Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.117
Filter
1.
Cytokine ; 157: 155959, 2022 09.
Article in English | MEDLINE | ID: mdl-35816926

ABSTRACT

BACKGROUND: Serious trauma due to various factors is a major global public issue, and sepsis is a major cause of trauma-associated mortality. Timely diagnosis and suitable treatment of post-traumatic sepsis are crucial to improve the hospital outcome of traumatic patients. IL-28 is a newly discovered member of IFN-λ family with multiple functions in inflammatory response. To date, its role in the pathogenic mechanisms of post-traumatic sepsis still remains unknown. METHODS: In total, 20 healthy controls, 55 traumatic patients without sepsis and 54 traumatic patients with sepsis were enrolled in this study. Serum IL-28A/B levels were investigated by ELISA. RESULTS: IL-28A/B levels were significantly increased in traumatic patients compared to healthy volunteers. Moreover, septic trauma patients displayed a significant increase in IL-28A/B levels compared with non-septic patients. In septic patients, IL-28A/B were negatively correlated with IFN-γ, IL-5, IL-13 and IL-17, and positively associated with IL-10. Moreover, IL-28A (AUC: 0.821, 95 %CI: 0.693-0.949) and IL-28B (AUC: 0.811, 95 %CI: 0.691-0.931) were both beneficial to predict increased mortality risk in septic trauma patients, though there was no statistical difference in the predictive value between them. CONCLUSIONS: Early serum levels of IL-28A/B were associated with the development of post-trauma sepsis and could be applied to assess the outcome of traumatic patients with sepsis. Thus, IL-28 may be a potential indicator for post-traumatic sepsis.


Subject(s)
Interferons/blood , Interleukins/blood , Sepsis , Biomarkers , Cytokines , Humans
2.
Proc Natl Acad Sci U S A ; 119(11)2022 03 15.
Article in English | MEDLINE | ID: mdl-35217532

ABSTRACT

The impacts of interferon (IFN) signaling on COVID-19 pathology are multiple, with both protective and harmful effects being documented. We report here a multiomics investigation of systemic IFN signaling in hospitalized COVID-19 patients, defining the multiomics biosignatures associated with varying levels of 12 different type I, II, and III IFNs. The antiviral transcriptional response in circulating immune cells is strongly associated with a specific subset of IFNs, most prominently IFNA2 and IFNG. In contrast, proteomics signatures indicative of endothelial damage and platelet activation associate with high levels of IFNB1 and IFNA6. Seroconversion and time since hospitalization associate with a significant decrease in a specific subset of IFNs. Additionally, differential IFN subtype production is linked to distinct constellations of circulating myeloid and lymphoid immune cell types. Each IFN has a unique metabolic signature, with IFNG being the most associated with activation of the kynurenine pathway. IFNs also show differential relationships with clinical markers of poor prognosis and disease severity. For example, whereas IFNG has the strongest association with C-reactive protein and other immune markers of poor prognosis, IFNB1 associates with increased neutrophil to lymphocyte ratio, a marker of late severe disease. Altogether, these results reveal specialized IFN action in COVID-19, with potential diagnostic and therapeutic implications.


Subject(s)
Blood/metabolism , COVID-19/immunology , Interferons/blood , Proteome , Transcriptome , COVID-19/blood , Case-Control Studies , Datasets as Topic , Humans , Inpatients
3.
Front Immunol ; 12: 723196, 2021.
Article in English | MEDLINE | ID: mdl-34497613

ABSTRACT

Hepatitis C virus (HCV) cure after all-oral direct-acting antiviral (DAA) therapy greatly improves the liver and immune system. We aimed to assess the impact of this HCV clearance on immune system-related markers in plasma and the gene expression profile in human immunodeficiency virus (HIV)/HCV-coinfected patients with advanced cirrhosis. We performed a prospective study on 33 HIV/HCV-coinfected patients at baseline and 36 weeks after the sustained virological response. Gene expression was evaluated by RNA-seq analysis on peripheral blood mononuclear cells (PBMCs) and plasma biomarkers by multiplex immunoassays. We found a decrease in plasma biomarkers (PD1, PDL1, CXCL10, CXCL8, IL12p70, IL10, and TGFß) and liver disease markers (stiffness measurement (LSM), hepatic venous pressure gradient (HVPG), and transaminases, among others). Furthermore, decreased plasma levels of CXCL8, CXCL10, IL10, and PD1 were associated with reduced LSM values. We also found two upregulated (HAS1 and IRG1) and 15 downregulated (CXCL11, CCL8, CCL7, CCL2, ADARB2, RRAD, MX1, SIGLEC1, IFI44L, IFI44, IFI27, IFI6, IFIT3, IFIT1B, and IFIT1) genes at the end of follow-up, all interferon-stimulated genes (ISGs) grouped into four pathways ("cytokine-cytokine receptor interaction", "viral protein interaction with cytokine and cytokine receptor", "chemokine signaling pathway", and "hepatitis C"). Additionally, the decrease in most of these ISGs was significantly related to reduced LSM and HVPG values. In conclusion, HIV/HCV-coinfected patients with advanced-HCV-related cirrhosis who eradicated HCV following DAA therapy exhibited an improvement in liver disease markers and a significant decrease in plasma biomarkers and gene expression related to antiviral/inflammatory response, particularly in levels of several chemokines and ISGs.


Subject(s)
Antiviral Agents/therapeutic use , Biomarkers/blood , Hepacivirus/genetics , Hepatitis C/drug therapy , Liver Cirrhosis/diagnosis , Aged , Chemokines/blood , Coinfection/drug therapy , Female , Gene Expression , HIV Infections/drug therapy , Hepatitis C/complications , Hepatitis C/virology , Humans , Interferons/blood , Liver Cirrhosis/blood , Liver Cirrhosis/etiology , Liver Function Tests , Male , Middle Aged , Prospective Studies , Sustained Virologic Response
4.
J Infect Dis ; 224(5): 777-782, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34467988

ABSTRACT

We analyzed plasma levels of interferons (IFNs) and cytokines, and expression of IFN-stimulated genes in peripheral blood mononuclear cells in patients with coronavirus disease 2019 of varying disease severity. Patients hospitalized with mild disease exhibited transient type I IFN responses, while intensive care unit patients had prolonged type I IFN responses. Type II IFN responses were compromised in intensive care unit patients. Type III IFN responses were induced in the early phase of infection, even in convalescent patients. These results highlight the importance of early type I and III IFN responses in controlling coronavirus disease 2019 progression.


Subject(s)
COVID-19/immunology , Interferon Type I/immunology , Interferon-gamma/immunology , Interferons/immunology , COVID-19/blood , Chemokines/blood , Cytokines/blood , Humans , Interferon Type I/blood , Interferon Type I/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Interferons/blood , Leukocytes, Mononuclear/immunology , SARS-CoV-2/isolation & purification , Interferon Lambda
5.
Nat Immunol ; 22(11): 1428-1439, 2021 11.
Article in English | MEDLINE | ID: mdl-34471264

ABSTRACT

Coordinated local mucosal and systemic immune responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection either protect against coronavirus disease 2019 (COVID-19) pathologies or fail, leading to severe clinical outcomes. To understand this process, we performed an integrated analysis of SARS-CoV-2 spike-specific antibodies, cytokines, viral load and bacterial communities in paired nasopharyngeal swabs and plasma samples from a cohort of clinically distinct patients with COVID-19 during acute infection. Plasma viral load was associated with systemic inflammatory cytokines that were elevated in severe COVID-19, and also with spike-specific neutralizing antibodies. By contrast, nasopharyngeal viral load correlated with SARS-CoV-2 humoral responses but inversely with interferon responses, the latter associating with protective microbial communities. Potential pathogenic microorganisms, often implicated in secondary respiratory infections, were associated with mucosal inflammation and elevated in severe COVID-19. Our results demonstrate distinct tissue compartmentalization of SARS-CoV-2 immune responses and highlight a role for the nasopharyngeal microbiome in regulating local and systemic immunity that determines COVID-19 clinical outcomes.


Subject(s)
COVID-19/immunology , Microbiota/immunology , Nasopharynx/immunology , SARS-CoV-2/physiology , Acute Disease , Adolescent , Adult , Aged , Antibodies, Viral/blood , Cohort Studies , Female , Humans , Immunity, Humoral , Immunity, Mucosal , Interferons/blood , Male , Middle Aged , Nasopharynx/microbiology , Spike Glycoprotein, Coronavirus/immunology , Viral Load , Young Adult
6.
Front Immunol ; 12: 716075, 2021.
Article in English | MEDLINE | ID: mdl-34394120

ABSTRACT

The existence of asymptomatic and re-detectable positive coronavirus disease 2019 (COVID-19) patients presents the disease control challenges of COVID-19. Most studies on immune responses in COVID-19 have focused on moderately or severely symptomatic patients; however, little is known about the immune response in asymptomatic and re-detectable positive (RP) patients. Here we performed a comprehensive analysis of the transcriptomic profiles of peripheral blood mononuclear cells (PBMCs) from 48 COVID-19 patients which included 8 asymptomatic, 13 symptomatic, 15 recovered and 12 RP patients. The weighted gene co-expression network analysis (WGCNA) identified six co-expression modules, of which the turquoise module was positively correlated with the asymptomatic, symptomatic, and recovered COVID-19 patients. The red module positively correlated with symptomatic patients only and the blue and brown modules positively correlated with the RP patients. The analysis by single sample gene set enrichment analysis (ssGSEA) revealed a lower level of IFN response and complement activation in the asymptomatic patients compared with the symptomatic, indicating a weaker immune response of the PBMCs in the asymptomatic patients. In addition, gene set enrichment analysis (GSEA) analysis showed the enrichment of TNFα/NF-κB and influenza infection in the RP patients compared with the recovered patients, indicating a hyper-inflammatory immune response in the PBMC of RP patients. Thus our findings could extend our understanding of host immune response during the progression of COVID-19 disease and assist clinical management and the immunotherapy development for COVID-19.


Subject(s)
Asymptomatic Diseases , COVID-19/immunology , Carrier State/immunology , Leukocytes, Mononuclear/immunology , SARS-CoV-2/immunology , Transcriptome/genetics , Adult , Carrier State/virology , Complement Activation/immunology , Female , Gene Expression Profiling , Humans , Inflammation/immunology , Influenza, Human/complications , Interferons/blood , Interferons/immunology , Male , Middle Aged , NF-kappa B/metabolism , Transcriptome/immunology , Tumor Necrosis Factor-alpha/metabolism , Young Adult
7.
Nat Commun ; 12(1): 4558, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34315904

ABSTRACT

Patients with advanced stage cancers frequently suffer from severe pain as a result of bone metastasis and bone destruction, for which there is no efficacious treatment. Here, using multiple mouse models of bone cancer, we report that agonists of the immune regulator STING (stimulator of interferon genes) confer remarkable protection against cancer pain, bone destruction, and local tumor burden. Repeated systemic administration of STING agonists robustly attenuates bone cancer-induced pain and improves locomotor function. Interestingly, STING agonists produce acute pain relief through direct neuronal modulation. Additionally, STING agonists protect against local bone destruction and reduce local tumor burden through modulation of osteoclast and immune cell function in the tumor microenvironment, providing long-term cancer pain relief. Finally, these in vivo effects are dependent on host-intrinsic STING and IFN-I signaling. Overall, STING activation provides unique advantages in controlling bone cancer pain through distinct and synergistic actions on nociceptors, immune cells, and osteoclasts.


Subject(s)
Bone Neoplasms/complications , Cancer Pain/etiology , Cancer Pain/immunology , Membrane Proteins/metabolism , Neurons/metabolism , Analgesics/pharmacology , Animals , Bone Neoplasms/blood , Cancer Pain/blood , Cell Line, Tumor , Disease Models, Animal , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/pathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Homeodomain Proteins/metabolism , Hyperalgesia/complications , Interferons/blood , Interferons/metabolism , Male , Mammary Neoplasms, Animal/complications , Membrane Proteins/agonists , Mice, Inbred C57BL , Neoplasm Metastasis , Neurons/drug effects , Nociception/drug effects , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/drug effects , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Xanthones/pharmacology
8.
Nat Commun ; 12(1): 3523, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112769

ABSTRACT

There is currently no approved treatment for primary Sjögren's syndrome, a disease that primarily affects adult women. The difficulty in developing effective therapies is -in part- because of the heterogeneity in the clinical manifestation and pathophysiology of the disease. Finding common molecular signatures among patient subgroups could improve our understanding of disease etiology, and facilitate the development of targeted therapeutics. Here, we report, in a cross-sectional cohort, a molecular classification scheme for Sjögren's syndrome patients based on the multi-omic profiling of whole blood samples from a European cohort of over 300 patients, and a similar number of age and gender-matched healthy volunteers. Using transcriptomic, genomic, epigenetic, cytokine expression and flow cytometry data, combined with clinical parameters, we identify four groups of patients with distinct patterns of immune dysregulation. The biomarkers we identify can be used by machine learning classifiers to sort future patients into subgroups, allowing the re-evaluation of response to treatments in clinical trials.


Subject(s)
Cytokines/blood , DNA Methylation , Interferons/blood , Proteome/metabolism , Sjogren's Syndrome/immunology , Transcriptome/genetics , Adult , Autoantibodies/blood , Biomarkers/blood , Chemokines/analysis , Chemokines/genetics , Chemokines/metabolism , Cohort Studies , Computational Biology , Computer Simulation , Cross-Sectional Studies , Cytokines/analysis , Cytokines/genetics , DNA Methylation/genetics , Databases, Genetic , Databases, Protein , Female , Flow Cytometry , Genome-Wide Association Study , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferons/genetics , Male , Middle Aged , Multigene Family , Polymorphism, Single Nucleotide , Proteome/genetics , RNA-Seq , Sjogren's Syndrome/blood , Sjogren's Syndrome/genetics , Sjogren's Syndrome/physiopathology
9.
J Interferon Cytokine Res ; 41(4): 149-152, 2021 04.
Article in English | MEDLINE | ID: mdl-33885337

ABSTRACT

The most recently discovered interferon (IFN) family, type III IFNs or lambda IFNs (IFN-λs) are caused by viral infection and act in mucosal barriers, such as the respiratory tract. In this study, we assessed the serum levels of IFN-λs in new coronavirus disease-2019 (COVID-19) patients. Sixty-four COVID-19 patients were enrolled in this study. All cases were divided into the intensive care unit (ICU) and non-ICU groups according to their symptoms. Fourteen samples of healthy controls were also included. The serum levels of IFN-λ1 and IFN-λ2 were analyzed by specific enzyme-linked immunosorbent assay (ELISA) kits. The concentrations of IFN-λ1 and IFN-λ2 induced in the serum of non-ICU patients (836.7 ± 284.6 and 798.8 ± 301.5 pg/mL, respectively) were higher than found in ICU patients (81.57 ± 34.25 and 48.32 ± 28.13 pg/mL, respectively) (P = 0.004 and P = 0.006, respectively) and healthy controls (85.57 ± 33.63 and 65.82 ± 21.26 pg/mL, respectively) (P = 0.03 and P = 0.04, respectively). Meanwhile, no significant differences were found in the concentration of both cytokines between the ICU patients and healthy controls. We conclude that higher levels of IFN-λs are associated with decreased clinical manifestations in COVID-19 patients. These cytokines could be a promising therapeutic agent to avoid the overwhelming consequences of COVID-19.


Subject(s)
COVID-19 , Interferons/blood , Interleukins/blood , SARS-CoV-2/metabolism , Adult , Aged , COVID-19/blood , COVID-19/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
10.
J Med Virol ; 93(7): 4559-4563, 2021 07.
Article in English | MEDLINE | ID: mdl-33811680

ABSTRACT

Coronavirus disease 2019 (COVID-19) is globally rampant, and to curb the growing burden of this disease, in-depth knowledge about its pathophysiology is needed. This was an observational study conducted at a single center to investigate serum cytokine and chemokine levels of COVID-19 patients, based on disease severity. We included 72 consecutive COVID-19 patients admitted to our hospital from March 21 to August 31, 2020. Patients were divided into Mild-Moderate I (mild) and Moderate II-Severe (severe) groups based on the COVID-19 severity classification developed by the Ministry of Health, Labor and Welfare (MHLW) of Japan. We compared the patient characteristics as well as the serum cytokine and chemokine levels on the day of admission between the two groups. Our findings indicated that the severe group had significantly higher levels of serum fibrinogen, d-dimer, lactate dehydrogenase, C-reactive protein, ferritin, Krebs von den Lungen-6, surfactant protein (SP)-D, and SP-A than the mild group. Strikingly, the levels of interleukin (IL)-28A/interferon (IFN)-λ2 were significantly lower in the severe group than in the mild group. We believe that reduced levels of type III interferons (IFN-λs) and alterations in the levels of other cytokines and chemokines may impact the severity of the disease.


Subject(s)
COVID-19/blood , Chemokines/blood , Interferons/blood , SARS-CoV-2/immunology , Adult , Aged , C-Reactive Protein/analysis , COVID-19/pathology , Down-Regulation , Female , Ferritins/blood , Fibrin Fibrinogen Degradation Products/analysis , Fibrinogen/analysis , Humans , Interferons/biosynthesis , Interleukins/blood , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Mucin-1/blood , Pulmonary Surfactant-Associated Protein A/blood , Pulmonary Surfactant-Associated Protein D/blood , Severity of Illness Index , Interferon Lambda
11.
Immunity ; 54(4): 753-768.e5, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33765435

ABSTRACT

Viral infections induce a conserved host response distinct from bacterial infections. We hypothesized that the conserved response is associated with disease severity and is distinct between patients with different outcomes. To test this, we integrated 4,780 blood transcriptome profiles from patients aged 0 to 90 years infected with one of 16 viruses, including SARS-CoV-2, Ebola, chikungunya, and influenza, across 34 cohorts from 18 countries, and single-cell RNA sequencing profiles of 702,970 immune cells from 289 samples across three cohorts. Severe viral infection was associated with increased hematopoiesis, myelopoiesis, and myeloid-derived suppressor cells. We identified protective and detrimental gene modules that defined distinct trajectories associated with mild versus severe outcomes. The interferon response was decoupled from the protective host response in patients with severe outcomes. These findings were consistent, irrespective of age and virus, and provide insights to accelerate the development of diagnostics and host-directed therapies to improve global pandemic preparedness.


Subject(s)
Immunity/genetics , Virus Diseases/immunology , Antigen Presentation/genetics , Cohort Studies , Hematopoiesis/genetics , Humans , Interferons/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Myeloid Cells/immunology , Myeloid Cells/pathology , Prognosis , Severity of Illness Index , Systems Biology , Transcriptome , Virus Diseases/blood , Virus Diseases/classification , Virus Diseases/genetics , Viruses/classification , Viruses/pathogenicity
12.
Clin Transl Sci ; 14(4): 1524-1534, 2021 07.
Article in English | MEDLINE | ID: mdl-33742764

ABSTRACT

RO6870868 is an oral prodrug of the toll-like receptor 7 (TLR7) specific agonist, RO6871765. TLR7 agonists augment host immune activity and are in development to treat hepatitis B infection. We evaluated the safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of RO6870868 in a first-in-human, phase I, randomized, single ascending oral dose study in 60 healthy volunteers at 6 dose levels (200-2000 mg). Single oral doses were generally well-tolerated with a predictable safety profile associated with dose-dependent increases in systemic interferon. No serious adverse events (AEs) were reported and no subject withdrew from the study due to an AE. No clinically significant changes were observed in vital signs, electrocardiograms, or laboratory parameters. Following oral RO6870868 doses, plasma RO6871765 concentrations increased rapidly, exhibiting mean terminal half-life ranging 2-6 h across all cohorts, with area under the plasma concentration versus time curve extrapolated to infinity (AUC0-∞ ) increasing proportionally with dose. A pattern of dose and time-dependent PD activity was demonstrated consistent with engagement of the TLR7 system. Single RO6870868 doses activated components of the TLR innate immune system in a dose-dependent manner with adequate safety and tolerability. Single-dose data in healthy volunteers are useful to evaluate safety, PK, and PD activity of TLR7 agonists and help to guide dose and regimen selection for further trials in patients with chronic hepatitis B.


Subject(s)
Immunologic Factors/adverse effects , Toll-Like Receptor 7/antagonists & inhibitors , Administration, Oral , Adolescent , Adult , Aged , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Half-Life , Healthy Volunteers , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/immunology , Humans , Immunity, Innate/drug effects , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics , Interferons/blood , Interferons/metabolism , Male , Middle Aged , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Signal Transduction/drug effects , Signal Transduction/immunology , Young Adult
13.
Nat Commun ; 12(1): 1112, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602937

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a worldwide health threat. In a prospective multicentric study, we identify IL-3 as an independent prognostic marker for the outcome during SARS-CoV-2 infections. Specifically, low plasma IL-3 levels is associated with increased severity, viral load, and mortality during SARS-CoV-2 infections. Patients with severe COVID-19 exhibit also reduced circulating plasmacytoid dendritic cells (pDCs) and low plasma IFNα and IFNλ levels when compared to non-severe COVID-19 patients. In a mouse model of pulmonary HSV-1 infection, treatment with recombinant IL-3 reduces viral load and mortality. Mechanistically, IL-3 increases innate antiviral immunity by promoting the recruitment of circulating pDCs into the airways by stimulating CXCL12 secretion from pulmonary CD123+ epithelial cells, both, in mice and in COVID-19 negative patients exhibiting pulmonary diseases. This study identifies IL-3 as a predictive disease marker for SARS-CoV-2 infections and as a potential therapeutic target for pulmunory viral infections.


Subject(s)
COVID-19/diagnosis , Interleukin-3/blood , Animals , COVID-19/mortality , Chemokine CXCL12/immunology , Dendritic Cells/cytology , Disease Models, Animal , Female , Germany , Humans , Immunity, Innate , Interferons/blood , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Prospective Studies , Severity of Illness Index , T-Lymphocytes/cytology , Viral Load
14.
Rheumatology (Oxford) ; 60(11): 5390-5396, 2021 11 03.
Article in English | MEDLINE | ID: mdl-33580248

ABSTRACT

OBJECTIVES: Heterogeneity of SLE patients in clinical trials remains a challenge for developing new therapies. This study used a combinatorial analysis of four molecular biomarkers to define key sources of heterogeneity. METHODS: Combinations of IFN (high/low), anti-dsDNA (+/-) and C3 and C4 (low/normal) were used to subset n = 1747 patients from two randomized phase III trials. A dichotomous classification scheme defined SLE (+) as: IFN (high), anti-dsDNA (+), C3 (low) and/or C4 (low). SLE (-) required all of the following: IFN (low), anti-dsDNA (-), C3 (normal) and C4 (normal). Additional analyses subset the data further by IFN, anti-dsDNA and complement. RESULTS: The trials enrolled n = 2262 patients of which n = 1747 patients had data for IFN, anti-dsDNA, C3 and C4 at baseline. There were n = 247 patients in the SLE (-) population and n = 1500 patients in the SLE (+) population. The SLE (-) population had more mucocutaneous and musculoskeletal disease at baseline, while SLE (+) had more haematological, renal and vascular involvement. There was lower concomitant medication use in the SLE (-) population for corticosteroids and immunosuppressants, except for MTX. Time to severe flare was significantly longer in SLE (-) vs SLE (+) (P < 0.0001) and SRI-4 response rate was significantly lower in SLE (-) vs SLE (+) (P = 0.00016). The USA had more SLE (-) patients (22%) than Mexico/Central America/South America (10%), Europe (7%) and the rest of the world (5%). CONCLUSION: Combinatorial analysis of four molecular biomarkers revealed subsets of SLE patients that discriminated by disease manifestations, concomitant medication use, geography, time to severe flare and SRI-4 response. These data may be useful for designing clinical trials and identifying subsets of patients for analysis. Rheumatology key messages SLE patients from a P3 trial were categorized by IFN, anti-dsDNA, C3 and C4 status. Patients lacking molecular markers of SLE distinguished from biomarker positive patients on multiple clinical parameters. Biomarker negative patients have distinct disease characteristics that may impact clinical trial outcomes.


Subject(s)
Antibodies, Antinuclear/blood , Complement C3/metabolism , Complement C4/metabolism , Interferons/blood , Lupus Erythematosus, Systemic/diagnosis , Humans , Lupus Erythematosus, Systemic/blood , Phenotype
15.
Eur Rev Med Pharmacol Sci ; 25(1): 459-465, 2021 01.
Article in English | MEDLINE | ID: mdl-33506937

ABSTRACT

OBJECTIVE: To investigate the correlations of Helicobacter pylori (HP) with liver function, inflammatory factors and serum levels of forkhead box P3 (FoxP3) and retinoic acid receptor-related orphan receptor gamma-t (RORγt) in patients with hepatitis B cirrhosis (HBC). PATIENTS AND METHODS: A total of 60 HBC patients were divided into HBC group (n=30) and HP-infected HBC group (HP&HBC group, n=30). QRT-PCR was conducted to determine the messenger ribonucleic acid (mRNA) levels of FoxP3 and RORγt in serum samples. ELISA was applied to measure the levels of relevant inflammatory factors. Besides, immunohistochemical staining was conducted to detect positive expressions of FoxP3 and RORγt in liver tissues of patients in the two groups. RESULTS: No significant differences in gender, drinking, smoking, diabetes and age were found between HBC group and HP&HBC group (p>0.05). Globulin and albumin levels were comparable between the two groups (p>0.05). Liver function indexes, including ALT, AST and TBIL were higher in HP&HBC group than those in HBC group (p<0.05). The HBV-DNA level was lower in HBC group in comparison with that in HP&HBC group. The interferon-gamma (IFN-γ) level was remarkably higher in HBC group than that in HP&HBC group (p<0.01), and the levels of interleukin (IL)-6, IL-10, IL-17 and transforming growth factor (TGF)-ß1 were notably lower in HBC group in comparison with those in HP&HBC group (p<0.01). Additionally, the mRNA levels of FoxP3 and RORγt in HBC group were distinctly lower than those in HP&HBC group (p<0.01). The mRNA levels of FoxP3 and RORγt were positively related to those of IL-6, IL-10, IL-17, and TGF-ß1, and negatively associated with IFN-γ level. Immunohistochemical results indicated that positive expression rates of FoxP3 and RORγt in the liver tissues were approximately 50% in HP&HBC group and B. Zhao, Q.-J. Sheng, Y. Qin, X.-L. Wang, H. Zhao, N. Zhaowere 15% in HBC group, and the difference was statistically significant (p<0.05). CONCLUSIONS: Expression levels of FoxP3 and RORγt in serum and liver tissues are elevated in HP-infected HBC patients, and inflammatory factors are correlated with their expressions, suggesting the aggravated liver damage.


Subject(s)
Helicobacter pylori/metabolism , Hepatitis B/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Adult , Female , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/genetics , Helicobacter pylori/isolation & purification , Hepatitis B/blood , Humans , Interferons/blood , Liver/microbiology , Liver Cirrhosis/blood , Male , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/blood , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , RNA, Messenger/blood , RNA, Messenger/genetics , Transforming Growth Factor beta1/blood
16.
Lupus ; 30(2): 315-319, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33086919

ABSTRACT

OBJECTIVE: To investigate the profiles of anti-RPLP0, anti-galectin3 antibodies, interferon-α (IFN-α), interferon-λ1(IFN-λ1) and interleukin-17A/F(IL-17A/F) in the subtypes of cutaneous lupus erythematosus (CLE) including acute CLE (ACLE), subacute CLE (SCLE) and discoid lupus erythematosus (DLE). METHODS: Serum levels of autoantibodies and cytokines were determined by enzyme-linked immunoabsorbent assay (ELISA). Lupus lesions were evaluated by cutaneous lupus erythematosus disease area and severity index (CLASI). RESULTS: Serum anti-RPLP0, anti-galectin3 antibodies and IFN-λ1 were higher in systemic lupus erythematosus (SLE) patients with skin lesions than those without skin lesions, compared to healthy controls. IFN-α, IL-17A and IL-17F was elevated in all patients regardless of skin lesions. The two antibodies, IFN-α and IL-17A were positively correlated with the CLASI score in all patients with CLE. In addition, serum IL-17A was positively correlated to the CLASI score of ACLE, SCLE and DLE, while anti-RPLP0 and anti-galectin3 antibodies were only correlated to the score of SCLE and IL-17F to DLE. CONCLUSION: Serum anti-RPLP0, anti-galectin3 antibodies, IFN-α, IFN-λ1 and IL-17A/F are associated with the occurrence of lupus skin lesions regardless of the systemic complications, whereas the profiles of these inflammatory mediators vary with the subtypes of lupus skin lesions.


Subject(s)
Autoantibodies/blood , Interferons/blood , Interleukin-17/blood , Lupus Erythematosus, Cutaneous/blood , Lupus Erythematosus, Cutaneous/immunology , Adult , Autoantibodies/immunology , Case-Control Studies , Female , Humans , Immunohistochemistry , Interferons/immunology , Interleukin-17/immunology , Lupus Erythematosus, Discoid/blood , Lupus Erythematosus, Discoid/immunology , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Male , Severity of Illness Index , Skin/immunology , Skin/metabolism
17.
Cytokine ; 138: 155353, 2021 02.
Article in English | MEDLINE | ID: mdl-33121876

ABSTRACT

BACKGROUND: Inflammatory cytokines participate in immune reactions and the pathogenesis of autoimmunity. Herein, we quantified four groups of inflammatory cytokines, including interferons (IFNs), the tumor necrosis factor (TNF) superfamily (TNFSF), interleukin (IL)-related cytokines, and bone and extracellular matrix remodeling-related cytokines to determine their contributions in women with overt Graves' disease (GD). METHODS: Forty-three women with GD were enrolled in this cross-sectional study. Thirty-seven cytokines, thyroid-stimulating hormone (TSH), free thyroxine, and TSH receptor antibody (TSHRAb) were quantified. GD patients with a low TSH level at the time of sample collection were defined as having active GD. RESULTS: Patients with active GD had higher IFN-α2, IFN-γ, IFN-λ1, and IFN-λ2 levels than those with inactive GD. In addition, certain TNFSF cytokines, including soluble cluster of differentiation 30 (sCD30), TNFSF member 14 (TNFSF14), pentraxin (PTX)-3, soluble TNF receptor 2 (sTNF-R2), and thymic stromal lymphopoietin (TSLP) were higher in active GD than in inactive GD. Moreover, active GD patients had higher IL-2, IL-12(p40), osteocalcin (OCN), and matrix metalloproteinase (MMP)-3 than inactive GD patients. All IFNs except IFN-λ1 were correlated with TSHRAb titers. Moreover, TNFSF cytokines, consisting of B-cell-activating factor, sCD30, TNFSF14, PTX-3, sTNF-R2, and TSLP, were associated with TSHRAb levels. CONCLUSIONS: Serum IFNs could be the most remarkable cytokines in modulating the disease severity and TSHRAb titers in women with full-blown GD. Further molecular-based research to clarify the actual role of IFNs in the disease progression of GD is needed.


Subject(s)
Graves Disease/blood , Interferon-alpha/blood , Interferon-gamma/blood , Interferons/blood , Interleukins/blood , Receptors, Thyrotropin/blood , Thyroid Gland/metabolism , Adult , Aged , Autoantibodies/immunology , Bone and Bones/metabolism , Cross-Sectional Studies , Cytokines/blood , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Graves Disease/physiopathology , Humans , Inflammation , Middle Aged , Thyrotropin/blood , Thymic Stromal Lymphopoietin
18.
Clin Exp Immunol ; 204(1): 49-63, 2021 04.
Article in English | MEDLINE | ID: mdl-33336388

ABSTRACT

We investigated Toll-like receptor (TLR)-3/-7/-8/-9 and interferon (IFN)-α/ß/γ mRNA expression in whole blood and serum IFN-α/ß/γ levels in patients with mixed connective tissue disease (MCTD), systemic lupus erythematosus (SLE) and systemic sclerosis (SSc) and in healthy subjects to assess the association between the TLR-IFN expression and severity of and susceptibility to diseases, and identify potential biomarkers. Expression of the IFN-γ, TLR-3 and TLR-8 was detected only in SLE patients. TLR-7, IFN-α and IFN-ß expression was highest in SLE, while TLR-9 expression was highest in SSc patients. In SLE and MCTD patients a strong correlation was observed between TLR-7 and IFN-α expression and IFN-ß and IFN-α expression. In MCTD patients, negative correlation between IFN-α and TLR-9 and TLR-7 and TLR-9 was revealed. TLR-9 expression in anti-U1-70k-negative, anti-C negative and anti-SmB-negative MCTD patients was higher than in MCTD-positive patients. We observed negative correlations between serum IFN-α levels and TLR-7 expression and C3 and C4 levels in SLE patients. In SLE patients we observed that with increased IFN-γ, TLR-3 and TLR-8 expression increased the value of C3 and C4. Our results confirmed that the endosomal TLR-IFN pathway seems to be more important in SLE than in MCTD or SSc, and that IFN-α and IFN-ß may be possible biomarkers for SLE.


Subject(s)
Gene Expression Profiling/methods , Interferons/genetics , Lupus Erythematosus, Systemic/genetics , Mixed Connective Tissue Disease/genetics , Scleroderma, Systemic/genetics , Toll-Like Receptors/genetics , Adult , Aged , Aged, 80 and over , Endosomes/genetics , Endosomes/metabolism , Female , Humans , Interferon-alpha/blood , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-beta/blood , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/blood , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferons/blood , Interferons/metabolism , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/metabolism , Male , Middle Aged , Mixed Connective Tissue Disease/blood , Mixed Connective Tissue Disease/metabolism , Scleroderma, Systemic/blood , Scleroderma, Systemic/metabolism , Toll-Like Receptor 3/blood , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/blood , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/blood , Toll-Like Receptor 8/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 9/blood , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/blood , Toll-Like Receptors/metabolism , Young Adult
19.
Front Immunol ; 11: 580987, 2020.
Article in English | MEDLINE | ID: mdl-33193384

ABSTRACT

Covid-19 features a delayed onset of critical illness occurring approximately one week from the beginning of symptoms, which corresponds to the bridging of innate and adaptive immunity. We reasoned that the immune events occurring at the turning point of disease might mark the direction toward pathogenic versus protective inflammatory responses. Subjects with either severe (s; PaO2/FiO2 ratio <200) or mild (m; PaO2/FiO2 ratio>300) Covid-19 were enrolled. A range of chemokines and cytokines as well as reactive oxygen species (ROS) were measured in plasma. Dendritic and NK cell frequency, monocyte and B-/T-cell phenotype and SARS-CoV-2-specific T-cell responses were assessed in PBMC. Twenty mCovid-19 and 20 sCovid-19 individuals were studied. sCovid-19 patients displayed higher non-classical monocytes, plasma chemokines (CXCL8, CXCL9, CXCL10), cytokines (IL-6, IL-10), and ROS versus mCovid-19. sCovid-19 also showed significantly increased activated CD38+HLA-DR+ T-lymphocyte, and granzyme-B+/perforin+ pro-cytolytic T-cells. All Covid-19 patients showed SARS-CoV-2 specific-T-cell response with a predominance of Th1 bi- or trifunctional IFN-γ/IL-2/TNF-α-expressing CD4+, while no difference according to disease severity was observed. Severe Covid-19 features heightened circulating IFN-inducible chemokines and activated pro-cytolytic Th1 cell phenotype in the second week of illness, yet SARS-CoV-2-specific responses are similar to that of mild illness. Altogether, our observations suggest Th1 polarization coupled to higher cytolytic profile in sCovid-19 as correlate of disease pathogenesis and as potential targets to be investigated in the roadmap to therapy and vaccine development.


Subject(s)
COVID-19/blood , Chemokines/blood , Interferons/blood , Leukocytes, Mononuclear/immunology , SARS-CoV-2/physiology , Th1 Cells/immunology , Aged , COVID-19/immunology , COVID-19/virology , Female , Humans , Male , Middle Aged , Phenotype , SARS-CoV-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...