was significantly associated with CRP, pro-thrombin time, D-dimer, and albumin. In conclusion, lower concentrations of C3 and C4, indicating complement activation, were significantly associated with higher COVID-19 severity and mortality. C3 and C4 might be useful to predict adverse clinical consequences in these patients. Systematic Review Registration: PROSPERO, Registration number: CRD42021239634.


Asunto(s)
COVID-19/sangre , Complemento C3/metabolismo , Complemento C4/metabolismo , Biomarcadores/sangre , COVID-19/mortalidad , COVID-19/patología , Activación de Complemento , Humanos , SARS-CoV-2 , Índice de Severidad de la Enfermedad
14.
J Allergy Clin Immunol ; 147(6): 2083-2097.e6, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1272498

RESUMEN

BACKGROUND: Excessive inflammation triggered by a hitherto undescribed mechanism is a hallmark of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and is associated with enhanced pathogenicity and mortality. OBJECTIVE: Complement hyperactivation promotes lung injury and was observed in patients suffering from Middle East respiratory syndrome-related coronavirus, SARS-CoV-1, and SARS-CoV-2 infections. Therefore, we investigated the very first interactions of primary human airway epithelial cells on exposure to SARS-CoV-2 in terms of complement component 3 (C3)-mediated effects. METHODS: For this, we used highly differentiated primary human 3-dimensional tissue models infected with SARS-CoV-2 patient isolates. On infection, viral load, viral infectivity, intracellular complement activation, inflammatory mechanisms, and tissue destruction were analyzed by real-time RT-PCR, high content screening, plaque assays, luminex analyses, and transepithelial electrical resistance measurements. RESULTS: Here, we show that primary normal human bronchial and small airway epithelial cells respond to SARS-CoV-2 infection by an inflated local C3 mobilization. SARS-CoV-2 infection resulted in exaggerated intracellular complement activation and destruction of the epithelial integrity in monolayer cultures of primary human airway cells and highly differentiated, pseudostratified, mucus-producing, ciliated respiratory tissue models. SARS-CoV-2-infected 3-dimensional cultures secreted significantly higher levels of C3a and the proinflammatory cytokines IL-6, monocyte chemoattractant protein 1, IL-1α, and RANTES. CONCLUSIONS: Crucially, we illustrate here for the first time that targeting the anaphylotoxin receptors C3a receptor and C5a receptor in nonimmune respiratory cells can prevent intrinsic lung inflammation and tissue damage. This opens up the exciting possibility in the treatment of COVID-19.


Asunto(s)
Bronquios/inmunología , COVID-19/inmunología , Activación de Complemento , Células Epiteliales/inmunología , Receptor de Anafilatoxina C5a/inmunología , Mucosa Respiratoria/inmunología , SARS-CoV-2/inmunología , Bronquios/patología , Bronquios/virología , COVID-19/patología , COVID-19/virología , Línea Celular , Complemento C3/inmunología , Citocinas/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
16.
mBio ; 12(2)2021 04 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1206005

RESUMEN

SARS-CoV-2 infection causing the COVID-19 pandemic calls for immediate interventions to avoid viral transmission, disease progression, and subsequent excessive inflammation and tissue destruction. Primary normal human bronchial epithelial cells are among the first targets of SARS-CoV-2 infection. Here, we show that ColdZyme medical device mouth spray efficiently protected against virus entry, excessive inflammation, and tissue damage. Applying ColdZyme to fully differentiated, polarized human epithelium cultured at an air-liquid interphase (ALI) completely blocked binding of SARS-CoV-2 and increased local complement activation mediated by the virus as well as productive infection of the tissue model. While SARS-CoV-2 infection resulted in exaggerated intracellular complement activation immediately following infection and a drop in transepithelial resistance, these parameters were bypassed by single pretreatment of the tissues with ColdZyme mouth spray. Crucially, our study highlights the importance of testing already evaluated and safe drugs such as ColdZyme mouth spray for maintaining epithelial integrity and hindering SARS-CoV-2 entry within standardized three-dimensional (3D) in vitro models mimicking the in vivo human airway epithelium.IMPORTANCE Although our understanding of COVID-19 continuously progresses, essential questions regarding prophylaxis and treatment remain open. A hallmark of severe SARS-CoV-2 infection is a hitherto-undescribed mechanism leading to excessive inflammation and tissue destruction associated with enhanced pathogenicity and mortality. To tackle the problem at the source, transfer of SARS-CoV-2, subsequent binding, infection, and inflammatory responses have to be avoided. In this study, we used fully differentiated, mucus-producing, and ciliated human airway epithelial cultures to test the efficacy of ColdZyme medical device mouth spray in terms of protection from SARS-CoV-2 infection. Importantly, we found that pretreatment of the in vitro airway cultures using ColdZyme mouth spray resulted in significantly shielding the epithelial integrity, hindering virus binding and infection, and blocking excessive intrinsic complement activation within the airway cultures. Our in vitro data suggest that ColdZyme mouth spray may have an impact in prevention of COVID-19.


Asunto(s)
Antivirales/farmacología , Mucosa Respiratoria/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Bronquios/citología , COVID-19/prevención & control , COVID-19/virología , Complemento C3/inmunología , Células Epiteliales , Humanos , Inmunidad Innata/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Vaporizadores Orales , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , SARS-CoV-2/fisiología , Acoplamiento Viral/efectos de los fármacos
17.
Sci Rep ; 11(1): 7857, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1180263

RESUMEN

Given that a substantial proportion of the subgroup of COVID-19 patients that face a severe disease course are younger than 60 years, it is critical to understand the disease-specific characteristics of young COVID-19 patients. Risk factors for a severe disease course for young COVID-19 patients and possible non-linear influences remain unknown. Data were analyzed from COVID-19 patients with clinical outcome in a single hospital in Wuhan, China, collected retrospectively from Jan 24th to Mar 27th. Clinical, demographic, treatment and laboratory data were collected from patients' medical records. Uni- and multivariable analysis using logistic regression and random forest, with the latter allowing the study of non-linear influences, were performed to investigate the clinical characteristics of a severe disease course. A total of 762 young patients (median age 47 years, interquartile range [IQR] 38-55, range 18-60; 55.9% female) were included, as well as 714 elderly patients as a comparison group. Among the young patients, 362 (47.5%) had a severe/critical disease course and the mean age was statistically significantly higher in the severe subgroup than in the mild subgroup (59.3 vs. 56.0, Student's t-test: p < 0.001). The uni- and multivariable analysis suggested that several covariates such as elevated levels of serum amyloid A (SAA), C-reactive protein (CRP) and lactate dehydrogenase (LDH), and decreased lymphocyte counts influence disease severity independently of age. Elevated levels of complement C3 (odds ratio [OR] 15.6, 95% CI 2.41-122.3; p = 0.039) are particularly associated with the risk of developing severe COVID-19 specifically in young patients, whereas no such influence seems to exist for elderly patients. Additional analysis suggests that the influence of complement C3 in young patients is independent of age, gender, and comorbidities. Variable importance values and partial dependence plots obtained using random forests delivered additional insights, in particular indicating non-linear influences of risk factors on disease severity. This study identified increased levels of complement C3 as a unique risk factor for adverse outcomes specific to young COVID-19 patients.


Asunto(s)
COVID-19/sangre , Complemento C3/análisis , Adolescente , Adulto , Área Bajo la Curva , COVID-19/inmunología , China/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Análisis Multivariante , Dinámicas no Lineales , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Adulto Joven
18.
Clin Immunol ; 226: 108726, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1179332

RESUMEN

Recent studies suggest excessive complement activation in severe coronavirus disease-19 (COVID-19). The latter shares common characteristics with complement-mediated thrombotic microangiopathy (TMA). We hypothesized that genetic susceptibility would be evident in patients with severe COVID-19 (similar to TMA) and associated with disease severity. We analyzed genetic and clinical data from 97 patients hospitalized for COVID-19. Through targeted next-generation-sequencing we found an ADAMTS13 variant in 49 patients, along with two risk factor variants (C3, 21 patients; CFH,34 patients). 31 (32%) patients had a combination of these, which was independently associated with ICU hospitalization (p = 0.022). Analysis of almost infinite variant combinations showed that patients with rs1042580 in thrombomodulin and without rs800292 in complement factor H did not require ICU hospitalization. We also observed gender differences in ADAMTS13 and complement-related variants. In light of encouraging results by complement inhibitors, our study highlights a patient population that might benefit from early initiation of specific treatment.


Asunto(s)
Proteína ADAMTS13/genética , COVID-19/genética , Complemento C3/genética , Predisposición Genética a la Enfermedad/genética , Trombomodulina/genética , Anciano , Algoritmos , COVID-19/fisiopatología , Activación de Complemento , Factor H de Complemento/genética , Cuidados Críticos , Femenino , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Hospitalización/estadística & datos numéricos , Humanos , Unidades de Cuidados Intensivos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Índice de Severidad de la Enfermedad , Microangiopatías Trombóticas/genética
19.
Pharmacol Rev ; 73(2): 792-827, 2021 04.
Artículo en Inglés | MEDLINE | ID: covidwho-1125571

RESUMEN

The complement system was discovered at the end of the 19th century as a heat-labile plasma component that "complemented" the antibodies in killing microbes, hence the name "complement." Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. SIGNIFICANCE STATEMENT: The complement system is the host's defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host's enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.


Asunto(s)
COVID-19/epidemiología , COVID-19/fisiopatología , Proteínas del Sistema Complemento/fisiología , Enfermedades Raras/fisiopatología , Colectinas/metabolismo , Enzimas Activadoras de Complemento/metabolismo , Complemento C3/metabolismo , Inactivadores del Complemento/farmacología , Terapia Genética/métodos , Humanos , Mediadores de Inflamación/metabolismo , Lectinas/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Pandemias , SARS-CoV-2 , Sinapsis/metabolismo
20.
Front Immunol ; 12: 581469, 2021.
Artículo en Inglés | MEDLINE | ID: covidwho-1119544

RESUMEN

Background: Epidemiological factors, clinical characteristics, and risk factors for the mortality of COVID-19 patients have been studied, but the role of complementary systems, possible inflammatory and immune response mechanisms, and detailed clinical courses are uncertain and require further study. Methods: In this single center, retrospective case-control study, we included all COVID-19 inpatients transferred or admitted to Wuhan Tongji Hospital from January 3 to March 30 2020 who had definite clinical outcomes (cured or deceased) with complete laboratory and radiological results. Clinical data were extracted from the electronic medical records, and compared between the cured and deceased patients. ROC curves were used to evaluate the prognostic value of the clinical parameters, and multivariable logistic regression analysis was performed to explore the risk factors for mortality. The correlation between the variables was evaluated by Spearman correlation analysis. Results: 208 patients were included in this study, 182 patients were cured and discharged, 26 patients died from COVID-2019. Most patients had comorbidities, with hypertension as the most common chronic disease (80; 38%). The most common symptoms at onset were fever (149; 72%), cough (137; 66%), and dyspnea (113; 54%). Elevated leucocytes, neutrophils, inflammatory biomarkers (CRP, ferritin, IL6, IL8, procalcitonin), PT, D-dimer, myocardial enzymes, BUN, decreased lymphocyte and subsets (T cells, CD4 T cells, CD8 T cells, NK cells, T cells + B cells + NK cells), and immunological factors (C3, C4) indicated poor outcome. PT, C3, and T cells were confirmed as independent prognostic factors for mortality by logistic regression models. IL6 and CPR were positively correlated with neutrophils, but negatively with lymphocytes and lymphocyte subsets except B cells. IL8 and ferritin were negatively related to T cells and CD4 T cells. Positive associations existed between C3 and T cells, CD4 T cells, and CD8 T cells, whereas there was no significant correlation between C4 and lymphocyte subsets. PT was found positively correlated with IL6, IL8, and CRP. Reverse correlations were explored between C3, C4, and PT, CK-MB, total bilirubin. Conclusions: T cells, C3, and PT were identified as independent prognostic factors for mortality. Decreased C3 and C4, dysregulation of lymphocyte subsets and cytokines may lead to death after SARS-CoV-2 infection.


Asunto(s)
COVID-19/epidemiología , COVID-19/mortalidad , Complemento C3/análisis , Citocinas/sangre , Subgrupos de Linfocitos T/inmunología , Anciano , COVID-19/patología , Estudios de Casos y Controles , Comorbilidad , Femenino , Humanos , Hipertensión/complicaciones , Células Asesinas Naturales/inmunología , Recuento de Linfocitos , Linfopenia/patología , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA