Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 715
Filter
1.
Discov Med ; 36(186): 1386-1397, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054710

ABSTRACT

BACKGROUND: More and more evidence has shown the process of Parkinson's disease (PD). Probably, inflammation exerts a crucial role between them. Therefore, the aim of this study was to analyze the impact of interleukin-6 receptor (IL-6R) expression on the IL-6/signal transducer and activator of transcription 3 (STAT3)/hypoxia-inducible factor-1α (HIF-1α) inflammatory signaling pathway within a mouse model of PD with type 2 diabetes mellitus (T2DM) as co-morbidity. METHODS: We chose healthy wild-type C57BL/6J male mice at the age of 10 weeks to prepare a mouse model of PD with T2DM co-morbidity. Adeno-associated virus (AAV) overexpressing IL-6R or AAV IL-6R-shRNA genes were injected into the substantia nigra (SN) of the mice. The behavioral indices of the pole test were used for examining the motor function of the mice. Using immunofluorescence analysis, the impacts of IL-6R on the level of tyrosine hydroxylase (TH) and anti-ionized calcium-binding adaptor molecule 1 (IBA-1) on dopaminergic neurons and microglia were examined. Additionally, enzyme-linked immunosorbent assay (ELISA) was adopted for determining the expressions of HIF-1α and inflammatory cytokines like tumor necrosis factor-α (TNF-α), IL-1ß, IL-6, and IL-4 in the serum. In this study, the protein expression levels of TH, α-Synuclein (α-Syn), IBA-1, IL-6, IL-6R, phosphorylated and total signal transducer and activator of transcription 3 (p-STAT3 (Tyr705) and STAT3) and HIF-1α in the SN were tested via western blotting. To ascertain the mRNA expressions of TNF-α, IL-1ß, IL-6, IL-4, and HIF-1α, we used quantitative Real-Time Polymerase Chain Reaction (RT-qPCR). RESULTS: IL-6R-shRNA treatment could markedly shorten the total time of PD in the T2DM co-morbidity mouse model based on the pole test results, reverse the decrease in TH-positive neurons stimulated by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP), and lower the activation of microglia (all p < 0.05). Further, IL-6R-shRNA treatment hindered the expression of IL-6, p-STAT3 (Tyr705), and HIF-1α in the SN, lowered the levels of TNF-α, IL-1ß, IL-6, IL-4, and HIF-1α in the serum, and mRNA expressions of TNF-α, IL-1ß, IL-6, and HIF-1α in the SN (all p < 0.05). In contrast, IL-6R overexpression reduced TH levels, upregulated the level of IBA-1, IL-6, p-STAT3 (Tyr705), and HIF-1α, increased the level of IL-1ß, TNF-α, IL-6, IL-4, and HIF-1α (all p < 0.05) in the serum and SN in the PD mouse model with T2DM as a co-morbidity. CONCLUSIONS: PD progression with T2DM as a co-morbidity can be boosted by AAV IL-6R-overexpression through upregulation of the IL-6/STAT3/HIF-1α axis. Conversely, AAV IL-6R-shRNA treatment suppressed the IL-6/STAT3/HIF-1α pathway and alleviated neuroinflammation, thus weakening the development of PD with T2DM as a co-morbidity.


Subject(s)
Diabetes Mellitus, Type 2 , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-6 , Mice, Inbred C57BL , Parkinson Disease , Receptors, Interleukin-6 , STAT3 Transcription Factor , Signal Transduction , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Interleukin-6/metabolism , Male , Mice , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Parkinson Disease/metabolism , Parkinson Disease/genetics
2.
BMC Cardiovasc Disord ; 24(1): 365, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014329

ABSTRACT

BACKGROUND: M1 macrophages are closely associated with cardiac injury after myocardial infarction (MI). Increasing evidence shows that exosomes play a key role in pathophysiological regulation after MI, but the role of M1 macrophage-derived exosomes (M1-Exos) in myocardial regeneration remains unclear. In this study, we explored the impact of M1 macrophage-derived exosomes on cardiomyocytes regeneration in vitro and in vivo. METHODS: M0 macrophages were induced to differentiate into M1 macrophages with GM-CSF (50 ng/mL) and IFN-γ (20 ng/mL). Then M1-Exos were isolated and co-incubated with cardiomyocytes. Cardiomyocyte proliferation was detected by pH3 or ki67 staining. Quantitative real-time PCR (qPCR) was used to test the level of miR-155 in macrophages, macrophage-derived exosomes and exosome-treated cardiomyocytes. MI model was constructed and LV-miR-155 was injected around the infarct area, the proliferation of cardiomyocytes was counted by pH3 or ki67 staining. The downstream gene and pathway of miR-155 were predicted and verified by dual-luciferase reporter gene assay, qPCR and immunoblotting analysis. IL-6 (50 ng/mL) was added to cardiomyocytes transfected with miR-155 mimics, and the proliferation of cardiomyocytes was calculated by immunofluorescence. The protein expressions of IL-6R, p-JAK2 and p-STAT3 were detected by Western blot. RESULTS: The results showed that M1-Exos suppressed cardiomyocytes proliferation. Meanwhile, miR-155 was highly expressed in M1-Exos and transferred to cardiomyocytes. miR-155 inhibited the proliferation of cardiomyocytes and antagonized the pro-proliferation effect of interleukin 6 (IL-6). Furthermore, miR-155 targeted gene IL-6 receptor (IL-6R) and inhibited the Janus kinase 2(JAK)/Signal transducer and activator of transcription (STAT3) signaling pathway. CONCLUSION: M1-Exos inhibited cardiomyocyte proliferation by delivering miR-155 and inhibiting the IL-6R/JAK/STAT3 signaling pathway. This study provided new insight and potential treatment strategy for the regulation of myocardial regeneration and cardiac repair by macrophages.


Subject(s)
Cell Proliferation , Disease Models, Animal , Exosomes , Janus Kinase 2 , Macrophages , MicroRNAs , Myocardial Infarction , Myocytes, Cardiac , STAT3 Transcription Factor , Signal Transduction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/transplantation , Exosomes/genetics , Animals , Cell Proliferation/drug effects , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/genetics , Janus Kinase 2/metabolism , Male , Regeneration , Rats, Sprague-Dawley , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Cells, Cultured , Phosphorylation , Coculture Techniques , Mice, Inbred C57BL , Interleukin-6/metabolism
3.
J Cell Mol Med ; 28(13): e18515, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961677

ABSTRACT

There is a close relationship between immune-mediated inflammation and cancer, and there is still controversy over whether rheumatoid arthritis (RA) increases the risk of malignancy. We first used Mendelian randomization (MR) analysis to explore the potential causal relationship between RA and pan-cancer. And verify the effect of immune-mediated inflammation on cancer through intermediate MR analysis. Then we extracted the standardized incidence rate of malignancy in RA patients relative to the general population through large-scale meta-analysis. Finally, we performed pan-cancer analysis on the RA related genes obtained from MR analysis. And perform immune related analysis on key genes to reveal the association between RA and malignancy. The MR analysis demonstrated a negative correlation between RA and pan-cancer (p = 0.008). Autoimmune traits were the main mediating variable for the causal relationship between RA and pan-cancer. Based on the results of the meta-analysis, we validated that RA reduces the risk of developing colorectal cancer (SIR = 0.69, 95% CI 0.53-0.85). Pan-cancer analysis also showed that high expression of RA related genes was negatively correlated with colon adenocarcinoma. IL6R was the gene with the highest correlation among them, and its correlation with immune cells was higher in colorectal cancer than in other malignancy. Our MR study provides evidence that RA was associated with reduced risk of colorectal cancer. This effect is caused by immune-mediated inflammation, with IL6R being a key regulatory gene.


Subject(s)
Arthritis, Rheumatoid , Colorectal Neoplasms , Inflammation , Mendelian Randomization Analysis , Humans , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/complications , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Inflammation/genetics , Inflammation/complications , Inflammation/immunology , Risk Factors , Genetic Predisposition to Disease , Receptors, Interleukin-6/genetics
4.
Genes (Basel) ; 15(7)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39062668

ABSTRACT

OBJECTIVES: Interleukin 6 (IL-6) levels at hospital admission have been suggested for disease prognosis, and IL-6 antagonists have been suggested for the treatment of patients with severe COVID-19. However, less is known about the relationship between pre-COVID-19 IL-6 levels and the risk of severe COVID-19. To fill in this gap, here we extensively investigated the association of genetically instrumented IL-6 pathway components with the risk of severe COVID-19. METHODS: We used a two-sample Mendelian randomization study design and retrieved genetic instruments for blood biomarkers of IL-6 activation, including IL-6, soluble IL-6 receptor, IL-6 signal transducer, and CRP, from respective large available GWASs. To establish associations of these instruments with COVID-19 outcomes, we used data from the Host Genetics Initiative and GenOMICC studies. RESULTS: Our analyses revealed inverse associations of genetically instrumented levels of IL-6 and its soluble receptor with the risk of developing severe disease (OR = 0.60 and 0.94, respectively). They also demonstrated a positive association of severe disease with the soluble signal transducer level (OR = 1.13). Only IL-6 associations with severe COVID-19 outcomes reached the significance threshold corrected for multiple testing (p < 0.003; with COVID-19 hospitalization and critical illness). CONCLUSIONS: These potential causal relationships for pre-COVID-19 IL-6 levels with the risk of developing severe symptoms provide opportunities for further evaluation of these factors as prognostic/preventive markers of severe COVID-19. Further studies will need to clarify whether the higher risk for a severe disease course with lower baseline IL-6 levels may also extend to other infectious diseases.


Subject(s)
COVID-19 , Interleukin-6 , Mendelian Randomization Analysis , Receptors, Interleukin-6 , SARS-CoV-2 , Humans , Interleukin-6/blood , Interleukin-6/genetics , COVID-19/genetics , COVID-19/blood , COVID-19/virology , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/blood , Biomarkers/blood , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Signal Transduction/genetics
5.
Front Immunol ; 15: 1374967, 2024.
Article in English | MEDLINE | ID: mdl-38881895

ABSTRACT

Background: Cholangiocarcinoma (CCA) is a typical inflammation-induced malignancy, and elevated serum interleukin-6 (IL-6) levels have been reported to be linked to the onset and progression of CCA. We aim to investigate the potential prognostic value of the IL-6 pathway for CCA. Methods: We detected the expressions of IL-6, IL-6R, glycoprotein (gp130), C-reactive protein (CRP), Janus kinase 2 (JAK2), and signal transducer and activator of transcription 3 (STAT3) in CCA tissue microarray using multiplex immunofluorescence. Furthermore, the clinical associations and prognostic values were assessed. Finally, single-cell transcriptome analysis was performed to evaluate the expression level of IL-6 pathway genes in CCA. Results: The results revealed that the expression of IL-6 was lower, while the expression of STAT3 was higher in tumor tissues compared to normal tissues. Especially in tumor microenvironment, the expression of IL-6 pathway genes was generally downregulated. Importantly, gp130 was strongly correlated with JAK2 in tumor tissues, while it was moderately correlated with JAK2 in normal tissue. Although none of the gene expressions were directly associated with overall survival and disease-free survival, our study found that IL-6, IL-6R, CRP, gp130, and JAK2 were inversely correlated with vascular invasion, which is a risk factor for poor prognosis in patients with CCA. Conclusion: The findings from this study suggest that the IL-6 signaling pathway may have a potential prognostic value for CCA. Further investigation is needed to understand the underlying molecular mechanisms of the IL-6 pathway in CCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Cytokine Receptor gp130 , Interleukin-6 , Janus Kinase 2 , STAT3 Transcription Factor , Signal Transduction , Humans , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Male , Female , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Middle Aged , Prognosis , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Gene Expression Regulation, Neoplastic , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Aged , Biomarkers, Tumor/genetics , Gene Expression Profiling , Clinical Relevance
6.
Emerg Microbes Infect ; 13(1): 2366359, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38855910

ABSTRACT

Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.


Subject(s)
Coinfection , Granuloma , HIV Infections , Lung , Macrophages , Receptors, Interleukin-6 , STAT3 Transcription Factor , Humans , Coinfection/virology , Coinfection/immunology , Coinfection/microbiology , HIV Infections/complications , HIV Infections/immunology , Macrophages/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Granuloma/immunology , Lung/pathology , Lung/immunology , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, CD/metabolism , Antigens, CD/genetics , Signal Transduction , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/complications , Male , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/complications , Female , Adult , Interleukin-6/metabolism , Interleukin-6/genetics , CD68 Molecule
7.
J Transl Med ; 22(1): 581, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898459

ABSTRACT

Dysregulation of inflammation can lead to multiple chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD) and asthma. Interleukin-6 (IL6) is crucial in regulating the inflammatory cascade, but the causal link between IL6 signaling downregulation and respiratory diseases risk is unclear. This study uses Mendelian randomization to examine the effects of IL6R blockade on respiratory diseases. Analyzing data from 522,681 Europeans, 26 genetic variants were obtained to mimic IL6R inhibition. Our findings show that IL6R blockade significantly reduces the risk of COPD (OR = 0.71, 95% CI = 0.60-9.84) and asthma (OR = 0.82, 95% CI = 0.74-0.90), with protective trends for bronchitis, pulmonary embolism, and lung cancer. Results were consistent across methods, with no significant heterogeneity or pleiotropy. These insights suggest IL6R downregulation as a potential therapeutic target for respiratory diseases, meriting further clinical investigation.


Subject(s)
Receptors, Interleukin-6 , Signal Transduction , Humans , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Signal Transduction/genetics , Genetic Predisposition to Disease , Risk Factors , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Mendelian Randomization Analysis , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/metabolism , Asthma/genetics , Respiration Disorders/genetics
8.
Circ Genom Precis Med ; 17(3): e004374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38752343

ABSTRACT

BACKGROUND: The immune system's role in ST-segment-elevated myocardial infarction (STEMI) remains poorly characterized but is an important driver of recurrent cardiovascular events. While anti-inflammatory drugs show promise in reducing recurrence risk, their broad immune system impairment may induce severe side effects. To overcome these challenges, a nuanced understanding of the immune response to STEMI is needed. METHODS: For this, we compared peripheral blood mononuclear single-cell RNA-sequencing (scRNA-seq) and plasma protein expression over time (hospital admission, 24 hours, and 6-8 weeks post-STEMI) in 38 patients and 38 controls (95 995 diseased and 33 878 control peripheral blood mononuclear cells). RESULTS: Compared with controls, classical monocytes were increased and CD56dim natural killer cells were decreased in patients with STEMI at admission and persisted until 24 hours post-STEMI. The largest gene expression changes were observed in monocytes, associating with changes in toll-like receptor, interferon, and interleukin signaling activity. Finally, a targeted cardiovascular biomarker panel revealed expression changes in 33/92 plasma proteins post-STEMI. Interestingly, interleukin-6R, MMP9 (matrix metalloproteinase-9), and LDLR (low-density lipoprotein receptor) were affected by coronary artery disease-associated genetic risk variation, disease status, and time post-STEMI, indicating the importance of considering these aspects when defining potential future therapies. CONCLUSIONS: Our analyses revealed the immunologic pathways disturbed by STEMI, specifying affected cell types and disease stages. Additionally, we provide insights into patients expected to benefit most from anti-inflammatory treatments by identifying the genetic variants and disease stage at which these variants affect the outcome of these (drug-targeted) pathways. These findings advance our knowledge of the immune response post-STEMI and provide guidance for future therapeutic studies.


Subject(s)
Single-Cell Analysis , Humans , Male , Female , Middle Aged , ST Elevation Myocardial Infarction/immunology , ST Elevation Myocardial Infarction/genetics , ST Elevation Myocardial Infarction/blood , Aged , Monocytes/immunology , Monocytes/metabolism , Biomarkers/blood , Myocardial Infarction/immunology , Myocardial Infarction/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Case-Control Studies
9.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732195

ABSTRACT

Sport injuries, including the anterior crucial ligament rupture (ACLR) seem to be related to complex genetic backgrounds, including the genes responsible for inflammatory response. This review and meta-analysis investigated the contribution of the polymorphisms of genes encoding inflammatory cytokines and their receptors to the risk of ACLR. The scientific databases Science Direct, EBSCO host, Scopus, PubMed, and Google Scholar were screened (completed on 14 June 2023) according to the established inclusion/exclusion criteria (only fully accessible, original, human case-control studies written in English concerning the effect of interleukin genes' polymorphisms on the occurrence of ACL injury were included) and statistical meta-analysis using R version 4.0.3 was performed. The PRISMA methodology was used to review articles. The review protocol was registered under the number CRD42024514316 in the Prospero database. Eighty-nine studies were identified and narrowed down to three original case-control studies used for the meta-analysis. The studies analyzed Polish, South African, and Swedish cohorts, altogether 1282 participants. The candidate polymorphisms indicated in the studies involved IL6 rs1800795, IL6R rs2228145 and IL1B rs16944. The systematic review showed the relationships between IL6 rs1800795 polymorphism and ACLR in the Polish subpopulation, and IL6R rs2228145 and IL1B rs16944 in the South African subpopulations. The meta-analysis revealed that the IL6 rs1800795 CG genotype was over-represented (OR = 1.30, 95% CI 1.02-1.66), while the CC genotype was under-represented (OR = 0.75, 95% CI 0.54-1.03) in ACLR subjects, but no significant impact of IL6R rs2228145 was shown. Additionally, a tendency of the IL1B rs16944 CT genotype to be protective (OR 0.89, 95% CI 0.70-1.14), while the TT to be a risk genotype (OR 1.19, 95% CI 0.84-1.68) was observed. Thus, the relationship between the interleukin receptor IL6R rs2228145 and ACLR risk was not confirmed. However, the impact of genes coding pleiotropic IL6 rs1800795 on the incidences of ACLR was clear and the effect of pro-inflammatory IL1B rs16944 was possible.


Subject(s)
Anterior Cruciate Ligament Injuries , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Humans , Anterior Cruciate Ligament Injuries/genetics , Interleukin-6/genetics , Interleukin-1beta/genetics , Receptors, Interleukin-6/genetics , Interleukins/genetics , Risk Factors , Case-Control Studies
10.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791322

ABSTRACT

A keloid is a benign fibroproliferative hypertrophy of scar tissue that extends outside the original wound and invades adjacent healthy skin. Keloid formation is thought to be a complex process including overactivity of the interleukin-6 signaling pathway and genetic susceptibility. The aim of the study was to investigate possible associations between rs1800797, rs1800796, and rs1800795 polymorphisms in the promoter of the IL6 gene encoding interleukin-6 and the rs2228145 polymorphism in the IL6R gene encoding the interleukin-6 receptor subunit alpha with the predisposition to keloids in Polish patients. The genetic polymorphisms were identified either using Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) or sequencing of samples of genomic DNA extracted from blood leukocytes of 86 adult patients with keloids and 100 newborns comprising a control group. No significant differences in the distributions of IL6 or IL6R alleles or genotypes were found between keloid patients and newborn controls. There were also no significant differences between both groups in the distribution of IL6 haplotypes. The IL6 rs1800797, rs1800796 and rs1800795 and IL6R rs2228145 polymorphisms were not found to predispose individuals in the study group to keloids. IL6 promoter haplotypes were not found to be associated with a higher risk of keloids in the studied group.


Subject(s)
Genetic Predisposition to Disease , Interleukin-6 , Keloid , Polymorphism, Single Nucleotide , Receptors, Interleukin-6 , Humans , Keloid/genetics , Keloid/pathology , Interleukin-6/genetics , Receptors, Interleukin-6/genetics , Male , Female , Adult , Poland , Middle Aged , Promoter Regions, Genetic , Case-Control Studies , Haplotypes , Alleles , Adolescent , Young Adult , Gene Frequency , Genotype , Infant, Newborn , Genetic Association Studies
11.
Nutr J ; 23(1): 51, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750566

ABSTRACT

BACKGROUND: Previous research has extensively examined the role of interleukin 6 (IL-6) in sarcopenia. However, the presence of a causal relationship between IL-6, its receptor (IL-6R), and sarcopenia remains unclear. METHOD: In this study, we utilized summary-level data from genome-wide association studies (GWAS) focused on appendicular lean mass (ALM), hand grip strength, and walking pace. Single nucleotide polymorphisms (SNPs) were employed as genetic instruments for IL-6 and IL-6R to estimate the causal effect of sarcopenia traits. We adopted the Mendelian randomization (MR) approach to investigate these associations using the inverse variance weighted (IVW) method as the primary analytical approach. Additionally, we performed sensitivity analyses to validate the reliability of the MR results. RESULT: This study revealed a significant negative association between main IL-6R and eQTL IL-6R on the left grip strength were - 0.013 (SE = 0.004, p < 0.001) and -0.029 (SE = 0.007, p < 0.001), respectively. While for the right grip strength, the estimates were - 0.011 (SE = 0.001, p < 0.001) and - 0.021 (SE = 0.008, p = 0.005). However, no evidence of an association for IL-6R with ALM and walking pace. In addition, IL-6 did not affect sarcopenia traits. CONCLUSION: Our study findings suggest a negative association between IL-6R and hand grip strength. Additionally, targeting IL-6R may hold potential value as a therapeutic approach for the treatment of hand grip-related issues.


Subject(s)
Genome-Wide Association Study , Hand Strength , Interleukin-6 , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Receptors, Interleukin-6 , Sarcopenia , Humans , Interleukin-6/genetics , Interleukin-6/blood , Sarcopenia/genetics , Mendelian Randomization Analysis/methods , Receptors, Interleukin-6/genetics , Hand Strength/physiology , Genome-Wide Association Study/methods
12.
BMC Ophthalmol ; 24(1): 228, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822340

ABSTRACT

BACKGROUND: Several autoimmune disorders have been linked to polymorphisms in IL10 and IL6R genes. This research aimed to study whether single nucleotide polymorphisms (SNPs) in the genes of IL10 and IL6R were associated with acute anterior uveitis (AAU) in Han Chinese. METHODS: Genotyping was carried out by the iPLEX Gold Genotyping Assay. Our study comprised 420 patients with AAU and 918 healthy subjects from Han Chinese. Using the chi-square (χ2) test, alleles and genotypes were analyzed between AAU subjects and healthy controls. RESULTS: All ten SNPs were successfully genotyped and four SNPs (IL10/rs1800871, IL10/rs3021094, IL10/rs2222202, IL6R/rs4845618) exhibited weak associations with AAU, as indicated by their Puncorr values. However, upon applying the Bonferroni correction, there was no significant association between AAU and the control subjects. Additionally, the haplotype analysis of the ten SNPs revealed no association with AAU. CONCLUSION: Our findings suggested that polymorphisms of the tested ten SNPs on the IL10 and IL6R genes did not show any association with the risk of developing AAU among the Han Chinese population.


Subject(s)
Asian People , Genetic Predisposition to Disease , Genotype , Interleukin-10 , Polymorphism, Single Nucleotide , Receptors, Interleukin-6 , Uveitis, Anterior , Humans , Uveitis, Anterior/genetics , Male , Interleukin-10/genetics , Female , Receptors, Interleukin-6/genetics , Adult , China/epidemiology , Acute Disease , Middle Aged , Asian People/genetics , Case-Control Studies , Gene Frequency , Young Adult , Alleles , Haplotypes , Aged , East Asian People
13.
Sci Rep ; 14(1): 8021, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38580710

ABSTRACT

The Phenome-Wide Association Study (PheWAS) is increasingly used to broadly screen for potential treatment effects, e.g., IL6R variant as a proxy for IL6R antagonists. This approach offers an opportunity to address the limited power in clinical trials to study differential treatment effects across patient subgroups. However, limited methods exist to efficiently test for differences across subgroups in the thousands of multiple comparisons generated as part of a PheWAS. In this study, we developed an approach that maximizes the power to test for heterogeneous genotype-phenotype associations and applied this approach to an IL6R PheWAS among individuals of African (AFR) and European (EUR) ancestries. We identified 29 traits with differences in IL6R variant-phenotype associations, including a lower risk of type 2 diabetes in AFR (OR 0.96) vs EUR (OR 1.0, p-value for heterogeneity = 8.5 × 10-3), and higher white blood cell count (p-value for heterogeneity = 8.5 × 10-131). These data suggest a more salutary effect of IL6R blockade for T2D among individuals of AFR vs EUR ancestry and provide data to inform ongoing clinical trials targeting IL6 for an expanding number of conditions. Moreover, the method to test for heterogeneity of associations can be applied broadly to other large-scale genotype-phenotype screens in diverse populations.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Genetic Association Studies , Phenotype , Polymorphism, Single Nucleotide , Receptors, Interleukin-6/genetics
14.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167151, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565387

ABSTRACT

Immunoglobulin A vasculitis (IgAV) is the most common vasculitis of childhood. Disordered immune responses play important roles in its pathogenesis, but the comprehensive immune profile of the disease and the underlying mechanisms are still largely unknown. Here we found a potential disease biomarker cold inducible RNA binding protein (CIRP) in our pediatric IgAV cohort. Serum CIRP level in these patients were elevated and positively correlated with the increased early memory (CD45RA+CD62L+CD95+) T cells revealed using multicolor flow cytometry. Immune phenotyping of the patients showed they had more activated T cells with higher IL6Ra expression. T cell culture experiment showed CIRP further activated both human CD4+ and CD8+ T cells as indicated by increased perforin secretion and phosphorylation of STAT3. Blockade of IL6Rα attenuated CIRP-induced T cell toxicity in vitro. RNA-sequencing data further supported CIRP stimulation promoted human T cell activation and migration, fueled inflammation through the JAK-STAT signaling pathway. Therefore, IL6Ra-mediated T cell activation by extracellular CIRP may contribute to pathogenesis of IgAV in children, both CIRP and IL6Ra could be new therapeutic targets for IgAV.


Subject(s)
Lymphocyte Activation , RNA-Binding Proteins , Receptors, Interleukin-6 , STAT3 Transcription Factor , Adolescent , Child , Female , Humans , Male , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Hepatitis A Virus Cellular Receptor 2 , IgA Vasculitis/immunology , IgA Vasculitis/pathology , IgA Vasculitis/metabolism , Lymphocyte Activation/immunology , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/immunology , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
J Biol Chem ; 300(5): 107251, 2024 May.
Article in English | MEDLINE | ID: mdl-38569939

ABSTRACT

Ciliary neurotrophic factor (CNTF) activates cells via the non-signaling α-receptor CNTF receptor (CNTFR) and the two signaling ß-receptors glycoprotein 130 (gp130) and leukemia inhibitory factor receptor (LIFR). The CNTF derivate, Axokine, was protective against obesity and insulin resistance, but clinical development was halted by the emergence of CNTF antibodies. The chimeric cytokine IC7 used the framework of interleukin (IL-)6 with the LIFR-binding site from CNTF to activate cells via IL-6R:gp130:LIFR complexes. Similar to CNTF/Axokine, IC7 protected mice from obesity and insulin resistance. Here, we developed CNTF-independent chimeras that specifically target the IL-6R:gp130:LIFR complex. In GIL-6 and GIO-6, we transferred the LIFR binding site from LIF or OSM to IL-6, respectively. While GIO-6 signals via gp130:IL-6R:LIFR and gp130:IL-6R:OSMR complexes, GIL-6 selectively activates the IL-6R:gp130:LIFR receptor complex. By re-evaluation of IC7 and CNTF, we discovered the Oncostatin M receptor (OSMR) as an alternative non-canonical high-affinity receptor leading to IL-6R:OSMR:gp130 and CNTFR:OSMR:gp130 receptor complexes, respectively. The discovery of OSMR as an alternative high-affinity receptor for IC7 and CNTF designates GIL-6 as the first truly selective IL-6R:gp130:LIFR cytokine, whereas GIO-6 is a CNTF-free alternative for IC7.


Subject(s)
Ciliary Neurotrophic Factor , Cytokine Receptor gp130 , Interleukin-6 , Signal Transduction , Animals , Humans , Mice , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/genetics , Cytokine Receptor gp130/metabolism , Cytokine Receptor gp130/genetics , Interleukin-6/metabolism , Interleukin-6/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Models, Molecular , Protein Engineering/methods , Protein Structure, Tertiary , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Receptors, OSM-LIF/metabolism , Receptors, OSM-LIF/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Mice, Inbred C57BL
16.
Nat Commun ; 15(1): 2071, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38453915

ABSTRACT

IL-11 and IL-6 activate signalling via assembly of the cell surface receptor gp130; however, it is unclear how signals are transmitted across the membrane to instruct cellular responses. Here we solve the cryoEM structure of the IL-11 receptor recognition complex to discover how differences in gp130-binding interfaces may drive signalling outcomes. We explore how mutations in the IL6ST gene encoding for gp130, which cause severe immune deficiencies in humans, impair signalling without blocking cytokine binding. We use cryoEM to solve structures of both IL-11 and IL-6 complexes with a mutant form of gp130 associated with human disease. Together with molecular dynamics simulations, we show that the disease-associated variant led to an increase in flexibility including motion within the cytokine-binding core and increased distance between extracellular domains. However, these distances are minimized as the transmembrane helix exits the membrane, suggesting a stringency in geometry for signalling and dimmer switch mode of action.


Subject(s)
Interleukin-11 , Interleukin-6 , Humans , Interleukin-11/genetics , Interleukin-6/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Signal Transduction , Receptors, Interleukin-6/genetics
18.
Proc Natl Acad Sci U S A ; 121(2): e2315898120, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165930

ABSTRACT

Protection against endothelial damage is recognized as a frontline approach to preventing the progression of cytokine release syndrome (CRS). Accumulating evidence has demonstrated that interleukin-6 (IL-6) promotes vascular endothelial damage during CRS, although the molecular mechanisms remain to be fully elucidated. Targeting IL-6 receptor signaling delays CRS progression; however, current options are limited by persistent inhibition of the immune system. Here, we show that endothelial IL-6 trans-signaling promoted vascular damage and inflammatory responses via hypoxia-inducible factor-1α (HIF1α)-induced glycolysis. Using pharmacological inhibitors targeting HIF1α activity or mice with the genetic ablation of gp130 in the endothelium, we found that inhibition of IL-6R (IL-6 receptor)-HIF1α signaling in endothelial cells protected against vascular injury caused by septic damage and provided survival benefit in a mouse model of sepsis. In addition, we developed a short half-life anti-IL-6R antibody (silent anti-IL-6R antibody) and found that it was highly effective at augmenting survival for sepsis and severe burn by strengthening the endothelial glycocalyx and reducing cytokine storm, and vascular leakage. Together, our data advance the role of endothelial IL-6 trans-signaling in the progression of CRS and indicate a potential therapeutic approach for burns and sepsis.


Subject(s)
Cytokine Receptor gp130 , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-6 , Receptors, Interleukin-6 , Sepsis , Animals , Mice , Cytokine Receptor gp130/genetics , Cytokine Release Syndrome , Endothelial Cells , Receptors, Interleukin-6/genetics , Sepsis/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
20.
FEBS J ; 291(8): 1667-1683, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37994264

ABSTRACT

Interleukin-11 (IL-11) is a member of the IL-6 family of cytokines and is an important factor for bone homeostasis. IL-11 binds to and signals via the membrane-bound IL-11 receptor (IL-11R, classic signaling) or soluble forms of the IL-11R (sIL-11R, trans-signaling). Mutations in the IL11RA gene, which encodes the IL-11R, are associated with craniosynostosis, a human condition in which one or several of the sutures close prematurely, resulting in malformation of the skull. The biological mechanisms of how mutations within the IL-11R are linked to craniosynostosis are mostly unexplored. In this study, we analyze two variants of the IL-11R described in craniosynostosis patients: p.T306_S308dup, which results in a duplication of three amino-acid residues within the membrane-proximal fibronectin type III domain, and p.E364_V368del, which results in a deletion of five amino-acid residues in the so-called stalk region adjacent to the plasma membrane. The stalk region connects the three extracellular domains to the transmembrane and intracellular region of the IL-11R and contains cleavage sites for different proteases that generate sIL-11R variants. Using a combination of bioinformatics and different biochemical, molecular, and cell biology methods, we show that the IL-11R-T306_S308dup variant does not mature correctly, is intracellularly retained, and does not reach the cell surface. In contrast, the IL-11R-E364_V368del variant is fully biologically active and processed normally by proteases, thus allowing classic and trans-signaling of IL-11. Our results provide evidence that mutations within the IL11RA gene may not be causative for craniosynostosis and suggest that other regulatory mechanism(s) are involved but remain to be identified.


Subject(s)
Craniosynostoses , Interleukin-11 , Humans , Receptors, Interleukin-11/genetics , Receptors, Interleukin-11/chemistry , Receptors, Interleukin-11/metabolism , Interleukin-11/genetics , Interleukin-11/metabolism , Signal Transduction , Craniosynostoses/genetics , Peptide Hydrolases/metabolism , Receptors, Interleukin-6/genetics , Cytokine Receptor gp130/genetics
SELECTION OF CITATIONS
SEARCH DETAIL