1
|
Kuriakose D, Zhu HM, Zhao YL, Iraqi FA, Morahan G, Xiao ZC. Upstream regulation of microRNA-9 through a complex cellular machinery during neurogenesis. Brain Res 2025; 1848:149328. [PMID: 39547498 DOI: 10.1016/j.brainres.2024.149328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/16/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
While microRNAs (miRs) like miR-9 are crucial for neurogenesis and neuronal differentiation, their regulatory mechanisms are not well understood. miR-9 is highly expressed in the brain and plays a significant role in neurogenesis. Using the Collaborative Cross resource, we identified significant quantitative trait loci (QTL) through genetic analyses. We then characterized over 130 candidate genes within these QTL regions using RNA interference, qPCR, and neuronal differentiation assays, narrowing them down to 13 promising candidates. Among these, Panx2, Polr1c, and Mgea5 were found to colocalize in the neurogenic niches of the SVZ and DG regions, as shown by immunofluorescence. Further ChIP-seq and Co-IP analyses revealed their interaction and binding to the miR-9 locus, forming a DNA-protein regulatory complex we termed 'miRSome-9.' A 3C/ChIP-loop assay confirmed the chromatin organization of miRSome-9 at the miR-9 locus, shedding light on the upstream mechanisms regulating miR-9 expression during neurogenesis.
Collapse
Affiliation(s)
- Diji Kuriakose
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic 3800, Australia.
| | - Hong-Mei Zhu
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Yi-Ling Zhao
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Fuad A Iraqi
- Department of Human Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, University of Western Australia of Medical Research, Perth, Australia
| | - Zhi-Cheng Xiao
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic 3800, Australia; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China.
| |
Collapse
|
2
|
Urahashi M, Fujimoto T, Inoue-Mochita M, Inoue T. Effect of the IL-6 trans-signaling pathway in the absence or presence of TGF-β2 on Schlemm's canal endothelial cells. Exp Eye Res 2025; 251:110215. [PMID: 39710100 DOI: 10.1016/j.exer.2024.110215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/19/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Intraocular pressure (IOP) is regulated through the balance of production and drainage of aqueous humor. The main route of aqueous-humor outflow comprises the trabecular meshwork (TM) and Schlemm's canal (SC). We reported that IL-6 trans-signaling can inhibit TGF-β signaling in TM cells and may affect regulation of IOP. However, the function of IL-6 trans-signaling in SC cells remains unclear. Therefore, we investigated the role of IL-6 trans-signaling in monkey SC cells. Simultaneous treatment with IL-6 and soluble IL-6 receptor (sIL-6R) significantly decreased the trans-endothelial electrical resistance (TER) of SC cells and reduced aqueous-humor outflow resistance. Moreover, activation of IL-6 trans-signaling significantly reduced expression of fibronectin, ZO-1 and claudin-5, and increased that of several matrix metalloproteinases. We also investigated the effect of IL-6 trans-signaling on TGF-β2-induced changes in SC cells. Simultaneous treatment with IL-6 and sIL-6R significantly suppressed the TGF-β2-induced increase in the TER of SC cells but did not affect the activity of the TGF-β2 signaling pathway. By contrast, the TGF-β2-induced increases in the expression of fibronectin and collagen type I were significantly decreased upon simultaneous treatment with IL-6 and sIL-6R. The results show that IL-6 trans-signaling suppressed TGF-β2-induced increase in outflow resistance.
Collapse
Affiliation(s)
- Mai Urahashi
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomokazu Fujimoto
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Miyuki Inoue-Mochita
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Toshihiro Inoue
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Ding W, Moattari C, Stohl LL, Wagner JA, Zhou XK, Granstein RD. IL-6 Signalling to Responding T Cells Is Key to Calcitonin Gene-Related Peptide-Exposed Endothelial Cell Enhancement of Th17 Immunity During Langerhans Cell Antigen Presentation. Immunology 2025. [PMID: 39829087 DOI: 10.1111/imm.13892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Calcitonin gene-related peptide (CGRP) biases Langerhans cell (LC) Ag presentation to CD4+ T cells towards Th17-type immunity through actions on endothelial cells (ECs). We now report further evidence that IL-6 signalling at responding T cells mediates this effect. This CGRP effect was absent with ECs from IL-6 KO mice. Exposure of LCs, but not T cells, to IL-6 enhanced IL-6 and IL-17A production and reduced IFN-γ in the T-cell response. Pretreatment of LCs with IL-6 receptor α-chain (IL-6Rα) antibodies prior to IL-6 exposure significantly inhibited these responses. However, T-cell pretreatment with an IL-6/IL-6Rα chimera mimicked the effect of IL-6 pretreatment of LCs on T-cell responses. When this experiment was performed in the presence of the ADAM17 and ADAM10 inhibitor TAPI-1 during LC pretreatment of LCs and during the Ag presentation culture, release of soluble IL-6Rα chains into the medium was very significantly reduced, but this did not affect levels of T-cell cytokine release. Interestingly, LC exposure to IL-6 significantly increased LC IL-6 expression. Furthermore, pretreatment of T cells with antibodies against the IL-6 receptor β-chain significantly inhibited the IL-6 effect. CGRP may stimulate ECs in lymphatics and/or lymph nodes to produce IL-6 which likely results in migrating LCs nonclassically presenting IL-6. Furthermore, we found that IL-6 induces IL-6 production by LCs, suggesting an autocrine amplification pathway for this effect.
Collapse
Affiliation(s)
- Wanhong Ding
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Cameron Moattari
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Lori L Stohl
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - John A Wagner
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Xi K Zhou
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | | |
Collapse
|
4
|
Wang X, Xie J, Yang Y, Li M, Li G, Zhang X, Li J. The relationship between plasma interleukin-6 and cognition based on curve correlation in drug-naïve patients with major depressive disorder. J Affect Disord 2025; 369:211-217. [PMID: 39349223 DOI: 10.1016/j.jad.2024.09.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/26/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND The effect of interleukin-6 (IL-6) on cognition in patients with major depressive disorder (MDD) remains unclear. The aim of the present study was to investigate for the first time the non-linear relationship between plasma IL-6 and cognition and its sex-specific associations in patients with drug-naïve MDD. METHODS A total of 326 participants, including 237 drug-naïve MDD patients and 89 healthy controls (HCs), were included in this study. All participants completed the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and fasting venous blood was collected for IL-6 measurement. Patients with MDD completed the Hamilton Depression Scale-17 (HAMD-17) and the Hamilton Anxiety Scale-14 (HAMA-14) assessments. Two-way analysis of variance and curve estimation analyses were used to explore the effects of IL-6 on cognition and its sex differences. RESULTS We found that IL-6 and cognition were associated in different patterns in HCs and MDD patients. The best model for curve estimation between IL-6 and attention (F = 2.736, p = 0.045) and HAMA (F = 6.416, p < 0.001) in females with MDD was the cubic model. In male MDD patients, the best model for curve estimation between IL-6 and immediate memory was the cubic model (F = 3.077, p = 0.034). However, in HCs, the best model for curve estimation analysis between IL-6 and language was the quadratic model (F = 3.803, p = 0.026). LIMITATIONS The main limitations were cross-sectional design. CONCLUSION There was a non-linear and sex-specific relationship between IL-6 levels and cognition in patients with MDD.
Collapse
Affiliation(s)
- Xiaoli Wang
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Jun Xie
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Yuan Yang
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Meijuan Li
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Gang Li
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China; Chifeng Anding Hospital, Inner Mongolia, China
| | - Xue Zhang
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China; Chifeng Anding Hospital, Inner Mongolia, China
| | - Jie Li
- Tianjin Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
5
|
Koureta E, Karatzas P, Kanellopoulos PN, Papapanagiotou A, Lekakis V, Bamias G, Karamanolis G, Vlachogiannakos J, Papavassiliou AG, Papatheodoridis GV. The importance of growth differentiation factor 15 and interleukin 6 serum levels in inflammatory bowel diseases. J Physiol Biochem 2024:10.1007/s13105-024-01057-4. [PMID: 39560915 DOI: 10.1007/s13105-024-01057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
There are only scarce recent reports about the role of growth differentiation factor 15 (GDF-15) and some more data about interleukin-6 (IL-6) in inflammatory bowel diseases (IBD). We assessed GDF-15 and IL-6 serum levels in patients with IBD and associations with their characteristics. We included 122 and 71 stored samples from patients with Crohn's disease (CD) and ulcerative colitis (UC), respectively, and regular follow-up and 44 samples from healthy controls. Data regarding epidemiologic and disease characteristics were recorded. In CD, both GDF-15 and IL-6 levels were higher in active disease or all patients than controls (P ≤ 0.020) as well as patients with elevated CRP (P ≤ 0.008), endoscopically active disease (P ≤ 0.017), age ≥ 40 years (P ≤ 0.005) and active smokers (P ≤ 0.050) and were positively correlated with hospitalization numbers (P ≤ 0.019). GDF-15 levels were also positively correlated with flares within year-1 (P < 0.001). In UC, both GDF-15 and IL-6 levels were higher in clinically active or all patients than controls (P < 0.001), but they shared no other association with patient characteristics except for positive correlation with CRP. Only IL-6 levels were higher in active than inactive UC either clinically (P = 0.047) or endoscopically (P < 0.001) and were positively correlated with stool calprotectin (P = 0.021). GDF-15 was positively correlated to IL-6 levels only in UC (rs=0.591, P < 0.001) but not in CD. In conclusion, in CD, GDF-15 and IL-6 levels could constitute indexes of activity and even offer a prognostic index of disease progression. In UC, IL-6 could also represent an activity index, but the role of GDF-15 needs further evaluation.
Collapse
Affiliation(s)
- Evgenia Koureta
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece
| | - Pantelis Karatzas
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece
| | - Panagiotis N Kanellopoulos
- Department of Biological Chemistry, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Angeliki Papapanagiotou
- Department of Biological Chemistry, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Vasileios Lekakis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece
| | - Giorgos Bamias
- GastroenteroIogy Unit, 3rd Department of Internal Medicine, Medical School of National, Kapodistrian University of Athens, "Sotiria" Hospital, Athens, Greece
| | - George Karamanolis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece
| | - Jiannis Vlachogiannakos
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - George V Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", 17 Agiou Thoma street, Athens, 11527, Greece.
| |
Collapse
|
6
|
Chen M, Chen W, Sun S, Lu Y, Wu G, Xu H, Yang H, Li C, He W, Xu M, Li X, Jiang D, Cai Y, Liu C, Zhang W, He Z. CDK4/6 inhibitor PD-0332991 suppresses hepatocarcinogenesis by inducing senescence of hepatic tumor-initiating cells. J Adv Res 2024:S2090-1232(24)00374-6. [PMID: 39218249 DOI: 10.1016/j.jare.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Owing to the limited treatment options for hepatocellular carcinoma (HCC), interventions targeting pre-HCC stages have attracted increasing attention. In the pre-HCC stage, hepatic tumor-initiating cells (hTICs) proliferate abnormally and contribute to hepatocarcinogenesis. Numerous studies have investigated targeted senescence induction as an HCC intervention. However, it remains to be clarified whether senescence induction of hTICs could serve as a pre-HCC intervention. OBJECTIVES This study was designed to investigate whether senescence induction of hTICs in the precancerous stage inhibit HCC initiation. METHODS AND RESULTS HCC models developed from chronic liver injury (CLI) were established by using Fah-/- mice and N-Ras + AKT mice. PD-0332991, a selective CDK4/6 inhibitor that blocks the G1/S transition in proliferating cells, was used to induce senescence during the pre-HCC stage. Upon administration of PD-0332991, we observed a significant reduction in HCC incidence following selective senescence induction in hTICs, and an alleviation liver injury in the CLI-HCC models. PD-0332991 also induced senescence in vitro in cultured hTICs isolated from CLI-HCC models. Moreover, RNA sequencing (RNA-seq) analysis delineated that the "Cyclin D-CDK4/6-INK4-Rb" pathway was activated in both mouse and human liver samples during the pre-HCC stage, while PD-0332991 exhibited substantial inhibition of this pathway, thereby inducing cellular senescence in hTICs. Regarding the immune microenvironment, we demonstrated that senescent hTICs secrete key senescence-associated secretory phenotypic (SASP) factors, CXCL10 and CCL2, to activate and recruit macrophages, and contribute to immune surveillance. CONCLUSION We found that hTICs can be targeted and induced into a senescent state during the pre-HCC stage. The SASP factors released by senescent hTICs further activate the immune response, facilitating the clearance of hTICs, and consequently suppressing HCC occurrence. We highlight the importance of pre-HCC interventions and propose that senescence-inducing drugs hold promise for preventing HCC initiation under CLI.
Collapse
Affiliation(s)
- Miaomiao Chen
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Wenjian Chen
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Shiwen Sun
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Yanli Lu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Hongyu Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Huiru Yang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New Area, Shanghai, China
| | - Weizhi He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Mingyang Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Xiuhua Li
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Dong Jiang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Yongchao Cai
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Changcheng Liu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Wencheng Zhang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China.
| |
Collapse
|
7
|
Dolla G, Nicolas S, Dos Santos LR, Bourgeois A, Pardossi-Piquard R, Bihl F, Zaghrini C, Justino J, Payré C, Mansuelle P, Garbers C, Ronco P, Checler F, Lambeau G, Petit-Paitel A. Ectodomain shedding of PLA2R1 is mediated by the metalloproteases ADAM10 and ADAM17. J Biol Chem 2024; 300:107480. [PMID: 38897568 PMCID: PMC11301074 DOI: 10.1016/j.jbc.2024.107480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2023] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Phospholipase A2 receptor 1 (PLA2R1) is a 180-kDa transmembrane protein that plays a role in inflammation and cancer and is the major autoantigen in membranous nephropathy, a rare but severe autoimmune kidney disease. A soluble form of PLA2R1 has been detected in mouse and human serum. It is likely produced by proteolytic shedding of membrane-bound PLA2R1 but the mechanism is unknown. Here, we show that human PLA2R1 is cleaved by A Disintegrin And Metalloprotease 10 (ADAM10) and ADAM17 in HEK293 cells, mouse embryonic fibroblasts, and human podocytes. By combining site-directed mutagenesis and sequencing, we determined the exact cleavage site within the extracellular juxtamembrane stalk of human PLA2R1. Orthologs and paralogs of PLA2R1 are also shed. By using pharmacological inhibitors and genetic approaches with RNA interference and knock-out cellular models, we identified a major role of ADAM10 in the constitutive shedding of PLA2R1 and a dual role of ADAM10 and ADAM17 in the stimulated shedding. We did not observe evidence for cleavage by β- or γ-secretase, suggesting that PLA2R1 may not be a substrate for regulated intramembrane proteolysis. PLA2R1 shedding occurs constitutively and can be triggered by the calcium ionophore ionomycin, the protein kinase C activator PMA, cytokines, and lipopolysaccharides, in vitro and in vivo. Altogether, our results show that PLA2R1 is a novel substrate for ADAM10 and ADAM17, producing a soluble form that is increased in inflammatory conditions and likely exerts various functions in physiological and pathophysiological conditions including inflammation, cancer, and membranous nephropathy.
Collapse
Affiliation(s)
- Guillaume Dolla
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Sarah Nicolas
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Ligia Ramos Dos Santos
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Alexandre Bourgeois
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Raphaëlle Pardossi-Piquard
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Franck Bihl
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Christelle Zaghrini
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Joana Justino
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Christine Payré
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Pascal Mansuelle
- Plateforme de Protéomique de l'Institut de Microbiologie de la Méditerranée (IMM), Marseille Protéomique (MaP), Aix Marseille Université (AMU), Centre National de la Recherche Scientifique (CNRS) FR3479, Marseille, France
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Pierre Ronco
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S1155, Paris, France; Sorbonne Université, Université Pierre et Marie Curie Paris 06, Paris, France
| | - Frédéric Checler
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Laboratoire d'Excellence DistALZ, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France
| | - Gérard Lambeau
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France.
| | - Agnès Petit-Paitel
- Centre National de la Recherche Scientifique, Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia Antipolis, Université Côte d'Azur (UniCa), Valbonne, France.
| |
Collapse
|
8
|
Mohamed AH, Ahmed AT, Al Abdulmonem W, Bokov DO, Shafie A, Al-Hetty HRAK, Hsu CY, Alissa M, Nazir S, Jamali MC, Mudhafar M. Interleukin-6 serves as a critical factor in various cancer progression and therapy. Med Oncol 2024; 41:182. [PMID: 38900329 DOI: 10.1007/s12032-024-02422-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/24/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Interleukin-6 (IL-6), a pro-inflammatory cytokine, plays a crucial role in host immune defense and acute stress responses. Moreover, it modulates various cellular processes, including proliferation, apoptosis, angiogenesis, and differentiation. These effects are facilitated by various signaling pathways, particularly the signal transducer and activator of transcription 3 (STAT3) and Janus kinase 2 (JAK2). However, excessive IL-6 production and dysregulated signaling are associated with various cancers, promoting tumorigenesis by influencing all cancer hallmarks, such as apoptosis, survival, proliferation, angiogenesis, invasiveness, metastasis, and notably, metabolism. Emerging evidence indicates that selective inhibition of the IL-6 signaling pathway yields therapeutic benefits across diverse malignancies, such as multiple myeloma, prostate, colorectal, renal, ovarian, and lung cancers. Targeting key components of IL-6 signaling, such as IL-6Rs, gp130, STAT3, and JAK via monoclonal antibodies (mAbs) or small molecules, is a heavily researched approach in preclinical cancer studies. The purpose of this study is to offer an overview of the role of IL-6 and its signaling pathway in various cancer types. Furthermore, we discussed current preclinical and clinical studies focusing on targeting IL-6 signaling as a therapeutic strategy for various types of cancer.
Collapse
Affiliation(s)
- Asma'a H Mohamed
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Babil, Hilla, 51001, Iraq
| | - Abdulrahman T Ahmed
- Department of Nursing, Al-Maarif University College, Ramadi, AL-Anbar Governorate, Iraq.
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy named after A.P. Nelyubin, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, Russian Federation, 119991
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, Russian Federation, 109240
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | | | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, 85004, USA
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shahid Nazir
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Mohammad Chand Jamali
- Faculty of Medical and Health Sciences, Liwa College, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mustafa Mudhafar
- Department of Medical Physics, College of Applied Medical Sciences, University of Kerbala, Karbala, 56001, Iraq
- Department of Anesthesia Techniques and Intensive Care, Al-Taff University College, Kerbala, 56001, Iraq
| |
Collapse
|
9
|
Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What Have We Learned? BioDrugs 2024; 38:61-71. [PMID: 37989892 PMCID: PMC10789669 DOI: 10.1007/s40259-023-00634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The use of different pathways in the treatment of rheumatoid arthritis has led to a significant decrease in the number of treatment-resistant patients. In this context, interleukin (IL)-6 inhibition has filled an important gap in rheumatoid arthritis treatment with its effectiveness and safety in both monotherapy and combinations. The process of IL-6 inhibition initiated with IL-6 receptor blockers has prompted questions regarding the potential impact and safety of different inhibitions of this pathway, such as the direct blockade of IL-6. Following the termination of the development of sirukumab because of mortality data in early studies, the investigation of olokizumab, which targets a different region of the IL-6 cytokine, has renewed the hope in this area and the safety concerns have been largely alleviated by the open-label extension data. In addition, the efficacy and safety of tocilizumab and sarilumab have led to a rapid investigation of biosimilars and new potent IL-6 receptor blockers. A comprehensive understanding of mechanisms of this pathway with further long-term clinical data and basic research may provide a decisive impact on selecting the appropriate mechanism as the first choice in personalized treatments.
Collapse
Affiliation(s)
- Ali Berkant Avci
- Department of Internal Medicine, Rheumatology, Medical Park Antalya Hospital, Antalya, Türkiye
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik Vogelsang-Gommern, Cooperation Partner of the Otto-von-Guericke University Magdeburg, Gommern, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Medizinische Klinik für Rheumatologie und Klinische Immunologie, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
10
|
Pereira VM, Pradhanang S, Prather JF, Nair S. Role of Metalloproteinases in Diabetes-associated Mild Cognitive Impairment. Curr Neuropharmacol 2024; 23:58-74. [PMID: 38963109 PMCID: PMC11519823 DOI: 10.2174/1570159x22666240517090855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 07/05/2024] Open
Abstract
Diabetes has been linked to an increased risk of mild cognitive impairment (MCI), a condition characterized by a subtle cognitive decline that may precede the development of dementia. The underlying mechanisms connecting diabetes and MCI involve complex interactions between metabolic dysregulation, inflammation, and neurodegeneration. A critical mechanism implicated in diabetes and MCI is the activation of inflammatory pathways. Chronic low-grade inflammation, as observed in diabetes, can lead to the production of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 beta (IL-1β), and interferon-gamma (IFNγ), each of which can exacerbate neuroinflammation and contribute to cognitive decline. A crucial enzyme involved in regulating inflammation is ADAM17, a disintegrin, and metalloproteinase, which can cleave and release TNF-α from its membrane-bound precursor and cause it to become activated. These processes, in turn, activate additional inflammation-related pathways, such as AKT, NF-κB, NLP3, MAPK, and JAK-STAT pathways. Recent research has provided novel insights into the role of ADAM17 in diabetes and neurodegenerative diseases. ADAM17 is upregulated in both diabetes and Alzheimer's disease, suggesting a shared mechanism and implicating inflammation as a possible contributor to much broader forms of pathology and pointing to a possible link between inflammation and the emergence of MCI. This review provides an overview of the different roles of ADAM17 in diabetes-associated mild cognitive impairment diseases. It identifies mechanistic connections through which ADAM17 and associated pathways may influence the emergence of mild cognitive impairment.
Collapse
Affiliation(s)
- Vitoria Mattos Pereira
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| | - Suyasha Pradhanang
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| | - Jonathan F. Prather
- Department of Zoology and Physiology, Program in Neuroscience, University of Wyoming, Laramie, WY 82071, USA
| | - Sreejayan Nair
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
11
|
Ciryam P, Gerzanich V, Simard JM. Interleukin-6 in Traumatic Brain Injury: A Janus-Faced Player in Damage and Repair. J Neurotrauma 2023; 40:2249-2269. [PMID: 37166354 PMCID: PMC10649197 DOI: 10.1089/neu.2023.0135] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/12/2023] Open
Abstract
Traumatic brain injury (TBI) is a common and often devastating illness, with wide-ranging public health implications. In addition to the primary injury, victims of TBI are at risk for secondary neurological injury by numerous mechanisms. Current treatments are limited and do not target the profound immune response associated with injury. This immune response reflects a convergence of peripheral and central nervous system-resident immune cells whose interaction is mediated in part by a disruption in the blood-brain barrier (BBB). The diverse family of cytokines helps to govern this communication and among these, Interleukin (IL)-6 is a notable player in the immune response to acute neurological injury. It is also a well-established pharmacological target in a variety of other disease contexts. In TBI, elevated IL-6 levels are associated with worse outcomes, but the role of IL-6 in response to injury is double-edged. IL-6 promotes neurogenesis and wound healing in animal models of TBI, but it may also contribute to disruptions in the BBB and the progression of cerebral edema. Here, we review IL-6 biology in the context of TBI, with an eye to clarifying its controversial role and understanding its potential as a target for modulating the immune response in this disease.
Collapse
Affiliation(s)
- Prajwal Ciryam
- Shock Trauma Neurocritical Care, Program in Trauma, R Adams Cowley Shock Trauma Center, University of Maryland Medical System, Baltimore, Maryland, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Edsfeldt A, Gonçalves I, Vigren I, Jovanović A, Engström G, Shore AC, Natali A, Khan F, Nilsson J. Circulating soluble IL-6 receptor associates with plaque inflammation but not with atherosclerosis severity and cardiovascular risk. Vascul Pharmacol 2023; 152:107214. [PMID: 37634789 DOI: 10.1016/j.vph.2023.107214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND The residual cardiovascular risk in subjects receiving guideline-recommended therapy is related to persistent vascular inflammation and IL-6 represents a target for its treatment. IL-6 binds to receptors on leukocytes and hepatocytes and/or by forming complexes with soluble IL-6 receptors (sIL-6R) binding to gp130 which is present on all cells. Here we aimed to estimate the associations of these two pathways with risk of cardiovascular disease (CVD). METHODS IL-6 and sIL-6R were analyzed using the proximity extension assay. Baseline plasma samples were obtained from participants in the prospective Malmö Diet and Cancer (MDC) study (n = 4661), the SUMMIT VIP study (n = 1438) and the Carotid Plaque Imaging Project (CPIP, n = 285). Incident clinical events were obtained through national registers. Plaques removed at surgery were analyzed by immunohistochemistry and biochemical methods. RESULTS During 23.1 ± 7.0 years follow-up, 575 subjects in the MDC cohort suffered a first myocardial infarction. Subjects in the highest tertile of IL-6 had an increased risk compared to the lowest tertile (HR and 95% CI 2.60 [2.08-3.25]). High plasma IL-6 was also associated with more atherosclerosis, increased arterial stiffness, and impaired endothelial function in SUMMIT VIP, but IL-6 was only weakly associated with plaque inflammation in CPIP. sIL-6R showed no independent association with risk of myocardial infarction, atherosclerosis severity or vascular function, but was associated with plaque inflammation. CONCLUSIONS Our findings show that sIL-6R is a poor marker of CVD risk and associated vascular changes. However, the observation that sIL-6R reflects plaque inflammation highlights the complexity of the role of IL-6 in CVD.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Cardiology, Skåne University Hospital, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Cardiology, Skåne University Hospital, Sweden
| | - Isa Vigren
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Anja Jovanović
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Faisel Khan
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Sweden.
| |
Collapse
|
13
|
Schumacher N, Thomsen I, Brundert F, Hejret V, Düsterhöft S, Tichý B, Schmidt-Arras D, Voss M, Rose-John S. EGFR stimulation enables IL-6 trans-signalling via iRhom2-dependent ADAM17 activation in mammary epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119489. [PMID: 37271223 DOI: 10.1016/j.bbamcr.2023.119489] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/16/2022] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023]
Abstract
The cytokine interleukin-6 (IL-6) has considerable pro-inflammatory properties and is a driver of many physiological and pathophysiological processes. Cellular responses to IL-6 are mediated by membrane-bound or soluble forms of the IL-6 receptor (IL-6R) complexed with the signal-transducing subunit gp130. While expression of the membrane-bound IL-6R is restricted to selected cell types, soluble IL-6R (sIL-6R) enables gp130 engagement on all cells, a process termed IL-6 trans-signalling and considered to be pro-inflammatory. sIL-6R is predominantly generated through proteolytic processing by the metalloproteinase ADAM17. ADAM17 also liberates ligands of the epidermal growth factor receptor (EGFR), which is a prerequisite for EGFR activation and results in stimulation of proliferative signals. Hyperactivation of EGFR mostly due to activating mutations drives cancer development. Here, we reveal an important link between overshooting EGFR signalling and the IL-6 trans-signalling pathway. In epithelial cells, EGFR activity induces not only IL-6 expression but also the proteolytic release of sIL-6R from the cell membrane by increasing ADAM17 surface activity. We find that this derives from the transcriptional upregulation of iRhom2, a crucial regulator of ADAM17 trafficking and activation, upon EGFR engagement, which results in increased surface localization of ADAM17. Also, phosphorylation of the EGFR-downstream mediator ERK mediates ADAM17 activity via interaction with iRhom2. In sum, our study reveals an unforeseen interplay between EGFR activation and IL-6 trans-signalling, which has been shown to be fundamental in inflammation and cancer.
Collapse
Affiliation(s)
- Neele Schumacher
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany.
| | - Ilka Thomsen
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Florian Brundert
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Vaclav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital Aachen/RWTH, Aachen, Germany
| | - Boris Tichý
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | | | - Matthias Voss
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| |
Collapse
|
14
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
15
|
D'Elia L, Masulli M, Iacone R, Russo O, Strazzullo P, Galletti F. Relationship between leptin and white blood cells: a potential role in infection susceptibility and severity-the Olivetti Heart Study. Intern Emerg Med 2023; 18:1429-1436. [PMID: 37217748 PMCID: PMC10202358 DOI: 10.1007/s11739-023-03313-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/13/2022] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
A number of evidence showed an emerging role of leptin on immune system, involving inflammation, and innate and adaptive immunity. Few observational studies have evaluated the relationship between leptin and immunity, albeit with low statistical power and methodological differences. Therefore, the aim of this study was to evaluate the potential role of leptin on the immunity, expressed as white blood cells (WBC)-and its subpopulations, by comprehensive multivariate models in a sample of adult men. A cross-sectional evaluation of a general population comprised 939 subjects participating in the Olivetti Heart Study, with available leptin levels and WBC-and its subpopulations. WBC were significantly and positively associated with leptin, C-reactive protein and HOMA index (p < 0.05), but not with age and anthropometric indices (p > 0.05). The multivariate analysis confirmed the association between leptin and WBC, after accounting for main confounders (p < 0.05). Additional analysis on WBC subpopulations showed a positive and significant correlation between leptin and lymphocytes, monocytes and eosinophils (p < 0.05), but not with neutrophils and basophils (p > 0.05). After stratification by body weight, the positive and significant association between leptin and WBC-and its subpopulations-was found in excess body weight participants. The results of this study indicate a direct relationship between leptin levels and WBC-and its subpopulations-in excess body weight participants. These results support the hypothesis that leptin has modulatory functions on immunity and role in the pathophysiology of immune-related diseases, in particular in those associated with excess body weight.
Collapse
Affiliation(s)
- Lanfranco D'Elia
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy.
| | - Maria Masulli
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy
| | - Roberto Iacone
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy
| | - Ornella Russo
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy
| | - Pasquale Strazzullo
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy
| | - Ferruccio Galletti
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Via S. Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
16
|
Lin W, Song H, Shen J, Wang J, Yang Y, Yang Y, Cao J, Xue L, Zhao F, Xiao T, Lin R. Functional role of skeletal muscle-derived interleukin-6 and its effects on lipid metabolism. Front Physiol 2023; 14:1110926. [PMID: 37555019 PMCID: PMC10405179 DOI: 10.3389/fphys.2023.1110926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2022] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
The detrimental impact of obesity on human health is increasingly evident with the rise in obesity-related diseases. Skeletal muscle, the crucial organ responsible for energy balance metabolism, plays a significant role as a secretory organ by releasing various myokines. Among these myokines, interleukin 6 (IL-6) is closely associated with skeletal muscle contraction. IL-6 triggers the process of lipolysis by mobilizing energy-storing adipose tissue, thereby providing energy for physical exercise. This phenomenon also elucidates the health benefits of regular exercise. However, skeletal muscle and adipose tissue maintain a constant interaction, both directly and indirectly. Direct interaction occurs through the accumulation of excess fat within skeletal muscle, known as ectopic fat deposition. Indirect interaction takes place when adipose tissue is mobilized to supply the energy for skeletal muscle during exercise. Consequently, maintaining a functional balance between skeletal muscle and adipose tissue becomes paramount in regulating energy metabolism and promoting overall health. IL-6, as a representative cytokine, participates in various inflammatory responses, including non-classical inflammatory responses such as adipogenesis. Skeletal muscle influences adipogenesis through paracrine mechanisms, primarily by secreting IL-6. In this research paper, we aim to review the role of skeletal muscle-derived IL-6 in lipid metabolism and other physiological activities, such as insulin resistance and glucose tolerance. By doing so, we provide valuable insights into the regulatory function of skeletal muscle-derived myokines in lipid metabolism.
Collapse
Affiliation(s)
- Weimin Lin
- *Correspondence: Weimin Lin, ; Ruiyi Lin,
| | | | | | | | | | | | | | | | | | | | - Ruiyi Lin
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
17
|
Shahini A, Shahini A. Role of interleukin-6-mediated inflammation in the pathogenesis of inflammatory bowel disease: focus on the available therapeutic approaches and gut microbiome. J Cell Commun Signal 2023; 17:55-74. [PMID: 36112307 PMCID: PMC10030733 DOI: 10.1007/s12079-022-00695-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/16/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is considered a chronic inflammatory and multifactorial disease of the gastrointestinal tract. Crohn's disease (CD) and ulcerative colitis (UC) are two types of chronic IBD. Although there is no accurate information about IBD pathophysiology, evidence suggests that various factors, including the gut microbiome, environment, genetics, lifestyle, and a dysregulated immune system, may increase susceptibility to IBD. Moreover, inflammatory mediators such as interleukin-6 (IL-6) are involved in the immunopathogenesis of IBDs. IL-6 contributes to T helper 17 (Th17) differentiation, mediating further destructive inflammatory responses in CD and UC. Moreover, Th1-mediated responses participate in IBD, and the antiapoptotic IL-6/IL-6 receptor (IL-6R)/signal transducer and activator of transcription 3 (STAT3) signals are responsible for preserving Th1 cells in the site of inflammation. It has been revealed that fecal bacteria isolated from UC-active and UC-remission patients stimulate the hyperproduction of several cytokines, such as IL-6, tumor necrosis factor-α (TNF-α), IL-10, and IL-12. Given the importance of the IL-6/IL-6R axis, various therapeutic options exist for controlling or treating IBD. Therefore, alternative therapeutic approaches such as modulating the gut microbiome could be beneficial due to the failure of the target therapies so far. This review article summarizes IBD immunopathogenesis focusing on the IL-6/IL-6R axis and discusses available therapeutic approaches based on the gut microbiome alteration and IL-6/IL-6R axis targeting and treatment failure.
Collapse
Affiliation(s)
- Arshia Shahini
- Department of Laboratory Sciences, School of Allied Medical Sciences, Arak University of Medical Sciences, Arak, Iran
| | - Ali Shahini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Müller SA, Shmueli MD, Feng X, Tüshaus J, Schumacher N, Clark R, Smith BE, Chi A, Rose-John S, Kennedy ME, Lichtenthaler SF. The Alzheimer's disease-linked protease BACE1 modulates neuronal IL-6 signaling through shedding of the receptor gp130. Mol Neurodegener 2023; 18:13. [PMID: 36810097 PMCID: PMC9942414 DOI: 10.1186/s13024-023-00596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2022] [Accepted: 01/11/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND The protease BACE1 is a major drug target for Alzheimer's disease, but chronic BACE1 inhibition is associated with non-progressive cognitive worsening that may be caused by modulation of unknown physiological BACE1 substrates. METHODS To identify in vivo-relevant BACE1 substrates, we applied pharmacoproteomics to non-human-primate cerebrospinal fluid (CSF) after acute treatment with BACE inhibitors. RESULTS Besides SEZ6, the strongest, dose-dependent reduction was observed for the pro-inflammatory cytokine receptor gp130/IL6ST, which we establish as an in vivo BACE1 substrate. Gp130 was also reduced in human CSF from a clinical trial with a BACE inhibitor and in plasma of BACE1-deficient mice. Mechanistically, we demonstrate that BACE1 directly cleaves gp130, thereby attenuating membrane-bound gp130 and increasing soluble gp130 abundance and controlling gp130 function in neuronal IL-6 signaling and neuronal survival upon growth-factor withdrawal. CONCLUSION BACE1 is a new modulator of gp130 function. The BACE1-cleaved, soluble gp130 may serve as a pharmacodynamic BACE1 activity marker to reduce the occurrence of side effects of chronic BACE1 inhibition in humans.
Collapse
Affiliation(s)
- Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Merav D Shmueli
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xiao Feng
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Ryan Clark
- Neuroscience, Merck & Co. Inc., Boston, MA, USA
| | - Brad E Smith
- Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - An Chi
- Chemical Biology, Merck & Co. Inc., Boston, MA, USA
| | | | | | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. .,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
19
|
Garbers C, Rose-John S. Dissecting Interleukin-6 Classic and Trans-signaling in Inflammation and Cancer. Methods Mol Biol 2023; 2691:207-224. [PMID: 37355548 DOI: 10.1007/978-1-0716-3331-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 06/26/2023]
Abstract
Interleukin-6 (IL-6) is a cytokine synthesized by many cells in the human body. IL-6 binds to a membrane-bound receptor (IL-6R), which is only present on hepatocytes, some epithelial cells, and some leukocytes. The complex of IL-6 and IL-6R binds to the ubiquitously expressed receptor subunit gp130, which forms a homodimer and thereby initiates intracellular signaling, e.g., the JAK/STAT and MAPK pathways. Proteases can cleave the membrane-bound IL-6R from the cell surface and generate a soluble IL-6R (sIL-6R), which retains its ability to bind IL-6. The IL-6/sIL-6R complex associates with gp130 and induces signaling even on cells which do not express the IL-6R. This paradigm has been called IL-6 trans-signaling, whereas signaling via the membrane-bound IL-6R is referred to as classic signaling. We have generated several molecular tools to differentiate between both pathways and to analyze the consequences of cellular IL-6 signaling in vivo. One of these tools is soluble gp130Fc, which selectively inhibits IL-6 trans-signaling. This protein under the WHO name Olamkicept has successfully undergone phase II clinical trials in patients with autoimmune diseases. Here, in this chapter, we describe several molecular tools to differentiate between IL-6 classic and trans-signaling and to analyze the consequences of cellular IL-6 signaling in vivo.
Collapse
Affiliation(s)
- Christoph Garbers
- Medical Faculty, Department of Pathology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany.
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany.
| | | |
Collapse
|
20
|
Pinci F, Gaidt MM, Jung C, Nagl D, Kuut G, Hornung V. Tumor necrosis factor is a necroptosis-associated alarmin. Front Immunol 2022; 13:1074440. [PMID: 36578489 PMCID: PMC9791252 DOI: 10.3389/fimmu.2022.1074440] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
Necroptosis is a form of regulated cell death that can occur downstream of several immune pathways. While previous studies have shown that dysregulated necroptosis can lead to strong inflammatory responses, little is known about the identity of the endogenous molecules that trigger these responses. Using a reductionist in vitro model, we found that soluble TNF is strongly released in the context of necroptosis. On the one hand, necroptosis promotes TNF translation by inhibiting negative regulatory mechanisms acting at the post-transcriptional level. On the other hand, necroptosis markedly enhances TNF release by activating ADAM proteases. In studying TNF release at single-cell resolution, we found that TNF release triggered by necroptosis is activated in a switch-like manner that exceeds steady-state TNF processing in magnitude and speed. Although this shedding response precedes massive membrane damage, it is closely associated with lytic cell death. Further, we found that lytic cell death induction using a pore-forming toxin also triggers TNF shedding, indicating that the activation of ADAM proteases is not strictly related to the necroptotic pathway but likely associated with biophysical changes of the cell membrane upon lytic cell death. These results demonstrate that lytic cell death, particularly necroptosis, is a critical trigger for TNF release and thus qualify TNF as a necroptosis-associated alarmin.
Collapse
|
21
|
Inflammatory Cytokines and Radiotherapy in Pancreatic Ductal Adenocarcinoma. Biomedicines 2022; 10:biomedicines10123215. [PMID: 36551971 PMCID: PMC9775272 DOI: 10.3390/biomedicines10123215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a therapeutic challenge in clinical oncology. Surgery is the only potentially curative treatment. However, the majority of PDAC patients present with locally advanced/unresectable or metastatic disease, where palliative multiagent chemotherapy is the first-line treatment with the therapeutic intent to delay progression and prolong survival. For locally advanced/unresectable pancreatic cancer patients who are treated with chemotherapy, consolidative radiotherapy in the form concurrent chemoradiation or stereotactic ablative radiotherapy improves locoregional control and pain/symptom control. To improve clinical outcomes of PDAC patients, there is a dire need for discoveries that will shed more light on the pathophysiology of the disease and lead to the development of more efficacious treatment strategies. Inflammatory cytokines are known to play a role in mediating tumor progression, chemoresistance, and radioresistance in PDAC. A PubMed search on published articles related to radiotherapy, inflammatory cytokines, and pancreatic cancer patients in the English language was performed. This article primarily focuses on reviewing the clinical literature that examines the association of inflammatory cytokines with clinical outcomes and the effects of radiotherapy on inflammatory cytokines in PDAC patients.
Collapse
|
22
|
Cooley A, Rayford KJ, Arun A, Villalta F, Lima MF, Pratap S, Nde PN. Trypanosoma cruzi Dysregulates piRNAs Computationally Predicted to Target IL-6 Signaling Molecules During Early Infection of Primary Human Cardiac Fibroblasts. Immune Netw 2022; 22:e51. [PMID: 36627941 PMCID: PMC9807959 DOI: 10.4110/in.2022.22.e51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/17/2022] [Revised: 09/20/2022] [Accepted: 10/26/2022] [Indexed: 12/31/2022] Open
Abstract
Trypanosoma cruzi, the etiological agent of Chagas disease, is an intracellular protozoan parasite, which is now present in most industrialized countries. About 40% of T. cruzi infected individuals will develop severe, incurable cardiovascular, gastrointestinal, or neurological disorders. The molecular mechanisms by which T. cruzi induces cardiopathogenesis remain to be determined. Previous studies showed that increased IL-6 expression in T. cruzi patients was associated with disease severity. IL-6 signaling was suggested to induce pro-inflammatory and pro-fibrotic responses, however, the role of this pathway during early infection remains to be elucidated. We reported that T. cruzi can dysregulate the expression of host PIWI-interacting RNAs (piRNAs) during early infection. Here, we aim to evaluate the dysregulation of IL-6 signaling and the piRNAs computationally predicted to target IL-6 molecules during early T. cruzi infection of primary human cardiac fibroblasts (PHCF). Using in silico analysis, we predict that piR_004506, piR_001356, and piR_017716 target IL6 and SOCS3 genes, respectively. We validated the piRNAs and target gene expression in T. cruzi challenged PHCF. Secreted IL-6, soluble gp-130, and sIL-6R in condition media were measured using a cytokine array and western blot analysis was used to measure pathway activation. We created a network of piRNAs, target genes, and genes within one degree of biological interaction. Our analysis revealed an inverse relationship between piRNA expression and the target transcripts during early infection, denoting the IL-6 pathway targeting piRNAs can be developed as potential therapeutics to mitigate T. cruzi cardiomyopathies.
Collapse
Affiliation(s)
- Ayorinde Cooley
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Ashutosh Arun
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Fernando Villalta
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Cell, Molecular, and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Maria F. Lima
- Department of Cell, Molecular, and Biomedical Sciences, School of Medicine, The City College of New York, New York, NY 10031, USA
| | - Siddharth Pratap
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
23
|
HDAC6-dependent deacetylation of TAK1 enhances sIL-6R release to promote macrophage M2 polarization in colon cancer. Cell Death Dis 2022; 13:888. [PMID: 36270986 PMCID: PMC9587286 DOI: 10.1038/s41419-022-05335-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Histone deacetylase 6 (HDAC6), a member of the HDAC family, has been identified as a potential therapeutic target for tumor therapy, but the function and underlying mechanisms of HDAC6 in colon cancer are incompletely characterized. Our study showed that the infiltration ratio of M2 macrophages was increased in colon cancer tissues with high HDAC6 expression. Similarly, the knockdown of HDAC6 in colon cancer cells inhibited cocultured macrophage M2 polarization in vitro. Analysis of the antibody chip revealed that HDAC6 promoted sIL-6R release to enhance macrophage M2 polarization. Mass spectrometry and immunoprecipitation demonstrated that, mechanistically, HDAC6 interacted with transforming growth factor β-activated kinase 1 (TAK1), deacetylated TAK1 at T178 and promoted TAK1 phosphorylation. TAK1-p38 MAPK signaling could further increase the phosphorylation and activity of ADAM17, which is responsible for shedding of IL-6R. Notably, the expression of phosphorylated TAK1 was positively correlated with HDAC6 expression and macrophage M2 polarization in human colon cancer tissues. Our study revealed a new HDAC6-TAK1-ADAM17 regulatory axis that mediates sIL-6R release and macrophage polarization in colon cancer.
Collapse
|
24
|
da Silva MC, dos Santos VM, da Silva MVB, Prazeres TCMM, Cartágenes MDSS, Calzerra NTM, de Queiroz TM. Involvement of shedding induced by ADAM17 on the nitric oxide pathway in hypertension. Front Mol Biosci 2022; 9:1032177. [PMID: 36310604 PMCID: PMC9614329 DOI: 10.3389/fmolb.2022.1032177] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/30/2022] [Accepted: 10/04/2022] [Indexed: 11/15/2022] Open
Abstract
A Disintegrin and Metalloprotease 17 (ADAM17), also called tumor necrosis factor-ɑ (TNF-ɑ) convertase (TACE), is a well-known protease involved in the sheddase of growth factors, chemokines and cytokines. ADAM17 is also enrolled in hypertension, especially by shedding of angiotensin converting enzyme type 2 (ACE2) leading to impairment of angiotensin 1–7 [Ang-(1–7)] production and injury in vasodilation, induction of renal damage and cardiac hypertrophy. Activation of Mas receptor (MasR) by binding of Ang-(1–7) induces an increase in the nitric oxide (NO) gaseous molecule, which is an essential factor of vascular homeostasis and blood pressure control. On the other hand, TNF-ɑ has demonstrated to stimulate a decrease in nitric oxide bioavailability, triggering a disrupt in endothelium-dependent vasorelaxation. In spite of the previous studies, little knowledge is available about the involvement of the metalloprotease 17 and the NO pathways. Here we will provide an overview of the role of ADAM17 and Its mechanisms implicated with the NO formation.
Collapse
Affiliation(s)
- Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | - Vanessa Maria dos Santos
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | - Matheus Vinícius B. da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| | | | | | | | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
- *Correspondence: Thyago Moreira de Queiroz,
| |
Collapse
|
25
|
Li X, Jiang W, Dong S, Li W, Zhu W, Zhou W. STAT3 Inhibitors: A Novel Insight for Anticancer Therapy of Pancreatic Cancer. Biomolecules 2022; 12:1450. [PMID: 36291659 PMCID: PMC9599947 DOI: 10.3390/biom12101450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
The signal transducer and activator of transcription (STAT) is a family of intracellular cytoplasmic transcription factors involved in many biological functions in mammalian signal transduction. Among them, STAT3 is involved in cell proliferation, differentiation, apoptosis, and inflammatory responses. Despite the advances in the treatment of pancreatic cancer in the past decade, the prognosis for patients with pancreatic cancer remains poor. STAT3 has been shown to play a pro-cancer role in a variety of cancers, and inhibitors of STAT3 are used in pre-clinical and clinical studies. We reviewed the relationship between STAT3 and pancreatic cancer and the latest results on the use of STAT3 inhibitors in pancreatic cancer, with the aim of providing insights and ideas around STAT3 inhibitors for a new generation of chemotherapeutic modalities for pancreatic cancer.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wenkai Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shi Dong
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wancheng Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Weixiong Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
26
|
Hu J, Zhang Y, Huang C, Feng X, He S, Zhang Y, Maze M. Interleukin-6 trans-signalling in hippocampal CA1 neurones mediates perioperative neurocognitive disorders in mice. Br J Anaesth 2022; 129:923-936. [DOI: 10.1016/j.bja.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
|
27
|
Cabrera-Rivera GL, Madera-Sandoval RL, León-Pedroza JI, Ferat-Osorio E, Salazar-Rios E, Hernández-Aceves JA, Guadarrama-Aranda U, López-Macías C, Wong-Baeza I, Arriaga-Pizano LA. Increased Tnf- Production In Response To Il-6 In Patients With Systemic Inflammation Without Infection. Clin Exp Immunol 2022; 209:225-235. [PMID: 35647912 DOI: 10.1093/cei/uxac055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/21/2021] [Revised: 03/28/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Acute systemic inflammation can lead to life-threatening organ dysfunction. In patients with sepsis, systemic inflammation is triggered in response to infection, but in other patients, a systemic inflammatory response syndrome (SIRS) is triggered by non-infectious events. IL-6 is a major mediator of inflammation, including systemic inflammatory responses. In homeostatic conditions, when IL-6 engages its membrane-bound receptor on myeloid cells, it promotes pro-inflammatory cytokine production, phagocytosis and cell migration. However, under non-physiologic conditions, such as SIRS and sepsis, leucocyte dysfunction could modify the response of these cells to IL-6. So, our aim was to evaluate the response to IL-6 of monocytes from patients diagnosed with SIRS or sepsis. We observed that monocytes from patients with SIRS, but not from patients with sepsis, produced significantly more TNF-α than monocytes from healthy volunteers, after stimulation with IL-6. Monocytes from SIRS patients had a significantly increased baseline phosphorylation of the p65 subunit of NF-κB, with no differences in STAT3 phosphorylation or SOCS3 levels, compared to monocytes from septic patients, and this increased phosphorylation was maintained during the IL-6 activation. We found no significant differences in the expression levels of the membrane-bound IL-6 receptor, or the serum levels of IL-6, soluble IL-6 receptor, or soluble gp130, between patients with SIRS and patients with sepsis. Our results suggest that, during systemic inflammation in the absence of infection, IL-6 promotes TNF-α production by activating NF-κB, and not the canonical STAT3 pathway.
Collapse
Affiliation(s)
- Graciela L Cabrera-Rivera
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ruth L Madera-Sandoval
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico
| | - José Israel León-Pedroza
- Coordinación de Investigación, Unidad 401-C, Urgencias Médicas, Hospital General de México "Dr. Eduardo Liceaga". Mexico City, Mexico.,Coordinación de Ciclos Básicos, Universidad Anáhuac, Mexico City, Mexico
| | - Eduardo Ferat-Osorio
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,División de Investigación en Salud, UMAE Hospital de Especialidades "Dr. Bernardo Sepúlveda Gutiérrez", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social. Mexico City, Mexico
| | - Enrique Salazar-Rios
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Medicina, Universidad Autónoma del Estado de Morelos. Mexico City, Mexico
| | - Juan A Hernández-Aceves
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Química, Universidad Nacional Autónoma de México. Mexico City, Mexico
| | - Uriel Guadarrama-Aranda
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México. Mexico City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico.,Visiting Professor of Immunology. Nuffield Department of Medicine. University of Oxford, UK
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Lourdes A Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica. Centro Medico Nacional "Siglo XXI". Instituto Mexicano del Seguro Social. Mexico City, Mexico
| |
Collapse
|
28
|
Manore SG, Doheny DL, Wong GL, Lo HW. IL-6/JAK/STAT3 Signaling in Breast Cancer Metastasis: Biology and Treatment. Front Oncol 2022; 12:866014. [PMID: 35371975 PMCID: PMC8964978 DOI: 10.3389/fonc.2022.866014] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/30/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women. Metastasis is the primary cause of mortality for breast cancer patients. Multiple mechanisms underlie breast cancer metastatic dissemination, including the interleukin-6 (IL-6)-mediated signaling pathway. IL-6 is a pleiotropic cytokine that plays an important role in multiple physiological processes including cell proliferation, immune surveillance, acute inflammation, metabolism, and bone remodeling. IL-6 binds to the IL-6 receptor (IL-6Rα) which subsequently binds to the glycoprotein 130 (gp130) receptor creating a signal transducing hexameric receptor complex. Janus kinases (JAKs) are recruited and activated; activated JAKs, in turn, phosphorylate signal transducer and activator of transcription 3 (STAT3) for activation, leading to gene regulation. Constitutively active IL-6/JAK/STAT3 signaling drives cancer cell proliferation and invasiveness while suppressing apoptosis, and STAT3 enhances IL-6 signaling to promote a vicious inflammatory loop. Aberrant expression of IL-6 occurs in multiple cancer types and is associated with poor clinical prognosis and metastasis. In breast cancer, the IL-6 pathway is frequently activated, which can promote breast cancer metastasis while simultaneously suppressing the anti-tumor immune response. Given these important roles in human cancers, multiple components of the IL-6 pathway are promising targets for cancer therapeutics and are currently being evaluated preclinically and clinically for breast cancer. This review covers the current biological understanding of the IL-6 signaling pathway and its impact on breast cancer metastasis, as well as, therapeutic interventions that target components of the IL-6 pathway including: IL-6, IL-6Rα, gp130 receptor, JAKs, and STAT3.
Collapse
Affiliation(s)
- Sara G Manore
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Daniel L Doheny
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Grace L Wong
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.,Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
29
|
Hyper IgE syndromes: A clinical approach. Clin Immunol 2022; 237:108988. [DOI: 10.1016/j.clim.2022.108988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/20/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
|
30
|
Villaescusa L, Zaragozá F, Gayo-Abeleira I, Zaragozá C. A New Approach to the Management of COVID-19. Antagonists of IL-6: Siltuximab. Adv Ther 2022; 39:1126-1148. [PMID: 35072887 PMCID: PMC8784859 DOI: 10.1007/s12325-022-02042-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/10/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
Since the beginning of the pandemic, numerous national and international clinical trials have been conducted with a large number of drugs. Many of them are intended for the treatment of other pathologies; however, despite the great effort made, no specific drug is available for the treatment of the symptoms of respiratory disease caused by SARS-CoV-2 infection. The aim of this article is to provide data to justify the use of drugs to tackle the effects produced by IL-6 as the main inflammatory mediator in patients with COVID-19 with severe respiratory complications, considering all clinical evidence linking the poor prognosis of these patients with increased IL-6 levels in the context of cytokine release syndrome. Furthermore, data are provided to justify the proposal of a rational dosing of siltuximab, a monoclonal antibody specifically targeting IL-6, based on RCP levels, considering the limited results published so far on the use of this drug in COVID-19. A literature search was conducted on the clinical trials of siltuximab published to date as well as on the different IL-6 signalling pathways and the effects of its overexpression. Knowledge of the mechanisms of action on these pathways may provide important information for the design of drugs useful in the treatment of these patients. This article describes the characteristics, properties, mechanism of action, therapeutic uses and clinical studies conducted with siltuximab so far. The results confirm that administration of siltuximab downregulates IL-6 levels, thereby reducing the inflammatory process in COVID-19 patients with severe respiratory disease, suggesting that it can be successfully used to prevent cytokine release syndrome and death from this cause.
Collapse
Affiliation(s)
- Lucinda Villaescusa
- Pharmacology Unit, Biomedical Sciences Department, University of Alcalá, Madrid, 28805, Alcalá de Henares, Spain.
| | - Francisco Zaragozá
- Pharmacology Unit, Biomedical Sciences Department, University of Alcalá, Madrid, 28805, Alcalá de Henares, Spain
| | - Irene Gayo-Abeleira
- Pharmacology Unit, Biomedical Sciences Department, University of Alcalá, Madrid, 28805, Alcalá de Henares, Spain
| | - Cristina Zaragozá
- Pharmacology Unit, Biomedical Sciences Department, University of Alcalá, Madrid, 28805, Alcalá de Henares, Spain
| |
Collapse
|
31
|
Choi Y, Nagel EM, Kharoud H, Johnson KE, Gallagher T, Duncan K, Kharbanda EO, Fields DA, Gale CA, Jacobs K, Jacobs DR, Demerath EW. Gestational Diabetes Mellitus Is Associated with Differences in Human Milk Hormone and Cytokine Concentrations in a Fully Breastfeeding United States Cohort. Nutrients 2022; 14:nu14030667. [PMID: 35277026 PMCID: PMC8838140 DOI: 10.3390/nu14030667] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2022] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 01/25/2023] Open
Abstract
It is unclear whether gestational diabetes mellitus (GDM) alters breast milk composition. We prospectively examined associations of GDM status with concentrations of six potentially bioactive elements (glucose, insulin, C-reactive protein (CRP), interleukin-6 (IL-6), leptin, and adiponectin) in human milk. These were measured at both 1 and 3 months postpartum in 189 fully breastfeeding women. Mixed-effects linear regression assessed GDM status-related differences in these milk bioactives, adjusting for demographics, maternal factors, and diet. At 1 and 3 months postpartum, milk CRP was higher (1.46 ± 0.31 ng/mL; p < 0.001 and 1.69 ± 0.31 ng/mL; p < 0.001) in women with GDM than in women without GDM, whereas milk glucose (−5.23 ± 2.22 mg/dL; p = 0.02 and −5.70 ± 2.22; p = 0.01) and milk insulin (−0.38 ± 0.17 μIU/mL; p = 0.03 and −0.53 ± 0.17; p = 0.003) were lower in women with GDM. These significant associations remained similar after additional adjustment for maternal weight status and its changes. No difference was found for milk IL-6, leptin, and adiponectin. There was no evidence of association between these milk bioactive compounds and 1 h non-fasting oral glucose challenge serum glucose in the women without GDM. This prospective study provides evidence that potentially bioactive elements of human milk composition are altered in women with GDM.
Collapse
Affiliation(s)
- Yuni Choi
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
- Correspondence: ; Tel.: +1-612-624-1818; Fax: +1-612-624-0315
| | - Emily M. Nagel
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
| | - Harmeet Kharoud
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
| | - Kelsey E. Johnson
- Department of Genetics, Cell Biology, and Development, University of Minnesota-Twin Cities, Minneapolis, MN 55454, USA;
| | - Tipper Gallagher
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
| | - Katy Duncan
- Department of Pediatrics, University of Oklahoma College of Medicine, Oklahoma City, OK 73104, USA; (K.D.); (D.A.F.)
| | | | - David A. Fields
- Department of Pediatrics, University of Oklahoma College of Medicine, Oklahoma City, OK 73104, USA; (K.D.); (D.A.F.)
| | - Cheryl A. Gale
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55454, USA;
| | - Katherine Jacobs
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Women’s Health, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA;
| | - David R. Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454, USA; (E.M.N.); (H.K.); (T.G.); (D.R.J.J.); (E.W.D.)
| |
Collapse
|
32
|
A hybrid soluble gp130/spike-nanobody fusion protein simultaneously blocks IL-6 trans-signaling and cellular infection with SARS-CoV2. J Virol 2021; 96:e0162221. [PMID: 34935434 PMCID: PMC8865451 DOI: 10.1128/jvi.01622-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can induce mild to life-threatening symptoms. Especially individuals over 60 years of age or with underlying comorbidities, including heart or lung disease and diabetes, or immunocompromised patients are at a higher risk. Fatal multiorgan damage in coronavirus disease 2019 (COVID-19) patients can be attributed to an interleukin-6 (IL-6)-dominated cytokine storm. Consequently, IL-6 receptor (IL-6R) monoclonal antibody treatment for severe COVID-19 cases has been approved for therapy. High concentrations of soluble IL-6R (sIL-6R) were found in COVID-19 intensive care unit patients, suggesting the involvement of IL-6 trans-signaling in disease pathology. Here, in analogy to bispecific antibodies (bsAbs), we developed the first bispecific IL-6 trans-signaling inhibitor, c19s130Fc, which blocks viral infection and IL-6 trans-signaling. c19s130Fc is a designer protein of the IL-6 trans-signaling inhibitor cs130 fused to a single-domain nanobody directed against the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. c19s130Fc binds with high affinity to IL-6:sIL-6R complexes as well as the spike protein of SARS-CoV-2, as shown by surface plasmon resonance. Using cell-based assays, we demonstrate that c19s130Fc blocks IL-6 trans-signaling-induced proliferation and STAT3 phosphorylation in Ba/F3-gp130 cells as well as SARS-CoV-2 infection and STAT3 phosphorylation in Vero cells. Taken together, c19s130Fc represents a new class of bispecific inhibitors consisting of a soluble cytokine receptor fused to antiviral nanobodies and principally demonstrates the multifunctionalization of trans-signaling inhibitors. IMPORTANCE The availability of effective SARS-CoV-2 vaccines is a large step forward in managing the pandemic situation. In addition, therapeutic options, e.g., monoclonal antibodies to prevent viral cell entry and anti-inflammatory therapies, including glucocorticoid treatment, are currently developed or in clinical use to treat already infected patients. Here, we report a novel dual-specificity inhibitor to simultaneously target SARS-CoV-2 infection and virus-induced hyperinflammation. This was achieved by fusing an inhibitor of viral cell entry with a molecule blocking IL-6, a key mediator of SARS-CoV-2-induced hyperinflammation. Through this dual action, this molecule may have the potential to efficiently ameliorate symptoms of COVID-19 in infected individuals.
Collapse
|
33
|
Ravindranath MH, El Hilali F, Filippone EJ. The Impact of Inflammation on the Immune Responses to Transplantation: Tolerance or Rejection? Front Immunol 2021; 12:667834. [PMID: 34880853 PMCID: PMC8647190 DOI: 10.3389/fimmu.2021.667834] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
Transplantation (Tx) remains the optimal therapy for end-stage disease (ESD) of various solid organs. Although alloimmune events remain the leading cause of long-term allograft loss, many patients develop innate and adaptive immune responses leading to graft tolerance. The focus of this review is to provide an overview of selected aspects of the effects of inflammation on this delicate balance following solid organ transplantation. Initially, we discuss the inflammatory mediators detectable in an ESD patient. Then, the specific inflammatory mediators found post-Tx are elucidated. We examine the reciprocal relationship between donor-derived passenger leukocytes (PLs) and those of the recipient, with additional emphasis on extracellular vesicles, specifically exosomes, and we examine their role in determining the balance between tolerance and rejection. The concept of recipient antigen-presenting cell "cross-dressing" by donor exosomes is detailed. Immunological consequences of the changes undergone by cell surface antigens, including HLA molecules in donor and host immune cells activated by proinflammatory cytokines, are examined. Inflammation-mediated donor endothelial cell (EC) activation is discussed along with the effect of donor-recipient EC chimerism. Finally, as an example of a specific inflammatory mediator, a detailed analysis is provided on the dynamic role of Interleukin-6 (IL-6) and its receptor post-Tx, especially given the potential for therapeutic interdiction of this axis with monoclonal antibodies. We aim to provide a holistic as well as a reductionist perspective of the inflammation-impacted immune events that precede and follow Tx. The objective is to differentiate tolerogenic inflammation from that enhancing rejection, for potential therapeutic modifications. (Words 247).
Collapse
Affiliation(s)
- Mepur H. Ravindranath
- Department of Hematology and Oncology, Children’s Hospital, Los Angeles, CA, United States
- Terasaki Foundation Laboratory, Santa Monica, CA, United States
| | | | - Edward J. Filippone
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
34
|
Selective Inhibition of IL-6 Trans-Signaling Has No Beneficial Effect on the Posttraumatic Cytokine Release after Multiple Trauma in Mice. Life (Basel) 2021; 11:life11111252. [PMID: 34833127 PMCID: PMC8617644 DOI: 10.3390/life11111252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/15/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 12/26/2022] Open
Abstract
While improvements in pre-hospital and in-hospital care allow more multiple trauma patients to advance to intensive care, the incidence of posttraumatic multiple organ dysfunction syndrome (MODS) is on the rise. Herein, the influence of a selective IL-6 trans-signaling inhibition on posttraumatic cytokine levels was investigated as an approach to prevent MODS caused by a dysbalanced posttraumatic immune reaction. Therefore, the artificial IL-6 trans-signaling inhibitor sgp130Fc was deployed in a murine multiple trauma model (femoral fracture plus bilateral chest trauma). The traumatized mice were treated with sgp130Fc (FP) and compared to untreated mice (WT) and IL-6 receptor knockout mice (RKO), which received the same traumas. The overall trauma mortality was 4.4%. Microscopic pulmonary changes were apparent after multiple trauma and after isolated bilateral chest trauma. Elevated IL-6, MCP-3 and RANTES plasma levels were measured after trauma, indicating a successful induction of a systemic inflammatory reaction. Significantly reduced IL-6 and RANTES plasma levels were visible in RKO compared to WT. Only a little effect was visible in FP compared to WT. Comparable cytokine levels in WT and FP indicate neither a protective nor an adverse effect of sgp130Fc on the cytokine release after femoral fracture and bilateral chest trauma.
Collapse
|
35
|
Lokau J, Garbers C. Interleukin-6-interleukin-11 receptor chimeras reveal ionomycin-induced proteolysis beyond ADAM10. FEBS Lett 2021; 595:3072-3082. [PMID: 34778975 DOI: 10.1002/1873-3468.14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/09/2022]
Abstract
Interleukin-6 (IL-6) and interleukin-11 (IL-11) are two important pleiotropic cytokines, both of which signal through a homodimer of the β-receptor gp130. Specificity is gained through the unique, nonsignaling α-receptors IL-6R and IL-11R. Soluble variants of IL-6R and IL-11R also exist. Both membrane-bound receptors can be cleaved by the metalloprotease ADAM10. Here, we use ten different chimeric receptors consisting of different parts of IL-6R and IL-11R and analyze their susceptibility toward cleavage by ADAM10. As expected, all chimeras are substrates of ADAM10. However, we observed that cleavage of chimeric receptors containing the stalk region of the IL-11R could be blocked by the protease inhibitor GI (selective for ADAM10), but not by the protease inhibitor GW (selective for both ADAM10 and ADAM17), suggesting that another protease besides ADAM10 is involved in cleavage of these chimeras.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
36
|
Rose-John S. Local and systemic effects of interleukin-6 (IL-6) in inflammation and cancer. FEBS Lett 2021; 596:557-566. [PMID: 34738234 DOI: 10.1002/1873-3468.14220] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2021] [Revised: 10/08/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Interleukin-6 (IL-6) is an inflammatory cytokine, the level of which is highly elevated in most, if not all, inflammatory states. IL-6 triggers cell type-specific responses and acts on target cells via a specific interleukin-6 receptor (IL-6R), which, together with IL-6, binds to and induces the dimerization of a second receptor subunit, gp130. IL-6 also binds to soluble IL-6R, and this complex interacts with gp130, regardless of IL-6R expression. This allows cells that do not express IL-6R and would be otherwise insensitive to IL-6 to respond to it. We have generated a constitutively active version of gp130 by forced leucine-zipper-mediated dimerization, named L-gp130. Once inserted into the Rosa26 locus of mice, L-gp130 can be activated in a cell-autonomous manner by crossing these mice with any Cre-recombinase transgenic mouse strain. Activation of gp130 in hepatocytes produced liver-specific effects such as the induction of acute-phase proteins, but it also had profound systemic effects on the immune system. Such local and systemic effects of interleukin-6 will be reviewed.
Collapse
|
37
|
New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology. Nat Rev Gastroenterol Hepatol 2021; 18:787-803. [PMID: 34211157 DOI: 10.1038/s41575-021-00473-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
IL-6 family cytokines are defined by the common use of the signal-transducing receptor chain glycoprotein 130 (gp130). Increasing evidence indicates that these cytokines are essential in the regulation of metabolic homeostasis as well as in the pathophysiology of multiple gastrointestinal and liver disorders, thus making them attractive therapeutic targets. Over the past few years, therapies modulating gp130 signalling have grown exponentially in several clinical settings including obesity, cancer and inflammatory bowel disease. A newly engineered gp130 cytokine, IC7Fc, has shown promising preclinical results for the treatment of type 2 diabetes, obesity and liver steatosis. Moreover, drugs that modulate gp130 signalling have shown promise in refractory inflammatory bowel disease in clinical trials. A deeper understanding of the main roles of the IL-6 family of cytokines during homeostatic and pathological conditions, their signalling pathways, sources of production and target cells will be crucial to the development of improved treatments. Here, we review the current state of the role of these cytokines in hepatology and gastroenterology and discuss the progress achieved in translating therapeutics targeting gp130 signalling into clinical practice.
Collapse
|
38
|
Martínez-Pérez C, Kay C, Meehan J, Gray M, Dixon JM, Turnbull AK. The IL6-like Cytokine Family: Role and Biomarker Potential in Breast Cancer. J Pers Med 2021; 11:1073. [PMID: 34834425 PMCID: PMC8624266 DOI: 10.3390/jpm11111073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
IL6-like cytokines are a family of regulators with a complex, pleiotropic role in both the healthy organism, where they regulate immunity and homeostasis, and in different diseases, including cancer. Here we summarise how these cytokines exert their effect through the shared signal transducer IL6ST (gp130) and we review the extensive evidence on the role that different members of this family play in breast cancer. Additionally, we discuss how the different cytokines, their related receptors and downstream effectors, as well as specific polymorphisms in these molecules, can serve as predictive or prognostic biomarkers with the potential for clinical application in breast cancer. Lastly, we also discuss how our increasing understanding of this complex signalling axis presents promising opportunities for the development or repurposing of therapeutic strategies against cancer and, specifically, breast neoplasms.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Charlene Kay
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - James Meehan
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Mark Gray
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - J. Michael Dixon
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
| | - Arran K. Turnbull
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| |
Collapse
|
39
|
ADAM17 orchestrates Interleukin-6, TNFα and EGF-R signaling in inflammation and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119141. [PMID: 34610348 DOI: 10.1016/j.bbamcr.2021.119141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023]
Abstract
It was realized in the 1990s that some membrane proteins such as TNFα, both TNF receptors, ligands of the EGF-R and the Interleukin-6 receptor are proteolytically cleaved and are shed from the cell membrane as soluble proteins. The major responsible protease is a metalloprotease named ADAM17. So far, close to 100 substrates, including cytokines, cytokine receptors, chemokines and adhesion molecules of ADAM17 are known. Therefore, ADAM17 orchestrates many different signaling pathways and is a central signaling hub in inflammation and carcinogenesis. ADAM17 plays an important role in the biology of Interleukin-6 (IL-6) since the generation of the soluble Interleukin-6 receptor (sIL-6R) is needed for trans-signaling, which has been identified as the pro-inflammatory activity of this cytokine. In contrast, Interleukin-6 signaling via the membrane-bound Interleukin-6 receptor is mostly regenerative and protective. Probably due to its broad substrate spectrum, ADAM17 is essential for life and most of the few human individuals identified with ADAM17 gene defects died at young age. Although the potential of ADAM17 as a therapeutic target has been recognized, specific blockade of ADAM17 is not trivial since the metalloprotease domain of ADAM17 shares high structural homology with other proteases, in particular matrix metalloproteases. Here, the critical functions of ADAM17 in IL-6, TNFα and EGF-R pathways and strategies of therapeutic interventions are discussed.
Collapse
|
40
|
Schumertl T, Lokau J, Rose-John S, Garbers C. Function and proteolytic generation of the soluble interleukin-6 receptor in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119143. [PMID: 34626681 DOI: 10.1016/j.bbamcr.2021.119143] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/01/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022]
Abstract
The pleiotropic cytokine interleukin-6 (IL-6) is involved in numerous physiological and pathophysiological functions that include development, immune cell differentiation, inflammation and cancer. IL-6 can signal via the membrane-bound IL-6 receptor (IL-6R, classic signaling) or via soluble forms of the IL-6R (sIL-6R, trans-signaling). Both modes of signaling induce the formation of a homodimer of the signal transducing β-receptor glycoprotein 130 (gp130) and the activation of several intracellular signaling cascades, e.g. the Jak/STAT pathway. Intriguingly, only IL-6 trans-signaling is required for the pro-inflammatory properties of IL-6, while regenerative and anti-inflammatory functions are mediated via classic signaling. The sIL-6R is generated by different molecular mechanisms, including alternative mRNA splicing, proteolysis of the membrane-bound IL-6R and the release of extracellular vesicles. In this review, we give an in-depth overview on these molecular mechanisms with a special emphasize on IL-6R cleavage by the metalloprotease ADAM17 and other proteases. We discuss the biological functions of the sIL-6R and highlight attempts to selectively block IL-6 trans-signaling in pre-clinical animal models as well as in clinical studies in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Tim Schumertl
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | | | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
41
|
Nara H, Watanabe R. Anti-Inflammatory Effect of Muscle-Derived Interleukin-6 and Its Involvement in Lipid Metabolism. Int J Mol Sci 2021; 22:ijms22189889. [PMID: 34576053 PMCID: PMC8471880 DOI: 10.3390/ijms22189889] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/12/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/11/2022] Open
Abstract
Interleukin (IL)-6 has been studied since its discovery for its role in health and diseases. It is one of the most important pro-inflammatory cytokines. IL-6 was reported as an exacerbating factor in coronavirus disease. In recent years, it has become clear that the function of muscle-derived IL-6 is different from what has been reported so far. Exercise is accompanied by skeletal muscle contraction, during which, several bioactive substances, collectively named myokines, are secreted from the muscles. Many reports have shown that IL-6 is the most abundant myokine. Interestingly, it was indicated that IL-6 plays opposing roles as a myokine and as a pro-inflammatory cytokine. In this review, we discuss why IL-6 has different functions, the signaling mode of hyper-IL-6 via soluble IL-6 receptor (sIL-6R), and the involvement of soluble glycoprotein 130 in the suppressive effect of hyper-IL-6. Furthermore, the involvement of a disintegrin and metalloprotease family molecules in the secretion of sIL-6R is described. One of the functions of muscle-derived IL-6 is lipid metabolism in the liver. However, the differences between the functions of IL-6 as a pro-inflammatory cytokine and the functions of muscle-derived IL-6 are unclear. Although the involvement of myokines in lipid metabolism in adipocytes was previously discussed, little is known about the direct relationship between nonalcoholic fatty liver disease and muscle-derived IL-6. This review is the first to discuss the relationship between the function of IL-6 in diseases and the function of muscle-derived IL-6, focusing on IL-6 signaling and lipid metabolism in the liver.
Collapse
|
42
|
Rose-John S. Blocking only the bad side of IL-6 in inflammation and cancer. Cytokine 2021; 148:155690. [PMID: 34474215 DOI: 10.1016/j.cyto.2021.155690] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/30/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Interleukin-6 (IL-6) is considered an inflammatory cytokine, which is involved not only in most inflammatory states but it also plays a prominent role in inflammation associated cancers. The response of cells to the cytokine strictly depends on the presence of the IL-6 receptor (IL-6R),which presents IL-6 to the signal transducing receptor subunit gp130, which is expressed on all cells of the body. The expression of IL-6R is limited to some cells, which are therefore IL-6 target cells. The IL-6R can be cleaved by proteases and the thus generated soluble IL-6R (sIL-6R) still binds the ligand IL-6. The complex of IL-6 and sIL-6R can bind to gp130 on any cell, induce dimerization of gp130 and intracellular signaling. This process has been named IL-6 trans-signaling. A fusion protein of soluble gp130 with the constant portion of human IgG1 (sgp130Fc) turned out to be a potent and specific inhibitor of IL-6 trans-signaling. In many animal models of human diseases the significance of IL-6 trans-signaling has been analyzed. It turned out that the activities of IL-6 mediated by the sIL-6R are the pro-inflammatory activities of the cytokine whereas activities of IL-6 mediated by the membrane-bound IL-6R are rather protective and regenerative. The sgp130Fc protein has recently been developed into a biologic. The possible consequences of a specific IL-6 trans-signaling blockade is discussed in the light of the recent successfully concluded phase II clinical trials in patients with inflammatory bowel disease.
Collapse
|
43
|
Hirani D, Alvira CM, Danopoulos S, Milla C, Donato M, Tian L, Mohr J, Dinger K, Vohlen C, Selle J, Koningsbruggen-Rietschel SV, Barbarino V, Pallasch C, Rose-John S, Odenthal M, Pryhuber GS, Mansouri S, Savai R, Seeger W, Khatri P, Al Alam D, Dötsch J, Alejandre Alcazar MA. Macrophage-derived IL-6 trans-signaling as a novel target in the pathogenesis of bronchopulmonary dysplasia. Eur Respir J 2021; 59:13993003.02248-2020. [PMID: 34446466 PMCID: PMC8850688 DOI: 10.1183/13993003.02248-2020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2020] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
Rationale Premature infants exposed to oxygen are at risk for bronchopulmonary dysplasia (BPD), which is characterised by lung growth arrest. Inflammation is important, but the mechanisms remain elusive. Here, we investigated inflammatory pathways and therapeutic targets in severe clinical and experimental BPD. Methods and results First, transcriptomic analysis with in silico cellular deconvolution identified a lung-intrinsic M1-like-driven cytokine pattern in newborn mice after hyperoxia. These findings were confirmed by gene expression of macrophage-regulating chemokines (Ccl2, Ccl7, Cxcl5) and markers (Il6, Il17A, Mmp12). Secondly, hyperoxia-activated interleukin 6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) signalling was measured in vivo and related to loss of alveolar epithelial type II cells (ATII) as well as increased mesenchymal marker. Il6 null mice exhibited preserved ATII survival, reduced myofibroblasts and improved elastic fibre assembly, thus enabling lung growth and protecting lung function. Pharmacological inhibition of global IL-6 signalling and IL-6 trans-signalling promoted alveolarisation and ATII survival after hyperoxia. Third, hyperoxia triggered M1-like polarisation, possibly via Krüppel-like factor 4; hyperoxia-conditioned medium of macrophages and IL-6-impaired ATII proliferation. Finally, clinical data demonstrated elevated macrophage-related plasma cytokines as potential biomarkers that identify infants receiving oxygen at increased risk of developing BPD. Moreover, macrophage-derived IL6 and active STAT3 were related to loss of epithelial cells in BPD lungs. Conclusion We present a novel IL-6-mediated mechanism by which hyperoxia activates macrophages in immature lungs, impairs ATII homeostasis and disrupts elastic fibre formation, thereby inhibiting lung growth. The data provide evidence that IL-6 trans-signalling could offer an innovative pharmacological target to enable lung growth in severe neonatal chronic lung disease. M1-like macrophage activation is linked to IL-6/STAT3 axis in clinical and experimental BPD. Inhibition of macrophage-related IL-6 trans-signalling promotes ATII survival and lung growth in experimental BPD as a new therapy for preterm infants.https://bit.ly/3AhF7GP
Collapse
Affiliation(s)
- Dharmesh Hirani
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Carlos Milla
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michele Donato
- Biomedical Informatics Research-Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California, USA
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, USA
| | - Jasmine Mohr
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Katharina Dinger
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany
| | - Christina Vohlen
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany.,Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany
| | - Silke V Koningsbruggen-Rietschel
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Verena Barbarino
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, University of Cologne, Koln, Germany
| | - Christian Pallasch
- Department I of Internal Medicine, Center for Integrated Oncology (CIO) Köln-Bonn, University of Cologne, Koln, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Margarete Odenthal
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Institute for Pathology, Koln, Germany
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Siavash Mansouri
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL)
| | - Werner Seeger
- Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL)
| | - Purvesh Khatri
- Biomedical Informatics Research-Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California, USA
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Koln, Germany
| | - Miguel A Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Koln, Germany .,University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Koln, Germany.,Institute for Lung Health (ILH), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL).,University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
44
|
Al-Salihi M, Bornikoel A, Zhuang Y, Stachura P, Scheller J, Lang KS, Lang PA. The role of ADAM17 during liver damage. Biol Chem 2021; 402:1115-1128. [PMID: 34192832 DOI: 10.1515/hsz-2021-0149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022]
Abstract
A disintegrin and metalloprotease (ADAM) 17 is a membrane bound protease, involved in the cleavage and thus regulation of various membrane proteins, which are critical during liver injury. Among ADAM17 substrates are tumor necrosis factor α (TNFα), tumor necrosis factor receptor 1 and 2 (TNFR1, TNFR2), the epidermal growth factor receptor (EGFR) ligands amphiregulin (AR) and heparin-binding-EGF-like growth factor (HB-EGF), the interleukin-6 receptor (IL-6R) and the receptor for a hepatocyte growth factor (HGF), c-Met. TNFα and its binding receptors can promote liver injury by inducing apoptosis and necroptosis in liver cells. Consistently, hepatocyte specific deletion of ADAM17 resulted in increased liver cell damage following CD95 stimulation. IL-6 trans-signaling is critical for liver regeneration and can alleviate liver damage. EGFR ligands can prevent liver damage and deletion of amphiregulin and HB-EGF can result in increased hepatocyte death and reduced proliferation. All of which indicates that ADAM17 has a central role in liver injury and recovery from it. Furthermore, inactive rhomboid proteins (iRhom) are involved in the trafficking and maturation of ADAM17 and have been linked to liver damage. Taken together, ADAM17 can contribute in a complex way to liver damage and injury.
Collapse
Affiliation(s)
- Mazin Al-Salihi
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
- School of Medicine, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Anna Bornikoel
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Yuan Zhuang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Pawel Stachura
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Jürgen Scheller
- Department of Biochemistry and Molecular Biology II, Medical Faculty, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Karl S Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, D-45147 Essen, Germany
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
45
|
Ren P, Lu L, Cai S, Chen J, Lin W, Han F. Alternative Splicing: A New Cause and Potential Therapeutic Target in Autoimmune Disease. Front Immunol 2021; 12:713540. [PMID: 34484216 PMCID: PMC8416054 DOI: 10.3389/fimmu.2021.713540] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Alternative splicing (AS) is a complex coordinated transcriptional regulatory mechanism. It affects nearly 95% of all protein-coding genes and occurs in nearly all human organs. Aberrant alternative splicing can lead to various neurological diseases and cancers and is responsible for aging, infection, inflammation, immune and metabolic disorders, and so on. Though aberrant alternative splicing events and their regulatory mechanisms are widely recognized, the association between autoimmune disease and alternative splicing has not been extensively examined. Autoimmune diseases are characterized by the loss of tolerance of the immune system towards self-antigens and organ-specific or systemic inflammation and subsequent tissue damage. In the present review, we summarized the most recent reports on splicing events that occur in the immunopathogenesis of systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) and attempted to clarify the role that splicing events play in regulating autoimmune disease progression. We also identified the changes that occur in splicing factor expression. The foregoing information might improve our understanding of autoimmune diseases and help develop new diagnostic and therapeutic tools for them.
Collapse
Affiliation(s)
- Pingping Ren
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Luying Lu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Shasha Cai
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nephrology, The First People’s Hospital of Wenling, Taizhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| | - Weiqiang Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University of Medicine, Hangzhou, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Nephropathy, Zhejiang Province, Hangzhou, China
- Institute of Nephropathy, Zhejiang University, Hangzhou, China
| |
Collapse
|
46
|
Jiang J, Wang J, Yao L, Lai S, Zhang X. What do we know about IL-6 in COVID-19 so far? BIOPHYSICS REPORTS 2021; 7:193-206. [PMID: 37287491 PMCID: PMC10244797 DOI: 10.52601/bpr.2021.200024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/08/2020] [Accepted: 04/01/2021] [Indexed: 11/05/2022] Open
Abstract
Interleukin 6 (IL-6) is a cytokine with dual functions of pro-inflammation and anti-inflammation. It is mainly produced by mononuclear macrophages, Th2 cells, vascular endothelial cells and fibroblasts. IL-6 binds to glycoprotein 130 and one of these two receptors, membrane-bound IL-6R or soluble IL-6R, forming hexamer (IL-6/IL-6R/gp130), which then activates different signaling pathways (classical pathway, trans-signaling pathway) to exert dual immune-modulatory effects of anti-inflammation or pro-inflammation. Abnormal levels of IL-6 can cause multiple pathological reactions, including cytokine storm. Related clinical studies have found that IL-6 levels in severe COVID-19 patients were much higher than in healthy population. A large number of studies have shown that IL-6 can trigger a downstream cytokine storm in patients with COVID-19, resulting in lung damages, aggravating clinical symptoms and developing excessive inflammation and acute respiratory distress syndrome (ARDS). Monoclonal antibodies against IL-6 or IL-6R, such as tocilizumab, sarilumab, siltuximab and olokizumab may serve as therapeutic options for COVID-19 infection.
Collapse
Affiliation(s)
- Jingrui Jiang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Biomedicine and Biopharmacology, Hubei University of Technology, Wuhan 430068, China
| | - Jun Wang
- Department of Biomedicine and Biopharmacology, Hubei University of Technology, Wuhan 430068, China
- National 111 Center for Cellular Regulation and Molecular Pharmaceutics, Wuhan 430068, China
| | - Lulu Yao
- Department of Biomedicine and Biopharmacology, Hubei University of Technology, Wuhan 430068, China
- National 111 Center for Cellular Regulation and Molecular Pharmaceutics, Wuhan 430068, China
| | - Shenghan Lai
- Department of Pathology, Johns Hopkins University School of Medicine, MD 21287, USA
| | - Xueji Zhang
- Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), School of Biomedical Engineering, Shenzhen University, Shenzhen 518037, Guangdong, China
| |
Collapse
|
47
|
Yang G, Cui M, Jiang W, Sheng J, Yang Y, Zhang X. Molecular switch in human diseases-disintegrin and metalloproteinases, ADAM17. Aging (Albany NY) 2021; 13:16859-16872. [PMID: 34182543 PMCID: PMC8266367 DOI: 10.18632/aging.203200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/13/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023]
Abstract
The ADAMs (a disintegrin and metalloproteinase) are a family of cell surface proteins with crucial roles in the regulation of cell adhesion, cell proliferation to migration, proteolysis and cell signaling transduction pathways. Among these enzymes, the ADAM17 shows significant effects in the “ectodomain shedding” of its substrates such as cytokines (e.g., tumor necrosis factor α, TNFα), growth factors (e.g., epidermal growth factor, EGF), adhesion proteins (e.g., L-selectin), and their receptors (e.g., IL-6R and TNFα). Several studies focus on the underlying molecular mechanisms of ADAM17 in diseased conditions. Here, we took several different approaches to elucidate the function of ADAM17, the participation of ADAM17 in several human diseases, and the potential as targeted therapy reagents. As more and more studies verify the miRNA-mediated expression variation of ADAM17, the specific regulation network of miRNAs and ADAM17 was exploited in this review as well.
Collapse
Affiliation(s)
- Guang Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Mengying Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Weibo Jiang
- Department of Orthopaedic, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yongsheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
48
|
Maurya R, Sebastian P, Namdeo M, Devender M, Gertler A. COVID-19 Severity in Obesity: Leptin and Inflammatory Cytokine Interplay in the Link Between High Morbidity and Mortality. Front Immunol 2021; 12:649359. [PMID: 34220807 PMCID: PMC8250137 DOI: 10.3389/fimmu.2021.649359] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/04/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity is one of the foremost risk factors in coronavirus infection resulting in severe illness and mortality as the pandemic progresses. Obesity is a well-known predisposed chronic inflammatory condition. The dynamics of obesity and its impacts on immunity may change the disease severity of pneumonia, especially in acute respiratory distress syndrome, a primary cause of death from SARS-CoV-2 infection. The adipocytes of adipose tissue secret leptin in proportion to individuals’ body fat mass. An increase in circulating plasma leptin is a typical characteristic of obesity and correlates with a leptin-resistant state. Leptin is considered a pleiotropic molecule regulating appetite and immunity. In immunity, leptin functions as a cytokine and coordinates the host’s innate and adaptive responses by promoting the Th1 type of immune response. Leptin induced the proliferation and functions of antigen-presenting cells, monocytes, and T helper cells, subsequently influencing the pro-inflammatory cytokine secretion by these cells, such as TNF-α, IL-2, or IL-6. Leptin scarcity or resistance is linked with dysregulation of cytokine secretion leading to autoimmune disorders, inflammatory responses, and increased susceptibility towards infectious diseases. Therefore, leptin activity by leptin long-lasting super active antagonist’s dysregulation in patients with obesity might contribute to high mortality rates in these patients during SARS-CoV-2 infection. This review systematically discusses the interplay mechanism between leptin and inflammatory cytokines and their contribution to the fatal outcomes in COVID-19 patients with obesity.
Collapse
Affiliation(s)
- Radheshyam Maurya
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Prince Sebastian
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Madhulika Namdeo
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Moodu Devender
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Arieh Gertler
- Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
49
|
Schmidt-Arras D, Rose-John S. Endosomes as Signaling Platforms for IL-6 Family Cytokine Receptors. Front Cell Dev Biol 2021; 9:688314. [PMID: 34141712 PMCID: PMC8204807 DOI: 10.3389/fcell.2021.688314] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin-6 (IL-6) is the name-giving cytokine of a family of eleven members, including IL-6, CNTF, LIF, and IL-27. IL-6 was first recognized as a B-cell stimulating factor but we now know that the cytokine plays a pivotal role in the orchestration of inflammatory processes as well as in inflammation associated cancer. Moreover, IL-6 is involved in metabolic regulation and it has been shown to be involved in major neural activities such as neuroprotection, which can help to repair and to reduce brain damage. Receptor complexes of all members formed at the plasma membrane contain one or two molecules of the signaling receptor subunit GP130 and the mechanisms of signal transduction are well understood. IL-6 type cytokines can also signal from endomembranes, in particular the endosome, and situations have been reported in which endocytosis of receptor complexes are a prerequisite of intracellular signaling. Moreover, pathogenic GP130 variants were shown to interfere with spatial activation of downstream signals. We here summarize the molecular mechanisms underlying spatial regulation of IL-6 family cytokine signaling and discuss its relevance for pathogenic processes.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
50
|
Abstract
The liver is a vital organ with multiple functions and a large regenerative capacity. Tumours of the liver are the second most frequently cause of cancer-related death and develop in chronically inflamed livers. IL-6-type cytokines are mediators of inflammation and almost all members signal via the receptor subunit gp130 and the downstream signalling molecule STAT3. We here summarize current knowledge on how gp130 signalling and STAT3 in tumour cells and cells of the tumour micro-environment drives hepatic tumorigenesis. We furthermore discuss very recent findings describing also anti-tumorigenic roles of gp130/STAT3 and important considerations for therapeutic interventions.
Collapse
|