1
|
Franke A, Dahl S, Funck M, Bakker H, Garbers C, Lokau J. Interleukin-2 receptor α (IL-2Rα/CD25) shedding is differentially regulated by N- and O-glycosylation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119863. [PMID: 39427744 DOI: 10.1016/j.bbamcr.2024.119863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/30/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
The cytokine interleukin-2 (IL-2) is a critical regulator of immune responses, with an especially well-characterized role in regulating T-cell homeostasis. IL-2 signaling involves three distinct receptor subunits: the IL-2Rα (CD25), IL-2Rβ, and IL-2Rγ. The intracellular transduction of IL-2-induced signals is strictly dependent on IL-2Rβ and IL-2Rγ, while the IL-2Rα is not directly involved in signaling. Instead, it has the highest affinity towards IL-2 and is thus responsible for regulating the affinity of a cell for IL-2. In addition to the membrane-bound IL-2Rα, a soluble form of the receptor (sIL-2Rα) has been described, which is present in the blood of healthy individuals, increased under various pathological conditions, and able to bind IL-2 and thus modulate its function. The sIL-2Rα is generated by proteolytic cleavage of the membrane-bound receptor. Here, we analyze whether glycosylation of the IL-2Rα regulates its proteolysis. We find that constitutive IL-2Rα shedding is affected by glycosylation and discover distinct roles for N- and O-glycosylation. Furthermore, we show that induced shedding by the metalloproteases ADAM10 and ADAM17 is also differentially regulated by distinct types of glycans. Finally, we identify a specific role for an N-glycan at an exosite in ADAM17-mediated proteolysis that does not affect ADAM10, indicating distinct substrate recognition mechanisms. These results further the understanding of the mechanisms leading to sIL-2Rα generation, and thus offer the opportunity to specifically modulate the generation of the soluble receptor.
Collapse
Affiliation(s)
- Amelie Franke
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sophia Dahl
- Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Monika Funck
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Hans Bakker
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
| | - Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany; Department of Pathology, Medical Faculty, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
2
|
Chongsaritsinsuk J, Rangel-Angarita V, Lucas TM, Mahoney KE, Enny OM, Katemauswa M, Malaker SA. Quantification and Site-Specific Analysis of Co-occupied N- and O-Glycopeptides. J Proteome Res 2024; 23:5449-5461. [PMID: 39498894 DOI: 10.1021/acs.jproteome.4c00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Protein glycosylation is a complex post-translational modification that is generally classified as N- or O-linked. Site-specific analysis of glycopeptides is accomplished with a variety of fragmentation methods, depending on the type of glycosylation being investigated and the instrumentation available. For instance, collisional dissociation methods are frequently used for N-glycoproteomic analysis with the assumption that one N-sequon exists per tryptic peptide. Alternatively, electron-based methods are preferable for O-glycosite localization. However, the presence of simultaneously N- and O-glycosylated peptides could suggest the necessity of electron-based fragmentation methods for N-glycoproteomics, which is not commonly performed. Thus, we quantified the prevalence of N- and O-glycopeptides in mucins and other glycoproteins. A much higher frequency of co-occupancy within mucins was detected whereas only a negligible occurrence occurred within nonmucin glycoproteins. This was demonstrated from analyses of recombinant and/or purified proteins, as well as more complex samples. Where co-occupancy occurred, O-glycosites were frequently localized to the Ser/Thr within the N-sequon. Additionally, we found that O-glycans in close proximity to the occupied Asn were predominantly unelaborated core 1 structures, while those further away were more extended. Overall, we demonstrate electron-based methods are required for robust site-specific analysis of mucins, wherein co-occupancy is more prevalent. Conversely, collisional methods are generally sufficient for analyses of other types of glycoproteins.
Collapse
Affiliation(s)
| | | | - Taryn M Lucas
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Keira E Mahoney
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Olivia M Enny
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Mitchelle Katemauswa
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| | - Stacy A Malaker
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
3
|
Yu X, Duan R, Jiang L, Wang T, Li Z, Zhang B, Su W, Lin Y. Interleukin-6 in non-infectious uveitis: Biology, experimentalevidence and treatment strategies. Biochem Pharmacol 2024; 230:116605. [PMID: 39491564 DOI: 10.1016/j.bcp.2024.116605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/10/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Uveitis is the leading cause of visual impairment worldwide. Interleukin-6 (IL-6), which is upregulated in response to inflammation, is one of the most important inflammatory cytokines associated with uveitis. Two major IL-6 receptors (IL-6R) mediate the pro-inflammatory and anti-inflammatory biological effects of IL-6. This review summarized multiple perspectives on the mechanism of IL-6-mediated uveitis, based on experimental evidence from clinical and animal models. It includes discussions on the roles of the downstream IL-6 signaling pathway, immunocytes, and the blood-retinal barrier. Therapeutic strategies aimed at blocking the action of IL-6 have progressed in clinical practice. However, due to the adverse events associated with existing biologics including infections, drugs that selectively inhibit intraocular IL-6 still require further development. The novel concept of converting the pro-inflammatory effects of IL-6 into protective effects also requires further research. In addition, the relationship between the trans-presentation of IL-6R and T-helper17 cells in uveitis remains unexplored. This review aims to consolidate our current understanding of the biology, signaling pathways, experimental models, and immune pathogenesis related to IL-6 and uveitis. We also discuss clinical strategies focused on blocking IL-6 as a treatment for uveitis. Targeting IL-6 provides unlimited potential for improving the diagnosis, treatment, and prognosis of uveitis.
Collapse
Affiliation(s)
- Xiaoyang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510060, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Loujing Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Tianfu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Bowen Zhang
- Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510060, China
| | - Wenru Su
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200000, China.
| | - Ying Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
4
|
Garbers C, Werfel T. Important molecular differences between therapeutic IL-6 receptor inhibition and its genetic mimicry in patients with AD. J Allergy Clin Immunol 2024; 154:1559. [PMID: 39340511 DOI: 10.1016/j.jaci.2024.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Affiliation(s)
- Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Liu L, Zhu L, Monteiro-Martins S, Griffin A, Vlahos LJ, Fujita M, Berrouet C, Zanoni F, Marasa M, Zhang JY, Zhou XJ, Caliskan Y, Akchurin O, Al-Akash S, Jankauskiene A, Bodria M, Chishti A, Esposito C, Esposito V, Claes D, Tesar V, Davis TK, Samsonov D, Kaminska D, Hryszko T, Zaza G, Flynn JT, Iorember F, Lugani F, Rizk D, Julian BA, Hidalgo G, Kallash M, Biancone L, Amoroso A, Bono L, Mani LY, Vogt B, Lin F, Sreedharan R, Weng P, Ranch D, Xiao N, Quiroga A, Matar RB, Rheault MN, Wenderfer S, Selewski D, Lundberg S, Silva C, Mason S, Mahan JD, Vasylyeva TL, Mucha K, Foroncewicz B, Pączek L, Florczak M, Olszewska M, Gradzińska A, Szczepańska M, Machura E, Badeński A, Krakowczyk H, Sikora P, Kwella N, Miklaszewska M, Drożdż D, Zaniew M, Pawlaczyk K, SiniewiczLuzeńczyk K, Bomback AS, Appel GB, Izzi C, Scolari F, Materna-Kiryluk A, Mizerska-Wasiak M, Berthelot L, Pillebout E, Monteiro RC, Novak J, Green TJ, Smoyer WE, Hastings MC, Wyatt RJ, Nelson R, Martin J, González-Gay MA, De Jager PL, Köttgen A, Califano A, Gharavi AG, Zhang H, Kiryluk K. Genome-wide studies define new genetic mechanisms of IgA vasculitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315041. [PMID: 39417133 PMCID: PMC11482997 DOI: 10.1101/2024.10.10.24315041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
IgA vasculitis (IgAV) is a pediatric disease with skin and systemic manifestations. Here, we conducted genome, transcriptome, and proteome-wide association studies in 2,170 IgAV cases and 5,928 controls, generated IgAV-specific maps of gene expression and splicing from blood of 255 pediatric cases, and reconstructed myeloid-specific regulatory networks to define disease master regulators modulated by the newly identified disease driver genes. We observed significant association at the HLA-DRB1 (OR=1.55, P=1.1×10-25) and fine-mapped specific amino-acid risk substitutions in DRβ1. We discovered two novel non-HLA loci: FCAR (OR=1.51, P=1.0×10-20) encoding a myeloid IgA receptor FcαR, and INPP5D (OR=1.34, P=2.2×10-9) encoding a known inhibitor of FcαR signaling. The FCAR risk locus co-localized with a cis-eQTL increasing FCAR expression; the risk alleles disrupted a PRDM1 binding motif within a myeloid enhancer of FCAR. Another risk locus was associated with a higher genetically predicted levels of plasma IL6R. The IL6R risk haplotype carried a missense variant contributing to accelerated cleavage of IL6R into a soluble form. Using systems biology approaches, we prioritized IgAV master regulators co-modulated by FCAR, INPP5D and IL6R in myeloid cells. We additionally identified 21 shared loci in a cross-phenotype analysis of IgAV with IgA nephropathy, including novel loci PAID4, WLS, and ANKRD55.
Collapse
Affiliation(s)
- Lili Liu
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Li Zhu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Sara Monteiro-Martins
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron Griffin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Lukas J. Vlahos
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Masashi Fujita
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Cecilia Berrouet
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Francesca Zanoni
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Jun Y. Zhang
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Xu-jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University, Saint Louis, MO, USA
| | | | | | | | - Monica Bodria
- MONICA BODRIA, MD, PHD, Primary Care Unit, Ausl Parma, south east district, Parma, Italy
| | - Aftab Chishti
- Division of Pediatric Nephrology, University of Kentucky, Kentucky Children’s Hospital, Lexington, KY, USA
| | - Ciro Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Vittoria Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Donna Claes
- Cinncinnati Children’s Hospital, Cincinnati, OH, USA
| | - Vladimir Tesar
- Department of Nephrology, 1st School of Medicine, Charles University Prague, Czech Republic
| | | | - Dmitry Samsonov
- Maria Fareri Children’s Hospital (MCF), New York Medical College, New York, NY, USA
| | - Dorota Kaminska
- Department of Non-Procedural Clinical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology, Hypertension and Internal Medicine, Medical University of Bialystok, Poland
| | - Gianluigi Zaza
- Renal, Dialysis and Transplant Unit, Department of Pharmacy, Health and Nutritional Sciences (DFSSN), University of Calabria
| | - Joseph T. Flynn
- Department of Pediatrics, University of Washington; and Division of Nephrology, Seattle Children’s Hospital
| | | | | | - Dana Rizk
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | - Luisa Bono
- Nephrology and Dialysis, A.R.N.A.S. Civico and Benfratellio, Palermo, Italy
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | - Scott Wenderfer
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Dave Selewski
- Mott Children’s Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Sigrid Lundberg
- Danderyd University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Cynthia Silva
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | - Sherene Mason
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | | | | | - Krzysztof Mucha
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michał Florczak
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | - Agnieszka Gradzińska
- Department of Dermatology and Pediatric Dermatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Helena Krakowczyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Norbert Kwella
- Department of Nephrology, Transplantology and Internal Diseases, University of Warmia and Mazury, Olsztyn, Poland
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna SiniewiczLuzeńczyk
- Department of Paediatrics, Immunology and Nephrology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | | | | | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Francesco Scolari
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | | | | | - Laureline Berthelot
- Nantes University, Inserm, CR2TI Center of Research on Translational Transplantation and Immunology, Nantes, France
| | - Evangeline Pillebout
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Renato C. Monteiro
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Jan Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Robert J. Wyatt
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | - Javier Martin
- Institute of Parasitology and Biomedicine Lopez-Neyra, Spanish National Research Council (CSIC), Granada, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, Spain
| | - Philip L. De Jager
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Chan Zuckerberg Biohub New York, New York, NY, USA
| | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
6
|
Runge J, Garbers C, Lokau J. The role of interleukin-6 classic and trans-signaling and interleukin-11 classic signaling in gastric cancer cells. Contemp Oncol (Pozn) 2024; 28:105-113. [PMID: 39421714 PMCID: PMC11480916 DOI: 10.5114/wo.2024.142211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/03/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction The cytokine interleukin-11 (IL-11) binds on its target cells to a membrane-bound IL-11R, which results in homodimerization of the signal-transducing β-receptor gp130. Recent studies have uncovered a pro- inflammatory role in several diseases, including different tumor entities, and mouse models have revealed a crucial role of the IL-11/IL-11R axis in gastric cancer. However, studies regarding human gastric cancer are sparse, and whether the results obtained in mouse models also hold true in the human situation is little investigated. Material and methods We analyzed gene expression of IL11RA, IL11, IL6R, IL6 and IL6ST in different gastric tumor and immune cell lines. Furthermore, we stimulated these cell lines with exogenous cytokines and determined intracellular signaling. Finally, we analyzed gene expression data of gastric tumor patients and correlated them with overall patient survival. Results This study showed that different gastric tumor cell lines respond to IL-6 classic and trans-signaling, but only slightly to stimulation with IL-11. We observed that monocyte-like cell lines expressed high levels of IL-6R and responded to IL-6, but not to stimulation with IL-11. Using gene expression data, we found that IL11RA and IL11 are not overexpressed in human gastric cancer tissue and do not correlate with patient survival. However, low IL6 expression is associated with higher overall survival. Conclusions We provided evidence for IL-6 but not IL-11 signaling in gastric tumor cells and demonstrated that IL6 expression in gastric tumors is associated with overall survival of patients.
Collapse
Affiliation(s)
- Josephine Runge
- Institute of Pathology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School (MHH), Hannover, Germany
| | - Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
7
|
Bento R, Scheller J, Parekkadan B. Intratumoral Delivery of Genetically Engineered Anti-IL-6 Trans-signaling Therapeutics. Mol Biotechnol 2024:10.1007/s12033-024-01230-6. [PMID: 38980514 DOI: 10.1007/s12033-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Interleukin-6 (IL-6) is a highly pro-inflammatory cytokine involved in the etiopathology of several inflammatory diseases and cancer. As so, the inhibition of IL-6 signaling pathways has emerged as an attractive therapeutic avenue for the treatment of several chronic diseases. Since IL-6 trans-signaling was described as the pathological branch of IL-6, selective inhibitors were developed. Next-generation variants with increased trans-signaling specificity and potency emerged as great candidates for the treatment of several diseases, with reduced off-target effects. The highly time-consuming and costly processes involving recombinant protein production, however, have hampered the progress of anti-cytokine pharmaceuticals in clinic so far. Herein, we developed gene therapeutic modalities of IL-6-trans-signaling inhibitors as alternatives for sustained recombinant protein secretion. By using an IL-6-dependent lymphoma cell line and xenograft tumor model, we demonstrated the superior inhibitory potential of second-generation anti-IL-6 trans-signaling therapeutic. We compared the efficiency of distinct gene delivery modalities using a bioluminescent biomarker probe and observed consistent protein production via cell-based delivery. When delivered intratumorally, genetically engineered sgp130FlyRFc-secreting cells significantly reduced tumor burden and increased animal survival, representing a promising therapeutic avenue to be explored in clinically relevant gene delivery applications.
Collapse
Affiliation(s)
- Raphaela Bento
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
8
|
Chongsaritsinsuk J, Rangel-Angarita V, Mahoney KE, Lucas TM, Enny OM, Katemauswa M, Malaker SA. Quantification and site-specific analysis of co-occupied N- and O-glycopeptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.06.602348. [PMID: 39005468 PMCID: PMC11245114 DOI: 10.1101/2024.07.06.602348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Protein glycosylation is a complex post-translational modification that is generally classified as N- or O-linked. Site-specific analysis of glycopeptides is accomplished with a variety of fragmentation methods, depending on the type of glycosylation being investigated and the instrumentation available. For instance, collisional dissociation methods are frequently used for N-glycoproteomic analysis with the assumption that one N-sequon exists per tryptic peptide. Alternatively, electron-based methods are indispensable for O-glycosite localization. However, the presence of simultaneously N- and O-glycosylated peptides could suggest the necessity of electron-based fragmentation methods for N-glycoproteomics, which is not commonly performed. Thus, we quantified the prevalence of N- and O-glycopeptides in mucins and other glycoproteins. A much higher frequency of co-occupancy within mucins was detected whereas only a negligible occurrence occurred within non-mucin glycoproteins. This was demonstrated from analyses of recombinant and/or purified proteins, as well as more complex samples. Where co-occupancy occurred, O-glycosites were frequently localized to the Ser/Thr within the N-sequon. Additionally, we found that O-glycans in close proximity to the occupied Asn were predominantly unelaborated core 1 structures, while those further away were more extended. Overall, we demonstrate electron-based methods are required for robust site-specific analysis of mucins, wherein co-occupancy is more prevalent. Conversely, collisional methods are generally sufficient for analyses of other types of glycoproteins.
Collapse
|
9
|
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] [Scholar 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
|
10
|
Wang MJ, Zhang HL, Chen F, Guo XJ, Liu QG, Hou J. The double-edged effects of IL-6 in liver regeneration, aging, inflammation, and diseases. Exp Hematol Oncol 2024; 13:62. [PMID: 38890694 PMCID: PMC11184755 DOI: 10.1186/s40164-024-00527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine and exerts its complex biological functions mainly through three different signal modes, called cis-, trans-, and cluster signaling. When IL-6 binds to its membrane or soluble receptors, the co-receptor gp130 is activated to initiate downstream signaling and induce the expression of target genes. In the liver, IL-6 can perform its anti-inflammatory activities to promote hepatocyte reprogramming and liver regeneration. On the contrary, IL-6 also exerts the pro-inflammatory functions to induce liver aging, fibrosis, steatosis, and carcinogenesis. However, understanding the roles and underlying mechanisms of IL-6 in liver physiological and pathological processes is still an ongoing process. So far, therapeutic agents against IL‑6, IL‑6 receptor (IL‑6R), IL-6-sIL-6R complex, or IL-6 downstream signal transducers have been developed, and determined to be effective in the intervention of inflammatory diseases and cancers. In this review, we summarized and highlighted the understanding of the double-edged effects of IL-6 in liver homeostasis, aging, inflammation, and chronic diseases, for better shifting the "negative" functions of IL-6 to the "beneficial" actions, and further discussed the potential therapeutic effects of targeting IL-6 signaling in the clinics.
Collapse
Affiliation(s)
- Min-Jun Wang
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Hai-Ling Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Neurology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Fei Chen
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Xiao-Jing Guo
- Department of Health Statistics, Faculty of Health Service, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Qing-Gui Liu
- Department of Cell Biology, Center for Stem Cell and Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
11
|
Swaroop AK, Negi P, Kar A, Mariappan E, Natarajan J, Namboori P K K, Selvaraj J. Navigating IL-6: From molecular mechanisms to therapeutic breakthroughs. Cytokine Growth Factor Rev 2024; 76:48-76. [PMID: 38220583 DOI: 10.1016/j.cytogfr.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/28/2023] [Indexed: 01/16/2024]
Abstract
This concise review navigates the intricate realm of Interleukin-6 (IL-6), an important member of the cytokine family. Beginning with an introduction to cytokines, this narrative review unfolds with the historical journey of IL-6, illuminating its evolving significance. A crucial section unravels the three distinct signaling modes employed by IL-6, providing a foundational understanding of its versatile interactions within cellular landscapes. Moving deeper, the review meticulously dissects IL-6's signaling mechanisms, unraveling the complexities of its pleiotropic effects in both physiological responses and pathological conditions. A significant focus is dedicated to the essential role IL-6 plays in inflammatory diseases, offering insights into its associations and implications for various health conditions. The review also takes a therapeutic turn by exploring the emergence of anti-IL-6 monoclonal inhibitors, marking a profound stride in treatment modalities. Diving into the molecular realm, the review explores small molecules as agents for IL-6 inhibition, providing a nuanced perspective on diverse intervention strategies. As the review embarks on the final chapters, it contemplates future aspects, offering glimpses into potential research trajectories and the evolving landscape of IL-6-related studies.
Collapse
Affiliation(s)
- Akey Krishna Swaroop
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Preeya Negi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Ayushi Kar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Esakkimuthukumar Mariappan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Jawahar Natarajan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Krishnan Namboori P K
- Amrita Molecular Modeling and Synthesis (AMMAS) Research lab, Amrita Vishwavidyapeetham, Amrita Nagar, Ettimadai, Coimbatore, Tamil Nadu, India
| | - Jubie Selvaraj
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India.
| |
Collapse
|
12
|
Shan C, Zhang C, Zhang C. The Role of IL-6 in Neurodegenerative Disorders. Neurochem Res 2024; 49:834-846. [PMID: 38227113 DOI: 10.1007/s11064-023-04085-6] [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] [Scholar Register] [Received: 10/11/2023] [Revised: 11/26/2023] [Accepted: 12/08/2023] [Indexed: 01/17/2024]
Abstract
"Neurodegenerative disorder" is an umbrella term for a group of fatal progressive neurological illnesses characterized by neuronal loss and inflammation. Interleukin-6 (IL-6), a pleiotropic cytokine, significantly affects the activities of nerve cells and plays a pivotal role in neuroinflammation. Furthermore, as high levels of IL-6 have been frequently observed in association with several neurodegenerative disorders, it may potentially be used as a biomarker for the progression and prognosis of these diseases. This review summarizes the production and function of IL-6 as well as its downstream signaling pathways. Moreover, we make a comprehensive review on the roles of IL-6 in neurodegenerative disorders and its potential clinical application.
Collapse
Affiliation(s)
- Chen Shan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Chuanbao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, People's Republic of China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
13
|
Garbers C, Lokau J. Cytokines of the interleukin-6 family as emerging targets in inflammatory bowel disease. Expert Opin Ther Targets 2024; 28:57-65. [PMID: 38217849 DOI: 10.1080/14728222.2024.2306341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is an umbrella term that includes different chronic inflammatory diseases of the gastrointestinal tract, most commonly Crohn's disease and ulcerative colitis. IBD affects more than 6 million people worldwide and constitutes not only a debilitating disease for the patients, but also a significant factor for society due to costs for health care and reduced working capacity. Despite the introduction of biologicals for the treatment of IBD, the identification of novel targets that could lead to novel therapeutics is still needed. AREAS COVERED In this review, we summarize current knowledge about the interleukin-6 family of cytokines as potential therapeutic targets for improving the therapy of patients with IBD. We discuss cytokines like IL-6 itself for which therapeutics such as inhibitory monoclonal antibodies have already entered the clinics, but also focus on other family members whose therapeutic potential has not been explored yet. EXPERT OPINION The different cytokines of the IL-6 family offer multiple therapeutic targets that can potentially be used to treat patients with inflammatory bowel disease, but unwanted side effects like inhibition of epithelial regeneration have to be considered.
Collapse
Affiliation(s)
- Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
14
|
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] [Scholar 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
|
15
|
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: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
16
|
Ren J, Wang XQ, Nakao T, Libby P, Shi GP. Differential Roles of Interleukin-6 in Severe Acute Respiratory Syndrome-Coronavirus-2 Infection and Cardiometabolic Diseases. CARDIOLOGY DISCOVERY 2023; 3:166-182. [PMID: 38152628 PMCID: PMC10750760 DOI: 10.1097/cd9.0000000000000096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can lead to a cytokine storm, unleashed in part by pyroptosis of virus-infected macrophages and monocytes. Interleukin-6 (IL-6) has emerged as a key participant in this ominous complication of COVID-19. IL-6 antagonists have improved outcomes in patients with COVID-19 in some, but not all, studies. IL-6 signaling involves at least 3 distinct pathways, including classic-signaling, trans-signaling, and trans-presentation depending on the localization of IL-6 receptor and its binding partner glycoprotein gp130. IL-6 has become a therapeutic target in COVID-19, cardiovascular diseases, and other inflammatory conditions. However, the efficacy of inhibition of IL-6 signaling in metabolic diseases, such as obesity and diabetes, may depend in part on cell type-dependent actions of IL-6 in controlling lipid metabolism, glucose uptake, and insulin sensitivity owing to complexities that remain to be elucidated. The present review sought to summarize and discuss the current understanding of how and whether targeting IL-6 signaling ameliorates outcomes following SARS-CoV-2 infection and associated clinical complications, focusing predominantly on metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Xiao-Qi Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Tetsushi Nakao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
17
|
Arai J, Otoyama Y, Nozawa H, Kato N, Yoshida H. The immunological role of ADAMs in the field of gastroenterological chronic inflammatory diseases and cancers: a review. Oncogene 2023; 42:549-558. [PMID: 36572816 PMCID: PMC9937921 DOI: 10.1038/s41388-022-02583-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Metalloproteinases cleave transmembrane proteins that play critical roles in inflammation and cancers. Metalloproteinases include a disintegrin and metalloprotease (ADAM), which we previously examined using a fluorescence assay system, and described their association with resistance to systemic therapy in cancer patients. There are also many reports on the relation between ADAM expression and the prognosis of patients with gastroenterological chronic inflammatory diseases and cancers. Inhibiting their immunomodulating activity in chronic inflammation restores innate immunity and potentially prevents the development of various cancers. Among the numerous critical immune system-related molecules, we focus on major histocompatibility complex class I polypeptide-related sequence A (MICA), MICB, intracellular adhesion molecule (ICAM)-1, TNF-α, IL-6 receptor (IL-6R), and Notch. This review summarizes our current understanding of the role of ADAMs in gastroenterological diseases with regard to the immune system. Several Food and Drug Administration (FDA)-approved inhibitors of ADAMs have been identified, and potential therapies for targeting ADAMs in the treatment of chronic inflammatory diseases and cancers are discussed. Some ongoing clinical trials for cancers targeting ADAMs are also introduced.
Collapse
Affiliation(s)
- Jun Arai
- Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Yumi Otoyama
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hisako Nozawa
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Naoya Kato
- grid.136304.30000 0004 0370 1101Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hitoshi Yoshida
- grid.410714.70000 0000 8864 3422Division of Gastroenterology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Lu M, Lee Y, Lillehoj HS. Evolution of developmental and comparative immunology in poultry: The regulators and the regulated. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 138:104525. [PMID: 36058383 DOI: 10.1016/j.dci.2022.104525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Avian has a unique immune system that evolved in response to environmental pressures in all aspects of innate and adaptive immune responses, including localized and circulating lymphocytes, diversity of immunoglobulin repertoire, and various cytokines and chemokines. All of these attributes make birds an indispensable vertebrate model for studying the fundamental immunological concepts and comparative immunology. However, research on the immune system in birds lags far behind that of humans, mice, and other agricultural animal species, and limited immune tools have hindered the adequate application of birds as disease models for mammalian systems. An in-depth understanding of the avian immune system relies on the detailed studies of various regulated and regulatory mediators, such as cell surface antigens, cytokines, and chemokines. Here, we review current knowledge centered on the roles of avian cell surface antigens, cytokines, chemokines, and beyond. Moreover, we provide an update on recent progress in this rapidly developing field of study with respect to the availability of immune reagents that will facilitate the study of regulatory and regulated components of poultry immunity. The new information on avian immunity and available immune tools will benefit avian researchers and evolutionary biologists in conducting fundamental and applied research.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Youngsub Lee
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture-Agricultural Research Service, Beltsville, MD, 20705, USA.
| |
Collapse
|
19
|
Werny L, Grogro A, Bickenbach K, Bülck C, Armbrust F, Koudelka T, Pathak K, Scharfenberg F, Sammel M, Sheikhouny F, Tholey A, Linder S, Becker-Pauly C. MT1-MMP and ADAM10/17 exhibit a remarkable overlap of shedding properties. FEBS J 2023; 290:93-111. [PMID: 35944080 DOI: 10.1111/febs.16586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/20/2022] [Accepted: 07/28/2022] [Indexed: 01/14/2023]
Abstract
Membrane-type-I matrix metalloproteinase (MT1-MMP) is one of six human membrane-bound MMPs and is responsible for extracellular matrix remodelling by degrading several substrates like fibrillar collagens, including types I-III, or fibronectin. Moreover, MT1-MMP was described as a key player in cancer progression and it is involved in various inflammatory processes, as well as in the pathogenesis of Alzheimer's disease (AD). The membrane-tethered metalloprotease meprin β as well as a disintegrin and metalloproteinase 10 (ADAM10) and ADAM17 are also associated with these diseases. Interestingly, meprin β, ADAM10/17 and MT1-MMP also have a shared substrate pool including the interleukin-6 receptor and the amyloid precursor protein. We investigated the interaction of these proteases, focusing on a possible connection between MT1-MMP and meprin β, to elucidate the potential mutual regulations of both enzymes. Herein, we show that besides ADAM10/17, MT1-MMP is also able to shed meprin β from the plasma membrane, leading to the release of soluble meprin β. Mass spectrometry-based cleavage site analysis revealed that the cleavage of meprin β by all three proteases occurs between Pro602 and Ser603 , N-terminal of the EGF-like domain. Furthermore, only inactive human pro-meprin β is shed by MT1-MMP, which is again in accordance with the shedding capability observed for ADAM10/17. Vice versa, meprin β also appears to shed MT1-MMP, indicating a complex regulatory network. Further studies will elucidate this well-orchestrated proteolytic web under distinct conditions in health and disease and will possibly show whether the loss of one of the above-mentioned sheddases can be compensated by the other enzymes.
Collapse
Affiliation(s)
- Ludwig Werny
- Institute of Biochemistry, University of Kiel, Germany
| | | | | | - Cynthia Bülck
- Institute of Biochemistry, University of Kiel, Germany
| | - Fred Armbrust
- Institute of Biochemistry, University of Kiel, Germany
| | - Tomas Koudelka
- Institute of Experimental Medicine, AG Proteomics & Bioanalytics, University of Kiel, Germany
| | - Kriti Pathak
- Institute of Biochemistry, University of Kiel, Germany
| | | | - Martin Sammel
- Institute of Biochemistry, University of Kiel, Germany
| | | | - Andreas Tholey
- Institute of Experimental Medicine, AG Proteomics & Bioanalytics, University of Kiel, Germany
| | - Stefan Linder
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | | |
Collapse
|
20
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232415937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
|
21
|
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] [Scholar 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
|
22
|
Mollinedo F. Raft platforms highly enriched in cholesterol: major scaffolds for IL-6 signalling assembly with implications in inflammation and cancer. FEBS J 2022; 289:5891-5894. [PMID: 35727702 PMCID: PMC9795985 DOI: 10.1111/febs.16547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022]
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine with complex and major roles in inflammation, which could be linked to the different ways IL-6 signals at the plasma membrane. In this issue of FEBS Journal, Woo and co-workers present evidence for the involvement of Eps15 homology domain-containing protein 1 (EHD1)-mediated lipid raft platforms, highly enriched in cholesterol, in the IL-6 signalling pathway. Because of the strong connection between IL-6, inflammation and cancer, one implication of this report is that agents or approaches targeting cholesterol-rich raft platforms may assist the development of novel strategies to treat inflammatory and malignant diseases. Comment on: https://doi.org/10.1111/febs.16458.
Collapse
Affiliation(s)
- Faustino Mollinedo
- Laboratory of Cell Death and Cancer Therapy, Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita SalasConsejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| |
Collapse
|
23
|
Aliyu M, Zohora FT, Anka AU, Ali K, Maleknia S, Saffarioun M, Azizi G. Interleukin-6 cytokine: An overview of the immune regulation, immune dysregulation, and therapeutic approach. Int Immunopharmacol 2022; 111:109130. [PMID: 35969896 DOI: 10.1016/j.intimp.2022.109130] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 12/19/2022]
Abstract
Several studies have shown that interleukin 6 (IL-6) is a multifunctional cytokine with both pro-inflammatory and anti-inflammatory activity, depending on the immune response context. Macrophages are among several cells that secrete IL-6, which they express upon activation by antigens, subsequently inducing fever and production of acute-phase proteins from the liver. Moreover, IL-6 induces the final maturation of B cells into memory B cells and plasma cells as well as an adaptive role for short-term energy allocation. Activation of IL-6 receptors results in the intracellular activation of the JAK/STAT pathway with resultant production of inflammatory cytokines. Several mechanisms-controlled IL-6 expression, but aberrant production was shown to be crucial in the pathogenesis of many diseases, which include autoimmune and chronic inflammatory diseases. IL-6 in combination with transforming growth factor β (TGF-β) induced differentiation of naïve T cells to Th17 cells, which is the cornerstone in autoimmune diseases. Recently, IL-6 secretion was shown to form the backbone of hypercytokinemia seen in the Coronavirus disease 2019 (COVID-19)-associated hyperinflammation and multiorgan failure. There are two classes of approved IL-6 inhibitors: anti-IL-6 receptor monoclonal antibodies (e.g., tocilizumab) and anti-IL-6 monoclonal antibodies (i.e., siltuximab). These drugs have been evaluated in patients with rheumatoid arthritis, juvenile idiopathic arthritis, cytokine release syndrome, and COVID-19 who have systemic inflammation. JAK/STAT pathway blockers were also successfully used in dampening IL-6 signal transduction. A better understanding of different mechanisms that modulate IL-6 expression will provide the much-needed solution with excellent safety and efficacy profiles for the treatment of autoimmune and inflammatory diseases in which IL-6 derives their pathogenesis.
Collapse
Affiliation(s)
- Mansur Aliyu
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, International Campus, TUMS-IC, Tehran, Iran; Department of Medical Microbiology, Faculty of Clinical Science, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Malaysia
| | - Abubakar Umar Anka
- Department of Medical Laboratory Science, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Kashif Ali
- Department of Pharmacy Abdul Wali, Khan University Mardan, Pakistan
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Saffarioun
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
24
|
Forcina L, Franceschi C, Musarò A. The hormetic and hermetic role of IL-6. Ageing Res Rev 2022; 80:101697. [PMID: 35850167 DOI: 10.1016/j.arr.2022.101697] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/24/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Interleukin-6 is a pleiotropic cytokine regulating different tissues and organs in diverse and sometimes discrepant ways. The dual and sometime hermetic nature of IL-6 action has been highlighted in several contexts and can be explained by the concept of hormesis, in which beneficial or toxic effects can be induced by the same molecule depending on the intensity, persistence, and nature of the stimulation. According with hormesis, a low and/or controlled IL-6 release is associated with anti-inflammatory, antioxidant, and pro-myogenic actions, whereas increased systemic levels of IL-6 can induce pro-inflammatory, pro-oxidant and pro-fibrotic responses. However, many aspects regarding the multifaceted action of IL-6 and the complex nature of its signal transduction remains to be fully elucidated. In this review we collect mechanistic insight into the molecular networks contributing to normal or pathologic changes during advancing age and in chronic diseases. We point out the involvement of IL-6 deregulation in aging-related diseases, dissecting the hormetic action of this key mediator in different tissues, with a special focus on skeletal muscle. Since IL-6 can act as an enhancer of detrimental factor associated with both aging and pathologic conditions, such as chronic inflammation and oxidative stress, this cytokine could represent a "Gerokine", a determinant of the switch from physiologic aging to age-related diseases.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14, Rome 00161, Italy.
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Scuola Superiore di Studi Avanzati Sapienza (SSAS), Via A. Scarpa, 14, Rome 00161, Italy.
| |
Collapse
|
25
|
Kirschke S, Ogunsulire I, Selvakumar B, Schumacher N, Sezin T, Rose-John S, Scheffold A, Garbers C, Lokau J. The metalloprotease ADAM10 generates soluble interleukin-2 receptor alpha (sCD25) in vivo. J Biol Chem 2022; 298:101910. [PMID: 35398356 PMCID: PMC9127578 DOI: 10.1016/j.jbc.2022.101910] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 12/04/2022] Open
Abstract
The cytokine interleukin-2 (IL-2) plays a critical role in controlling the immune homeostasis by regulating the proliferation and differentiation of immune cells, especially T cells. IL-2 signaling is mediated via the IL-2 receptor (IL-2R) complex, which consists of the IL-2Rα (CD25), the IL-2Rβ, and the IL-2Rγ. While the latter are required for signal transduction, IL-2Rα controls the ligand-binding affinity of the receptor complex. A soluble form of the IL-2Rα (sIL-2Rα) is found constitutively in human serum, though its levels are increased under various pathophysiological conditions. The sIL-2Rα originates partly from activated T cells through proteolytic cleavage, but neither the responsible proteases nor stimuli that lead to IL-2Rα cleavage are known. Here, we show that the metalloproteases ADAM10 and ADAM17 can cleave the IL-2Rα and generate a soluble ectodomain, which functions as a decoy receptor that inhibits IL-2 signaling in T cells. We demonstrate that ADAM10 is mainly responsible for constitutive shedding of the IL-2Rα, while ADAM17 is involved in IL-2Rα cleavage upon T cell activation. In vivo, we found that mice with a CD4-specific deletion of ADAM10, but not ADAM17, show reduced steady-state sIL-2Rα serum levels. We propose that the identification of proteases involved in sIL-2Rα generation will allow for manipulation of IL-2Rα cleavage, especially as constitutive and induced cleavage of IL-2Rα are executed by different proteases, and thus offer a novel opportunity to alter IL-2 function.
Collapse
Affiliation(s)
- Sophia Kirschke
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Ireti Ogunsulire
- Institute of Immunology, Kiel University & UKSH Schleswig-Holstein, Kiel, Germany
| | | | | | - Tanya Sezin
- Institute of Immunology, Kiel University & UKSH Schleswig-Holstein, Kiel, Germany
| | | | - Alexander Scheffold
- Institute of Immunology, Kiel University & UKSH Schleswig-Holstein, Kiel, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany.
| | - Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
26
|
Zhang Q, Xiong D, Pan J, Wang Y, Hardy M, Kalyanaraman B, You M. Chemoprevention of Lung Cancer with a Combination of Mitochondria-Targeted Compounds. Cancers (Basel) 2022; 14:cancers14102538. [PMID: 35626143 PMCID: PMC9140024 DOI: 10.3390/cancers14102538] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Previous reports showed that mitochondria-targeted honokiol and mitochondria-targeted lonidamine potently inhibit complex-I- and complexes-I/II-induced respiration and cancer cell proliferation. In this study, we investigated the efficacy of combining mitochondria-targeted honokiol and mitochondria-targeted lonidamine treatments for lung cancer prevention. We found that their combination exhibited striking tumor inhibition in the benzo[a]pyrene-induced murine lung tumor model without causing detectable side effects. Using single-cell RNA sequencing, we found combined treatment has a clear advantage in that it can significantly inhibit two oncogenic pathways—STAT3 signaling and AKT/mTOR/p70S6K signaling. Such dual inhibition may contribute to the greater efficacy of the combined drug treatment. Therefore, the combination provides a novel option for lung cancer chemoprevention. Abstract Combined treatment targeting mitochondria may improve the efficacy of lung cancer chemoprevention. Here, mitochondria-targeted honokiol (Mito-HNK), an inhibitor of mitochondrial complex I and STAT3 phosphorylation, and mitochondria-targeted lonidamine (Mito-LND), an inhibitor of mitochondrial complexes I/II and AKT/mTOR/p70S6K signaling, were evaluated for their combinational chemopreventive efficacy on mouse lung carcinogenesis. All chemopreventive treatments began one-week post-carcinogen treatment and continued daily for 24 weeks. No evidence of toxicity (including liver toxicity) was detected by monitoring serum levels of alanine aminotransferase and aspartate aminotransferase enzymes. Mito-HNK or Mito-LND treatment alone reduced tumor load by 56% and 48%, respectively, whereas the combination of Mito-HNK and Mito-LND reduced tumor load by 83%. To understand the potential mechanism(s) of action for the observed combinatorial effects, single-cell RNA sequencing was performed using mouse tumors treated with Mito-HNK, Mito-LND, and their combination. In lung tumor cells, Mito-HNK treatment blocked the expression of genes involved in mitochondrial complex ǀ, oxidative phosphorylation, glycolysis, and STAT3 signaling. Mito-LND inhibited the expression of genes for mitochondrial complexes I/II, oxidative phosphorylation, and AKT/mTOR/p70S6K signaling in lung tumor cells. In addition to these changes, a combination of Mito-HNK with Mito-LND decreased arginine and proline metabolism, N-glycan biosynthesis, and tryptophan metabolism in lung tumor cells. Our results demonstrate that Mito-LND enhanced the antitumor efficacy of Mito-HNK, where both compounds inhibited common targets (oxidative phosphorylation) as well as unique targets for each agent (STAT3 and mTOR signaling). Therefore, the combination of Mito-HNK with Mito-LND may present an effective strategy for lung cancer chemoprevention.
Collapse
Affiliation(s)
- Qi Zhang
- Center for Cancer Prevention, Houston Methodist Research Institute, Houston, TX 77030, USA; (Q.Z.); (D.X.); (J.P.); (Y.W.)
| | - Donghai Xiong
- Center for Cancer Prevention, Houston Methodist Research Institute, Houston, TX 77030, USA; (Q.Z.); (D.X.); (J.P.); (Y.W.)
| | - Jing Pan
- Center for Cancer Prevention, Houston Methodist Research Institute, Houston, TX 77030, USA; (Q.Z.); (D.X.); (J.P.); (Y.W.)
| | - Yian Wang
- Center for Cancer Prevention, Houston Methodist Research Institute, Houston, TX 77030, USA; (Q.Z.); (D.X.); (J.P.); (Y.W.)
| | - Micael Hardy
- Aix-Marseille University, CNRS, ICR, UMR 7273, 13013 Marseille, France;
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Ming You
- Center for Cancer Prevention, Houston Methodist Research Institute, Houston, TX 77030, USA; (Q.Z.); (D.X.); (J.P.); (Y.W.)
- Correspondence:
| |
Collapse
|
27
|
Sirisereephap K, Maekawa T, Tamura H, Hiyoshi T, Domon H, Isono T, Terao Y, Maeda T, Tabeta K. Osteoimmunology in Periodontitis: Local Proteins and Compounds to Alleviate Periodontitis. Int J Mol Sci 2022; 23:5540. [PMID: 35628348 PMCID: PMC9146968 DOI: 10.3390/ijms23105540] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/25/2023] Open
Abstract
Periodontitis is one of the most common oral diseases resulting in gingival inflammation and tooth loss. Growing evidence indicates that it results from dysbiosis of the oral microbiome, which interferes with the host immune system, leading to bone destruction. Immune cells activate periodontal ligament cells to express the receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL) and promote osteoclast activity. Osteocytes have active roles in periodontitis progression in the bone matrix. Local proteins are involved in bone regeneration through functional immunological plasticity. Here, we discuss the current knowledge of cellular and molecular mechanisms in periodontitis, the roles of local proteins, and promising synthetic compounds generating a periodontal regeneration effect. It is anticipated that this may lead to a better perception of periodontitis pathophysiology.
Collapse
Affiliation(s)
- Kridtapat Sirisereephap
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (K.S.); (H.T.); (K.T.)
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (T.H.); (T.M.)
- Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (T.H.); (T.M.)
| | - Hikaru Tamura
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (K.S.); (H.T.); (K.T.)
| | - Takumi Hiyoshi
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (T.H.); (T.M.)
| | - Hisanori Domon
- Division of Microbiology and Infectious Disease, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.D.); (T.I.); (Y.T.)
| | - Toshihito Isono
- Division of Microbiology and Infectious Disease, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.D.); (T.I.); (Y.T.)
| | - Yutaka Terao
- Division of Microbiology and Infectious Disease, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (H.D.); (T.I.); (Y.T.)
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (T.H.); (T.M.)
| | - Koichi Tabeta
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (K.S.); (H.T.); (K.T.)
| |
Collapse
|
28
|
Huo Y, Yang J, Zheng J, Xu D, Yang M, Tao L, Yao H, Fu X, Yang J, Liu D, Hua R, Zhang J, Sun Y, Hu L, Liu W. Increased SPON1 promotes pancreatic ductal adenocarcinoma progression by enhancing IL-6 trans-signalling. Cell Prolif 2022; 55:e13237. [PMID: 35487760 PMCID: PMC9136514 DOI: 10.1111/cpr.13237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES This study investigated the specific molecular mechanism and the roles of extracellular matrix protein Spondin 1 (SPON1) in the development of pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS The expression pattern and clinical relevance of SPON1 was determined in GEO, Ren Ji and TCGA datasets, further validated by immunohistochemical staining and Kaplan-Meier analysis. Loss and gain of function experiments were employed to investigate the cellular function of SPON1 in vitro. Gene set enrichment analysis, luciferase assay, immunofluorescence and Western blot and immunoprecipitation were applied to reveal the underlying molecular mechanisms. Subcutaneous xenograft model was used to test the role of SPON1 in tumour growth and maintenance in vivo. RESULTS SPON1 is significantly upregulated in PDAC tumour tissues and correlated with progression of PDAC. Loss and gain of function experiments showed that SPON1 promotes the growth and colony formation ability of pancreatic cancer cells. Combining bioinformatics assays and experimental signalling evidences, we found that SPON1 can enhance the IL-6/JAK/STAT3 signalling. Mechanistically, SPON1 exerts its oncogenic roles in pancreatic cancer by maintaining IL-6R trans-signalling through stabilizing the interaction of soluble IL-6R (sIL-6R) and glycoprotein-130 (gp130) in PDAC cells. Furthermore, SPON1 depletion greatly reduced the tumour burden, exerted positive effect with gemcitabine, prolonging PDAC mice overall survival. CONCLUSIONS Our data indicate that SPON1 expression is dramatically increased in PDAC and that SPON1 promotes tumorigenicity by activating the sIL-6R/gp130/STAT3 axis. Collectively, our current work suggests SPON1 may be a potential therapy target for PDAC patient.
Collapse
Affiliation(s)
- Yanmiao Huo
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jian Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jiahao Zheng
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Dapeng Xu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Minwei Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Lingye Tao
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Hongfei Yao
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Xueliang Fu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jianyu Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Dejun Liu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Rong Hua
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Junfeng Zhang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Yongwei Sun
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Lipeng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Wei Liu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| |
Collapse
|
29
|
Changes in immune function and immunomodulatory treatments of septic patients. Clin Immunol 2022; 239:109040. [DOI: 10.1016/j.clim.2022.109040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/23/2022] [Accepted: 05/07/2022] [Indexed: 12/25/2022]
|
30
|
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] [Scholar Register] [Received: 11/20/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
|
31
|
García-Juárez M, Camacho-Morales A. Defining the role of anti- and pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience 2022; 492:32-46. [DOI: 10.1016/j.neuroscience.2022.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
|
32
|
McElvaney OJ, McEvoy NL, Boland F, McElvaney OF, Hogan G, Donnelly K, Friel O, Browne E, Fraughen DD, Murphy MP, Clarke J, Choileáin ON, O'Connor E, McGuinness R, Boylan M, Kelly A, Hayden JC, Collins AM, Cullen A, Hyland D, Carroll TP, Geoghegan P, Laffey JG, Hennessy M, Martin-Loeches I, McElvaney NG, Curley GF. A randomized, double-blind, placebo-controlled trial of intravenous alpha-1 antitrypsin for acute respiratory distress syndrome secondary to COVID-19. MED 2022; 3:233-248.e6. [PMID: 35291694 PMCID: PMC8913266 DOI: 10.1016/j.medj.2022.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022]
Abstract
Background Patients with severe coronavirus disease 2019 (COVID-19) develop a febrile pro-inflammatory cytokinemia with accelerated progression to acute respiratory distress syndrome (ARDS). Here we report the results of a phase 2, multicenter, randomized, double-blind, placebo-controlled trial of intravenous (IV) plasma-purified alpha-1 antitrypsin (AAT) for moderate to severe ARDS secondary to COVID-19 (EudraCT 2020-001391-15). Methods Patients (n = 36) were randomized to receive weekly placebo, weekly AAT (Prolastin, Grifols, S.A.; 120 mg/kg), or AAT once followed by weekly placebo. The primary endpoint was the change in plasma interleukin (IL)-6 concentration at 1 week. In addition to assessing safety and tolerability, changes in plasma levels of IL-1β, IL-8, IL-10, and soluble tumor necrosis factor receptor 1 (sTNFR1) and clinical outcomes were assessed as secondary endpoints. Findings Treatment with IV AAT resulted in decreased inflammation and was safe and well tolerated. The study met its primary endpoint, with decreased circulating IL-6 concentrations at 1 week in the treatment group. This was in contrast to the placebo group, where IL-6 was increased. Similarly, plasma sTNFR1 was substantially decreased in the treatment group while remaining unchanged in patients receiving placebo. IV AAT did not definitively reduce levels of IL-1β, IL-8, and IL-10. No difference in mortality or ventilator-free days was observed between groups, although a trend toward decreased time on ventilator was observed in AAT-treated patients. Conclusions In patients with COVID-19 and moderate to severe ARDS, treatment with IV AAT was safe, feasible, and biochemically efficacious. The data support progression to a phase 3 trial and prompt further investigation of AAT as an anti-inflammatory therapeutic. Funding ECSA-2020-009; Elaine Galwey Research Bursary.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| | - Natalie L McEvoy
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fiona Boland
- Data Science Centre, Division of Biostatistics and Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Oisín F McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| | - Grace Hogan
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | - Daniel D Fraughen
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| | - Mark P Murphy
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jennifer Clarke
- Beaumont Hospital, Dublin, Ireland
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | | | | | - John C Hayden
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ann M Collins
- RCSI Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Ailbhe Cullen
- RCSI Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Deirdre Hyland
- RCSI Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Tomás P Carroll
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - John G Laffey
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, Galway, Ireland
| | - Martina Hennessy
- Department of Critical Care Medicine, St. James' Hospital, Dublin, Ireland
| | | | - Noel G McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Beaumont Hospital, Dublin, Ireland
| | - Gerard F Curley
- Beaumont Hospital, Dublin, Ireland
- Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
33
|
Kefaloyianni E. Soluble forms of cytokine and growth factor receptors: Mechanisms of generation and modes of action in the regulation of local and systemic inflammation. FEBS Lett 2022; 596:589-606. [PMID: 35113454 DOI: 10.1002/1873-3468.14305] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/16/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022]
Abstract
Cytokine and growth factor receptors are usually transmembrane proteins but they can also exist in soluble forms, either through cleavage and release of their ligand-binding extracellular domain, or through secretion of a soluble isoform. As an extension of this concept, transmembrane receptors on exosomes released into the circulation may act similarly to circulating soluble receptors. These soluble receptors add to the complexity of cytokine and growth factor signalling: they can function as decoy receptor that compete for ligand binding with their respective membrane-bound forms thereby attenuating signalling, or stabilize their ligands, or activate additional signalling events through interactions with other cell-surface proteins. Their soluble nature allows for a functional role away from the production sites, in remote cell types and organs. Accumulating evidence demonstrates that soluble receptors participate in the regulation and orchestration of various key cellular processes, particularly inflammatory responses. In this review, we will discuss release mechanisms of soluble cytokine and growth factor receptors, their mechanisms of action, as well as strategies for targeting their pathways in disease.
Collapse
Affiliation(s)
- Eirini Kefaloyianni
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
34
|
McConnell MJ, Kondo R, Kawaguchi N, Iwakiri Y. Covid-19 and Liver Injury: Role of Inflammatory Endotheliopathy, Platelet Dysfunction, and Thrombosis. Hepatol Commun 2022; 6:255-269. [PMID: 34658172 PMCID: PMC8652692 DOI: 10.1002/hep4.1843] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/18/2021] [Accepted: 10/10/2021] [Indexed: 02/06/2023] Open
Abstract
Liver injury, characterized predominantly by elevated aspartate aminotransferase and alanine aminotransferase, is a common feature of coronavirus disease 2019 (COVID-19) symptoms caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). Additionally, SARS-CoV-2 infection is associated with acute-on-chronic liver failure in patients with cirrhosis and has a notably elevated mortality in patients with alcohol-related liver disease compared to other etiologies. Direct viral infection of the liver with SARS-CoV-2 remains controversial, and alternative pathophysiologic explanations for its hepatic effects are an area of active investigation. In this review, we discuss the effects of SARS-CoV-2 and the inflammatory environment it creates on endothelial cells and platelets more generally and then with a hepatic focus. In doing this, we present vascular inflammation and thrombosis as a potential mechanism of liver injury and liver-related complications in COVID-19.
Collapse
Affiliation(s)
- Matthew J. McConnell
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Reiichiro Kondo
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of PathologyKurume University School of MedicineKurumeJapan
| | - Nao Kawaguchi
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Yasuko Iwakiri
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
35
|
Pleiotropic, Unique and Shared Responses Elicited by IL-6 Family Cytokines in Human Vascular Endothelial Cells. Int J Mol Sci 2022; 23:ijms23031448. [PMID: 35163371 PMCID: PMC8836206 DOI: 10.3390/ijms23031448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial cells express glycoprotein 130 (gp130), which is utilized as a signaling receptor by cytokines in the interleukin-6 (IL-6) family. Several IL-6 family cytokines can be found in the circulatory system during physiological or pathological conditions, and may influence endothelial function and response. This study evaluated and compared the cellular and molecular responses induced by IL-6 family cytokines in human endothelial cells. A proteomic analysis showed that IL-6 family cytokines induce the release of a range of proteins from endothelial cells, such as C-C motif chemokine ligand 23, hepatocyte growth factor, and IL-6. Pathway analysis indicated that gp130-signaling in endothelial cells regulates several functions related to angiogenesis and immune cell recruitment. The present investigation also disclosed differences and similarities between different IL-6 family cytokines in their ability to induce protein release and regulate gene expression and intracellular signaling, in regards to which oncostatin M showed the most pronounced effect. Further, this study showed that soluble gp130 preferentially blocks trans-signaling-induced responses, but does not affect responses induced by classic signaling. In conclusion, IL-6 family cytokines induce both specific and overlapping molecular responses in endothelial cells, and regulate genes and proteins involved in angiogenesis and immune cell recruitment.
Collapse
|
36
|
Puchinger J, Ryz S, Nixdorf L, Edlinger-Stanger M, Lassnigg A, Wiedemann D, Hiesmayr M, Spittler A, Bernardi MH. Characteristics of Interleukin-6 Signaling in Elective Cardiac Surgery—A Prospective Cohort Study. J Clin Med 2022; 11:jcm11030590. [PMID: 35160042 PMCID: PMC8836792 DOI: 10.3390/jcm11030590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 12/15/2022] Open
Abstract
Interleukin-6 (IL-6) can cause pro- and anti-inflammatory effects via different signaling pathways. This prospective study investigated the perioperative kinetics of IL-6, soluble IL-6 receptor (sIL-6R), and soluble glycoprotein 130 (sgp130) in elective patients undergoing cardiopulmonary bypass (CPB). IL-6, sIL-6R, and sgp130 were measured simultaneously and consecutively at 19 timepoints until the 10th postoperative day (POD). The proportion of pro- and anti-inflammatory pathways were determined by calculating sIL-6R/IL-6 and sIL-6R/sgp130 ratios. We analyzed 93 patients. IL-6 increased during surgery with reaching a plateau two hours after CPB and peaking on POD 1 (188.5 pg mL−1 (IQR, 126.6; 309.2)). sIL-6R decreased at the beginning of the surgical procedure, reaching a nadir level on POD 2 (26,311 pg mL−1 (IQR, 22,222; 33,606)). sgp130 dropped immediately after CPB initiation (0.13 ng mL−1 (IQR, 0.12; 0.15)), followed by a continuous recovery until POD10. The sIL-6R/IL-6 ratio decreased substantially at the beginning of the procedure, reaching a nadir on POD 1 (149.7 (IQR, 82.4; 237.4)), while the sIL-6R/sgp130 ratio increased simultaneously until 6 h post CPB (0.219 (IQR 0.18; 0.27)). In conclusion, IL-6 exhibited high inter-individual variability reflecting an inhomogeneous inflammatory response. Pro-inflammatory effects and overwhelming inflammation were rare and predominantly anti-inflammatory effects were found.
Collapse
Affiliation(s)
- Jürgen Puchinger
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
- Division for Internal Medicine 3, University Hospital of St. Poelten, Dunant-Platz 1, 3100 Sankt Poelten, Austria
| | - Sylvia Ryz
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
| | - Larissa Nixdorf
- Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (L.N.); (A.S.)
| | - Maximilian Edlinger-Stanger
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
| | - Andrea Lassnigg
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
| | - Dominik Wiedemann
- Department of Cardiac Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Michael Hiesmayr
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
| | - Andreas Spittler
- Department of Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (L.N.); (A.S.)
- Core Facilities, Core Facility Flow Cytometry, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin H. Bernardi
- Division of Cardiac Thoracic Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (S.R.); (M.E.-S.); (A.L.); (M.H.)
- Correspondence: ; Tel.: +43-1-40400-41090
| |
Collapse
|
37
|
Adu-Amankwaah J, Adzika GK, Adekunle AO, Ndzie Noah ML, Mprah R, Bushi A, Akhter N, Huang F, Xu Y, Adzraku SY, Nadeem I, Sun H. ADAM17, A Key Player of Cardiac Inflammation and Fibrosis in Heart Failure Development During Chronic Catecholamine Stress. Front Cell Dev Biol 2021; 9:732952. [PMID: 34966735 PMCID: PMC8710811 DOI: 10.3389/fcell.2021.732952] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
Heart failure development is characterized by persistent inflammation and progressive fibrosis owing to chronic catecholamine stress. In a chronic stress state, elevated catecholamines result in the overstimulation of beta-adrenergic receptors (βARs), specifically β2-AR coupling with Gαi protein. Gαi signaling increases the activation of receptor-stimulated p38 mitogen-activated-protein-kinases (p38 MAPKs) and extracellular signal-regulated kinases (ERKs). Phosphorylation by these kinases is a common way to positively regulate the catalytic activity of A Disintegrin and Metalloprotease 17 (ADAM17), a metalloprotease that has grown much attention in recent years and has emerged as a chief regulatory hub in inflammation, fibrosis, and immunity due to its vital proteolytic activity. ADAM17 cleaves and activates proinflammatory cytokines and fibrotic factors that enhance cardiac dysfunction via inflammation and fibrosis. However, there is limited information on the cardiovascular aspect of ADAM17, especially in heart failure. Hence, this concise review provides a comprehensive insight into the structure of ADAM17, how it is activated and regulated during chronic catecholamine stress in heart failure development. This review highlights the inflammatory and fibrotic roles of ADAM17’s substrates; Tumor Necrosis Factor α (TNFα), soluble interleukin-6 receptor (sIL-6R), and amphiregulin (AREG). Finally, how ADAM17-induced chronic inflammation and progressive fibrosis aggravate cardiac dysfunction is discussed.
Collapse
Affiliation(s)
| | | | | | | | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Nazma Akhter
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Fei Huang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yaxin Xu
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Seyram Yao Adzraku
- Key Laboratory of Bone Marrow Stem Cell, Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
38
|
Hromakova I, Sorochan P, Prokhach N, Hromakova I. Interleukin-6 and colorectal cancer development. УКРАЇНСЬКИЙ РАДІОЛОГІЧНИЙ ТА ОНКОЛОГІЧНИЙ ЖУРНАЛ 2021; 29:89-107. [DOI: 10.46879/ukroj.4.2021.89-107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Colorectal cancer (CRC) is one of the most common malignancies in the world. It ranks third in the structure of cancer morbidity and second in the structure of mortality. One of the important factors leading to CRC is chronic inflammation of the intestine, in which pro-inflammatory cytokines play a crucial role. Among proinflammatory cytokines, interleukin-6 occupies one of the leading places in the pathogenesis of CRC. Therefore, it is important to elucidate the role of interleukin-6 (IL-6) in the development and progression of CRC, determine the diagnostic and prognostic value of the cytokine and analyze the application of therapeutic strategies aimed at the IL-6 signaling pathway in CRC. Purpose – to analyze the role of proinflammatory cytokine IL-6 in the development of colorectal cancer, consider the mechanisms of oncogenic action of cytokine, evaluate the results of therapeutic strategies aimed at the IL-6 signaling pathway in colorectal cancer and characterize prognostic and diagnostic value of IL-6. Data sources. Data search for review was performed in databases Pubmed, Cochrane Library, ScienceDirect. The results of research performed before May 2021 are analyzed. Relevant unpublished studies have been found in clinical trial registry of U.S. National Institutes of Health www.clinicaltrials.gov. Results. The assessment of diagnostic and prognostic value of IL-6 in patients with CRC is given. The mechanisms of IL-6 regulation of tumor growth, angiogenesis, apoptosis, metastasis in CRC are elucidated. The results of preclinical and clinical testing of monoclonal antibodies to IL-6, IL-6R, low molecular weight compounds that affect cytokine receptor signaling through gp130 and JAK-STAT, as well as drugs and compounds of natural origin, that are able to inhibit IL-6/STAT3 signal pathway, are presented. Conclusions. Strategies to block IL-6 signaling may be potentially useful in malignancies, most likely as a component of combination therapy, or in preventing adverse symptoms associated with cancer immunotherapy. Further research is needed to elucidate the exact role of classical IL-6 signaling and trans-signaling in the pathogenesis of colorectal cancer, as this may provide a basis for more targeted inhibition of the functions of this pleiotropic cytokine.
Collapse
|
39
|
Tøllefsen IM, Shetelig C, Seljeflot I, Eritsland J, Hoffmann P, Andersen GØ. High levels of interleukin-6 are associated with final infarct size and adverse clinical events in patients with STEMI. Open Heart 2021; 8:openhrt-2021-001869. [PMID: 34933964 PMCID: PMC8693166 DOI: 10.1136/openhrt-2021-001869] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Inflammation has emerged as a new treatment target in patients with coronary artery disease and inflammation seems to play an important role in ischaemia/reperfusion injury that follows ST-elevation myocardial infarction (STEMI). We aimed to explore the role of acute and sustained interleukin 6 (IL-6) signalling, including soluble IL-6 receptor (IL-6R), with regard to infarct size, adverse remodelling and future cardiovascular events in patients with STEMI. Methods We included 269 patients with first-time STEMI, symptom duration <6 hours and treated with percutaneous coronary intervention. Blood sampling and cardiac MRI were performed in the acute phase and after 4 months. Clinical events and all-cause mortality were registered during 12-month and 70-month follow-up, respectively. Results IL-6 levels above median at all sampling points were significantly associated with increased infarct size and reduced left ventricular ejection fraction (LVEF). IL-6 levels in the highest quartile were at all sampling points associated with an increased risk of having an adverse clinical event during the first 12 months and with long-term all-cause mortality. IL-6R was not associated with infarct size, LVEF, myocardial salvage or long-term all-cause mortality. Conclusion Acute and sustained elevation of IL-6 measured 4 months after STEMI were associated with larger infarct size, reduced LVEF and adverse clinical events including all-cause mortality. The results add important information to the sustained role of inflammation in patients with STEMI and IL-6 as a potential target for long-term intervention. Trial registration number NCT00922675.
Collapse
Affiliation(s)
| | - Christian Shetelig
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo Faculty of Medicine, Oslo, Norway
| | - Jan Eritsland
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
| | - Pavel Hoffmann
- Section of Interventional Cardiology, Oslo universitetssykehus Ulleval, Oslo, Norway
| | | |
Collapse
|
40
|
Conroy LR, Hawkinson TR, Young LEA, Gentry MS, Sun RC. Emerging roles of N-linked glycosylation in brain physiology and disorders. Trends Endocrinol Metab 2021; 32:980-993. [PMID: 34756776 PMCID: PMC8589112 DOI: 10.1016/j.tem.2021.09.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/10/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022]
Abstract
N-linked glycosylation is a complex, co- and post-translational series of events that connects metabolism to signaling in almost all cells. Metabolic assembly of N-linked glycans spans multiple cellular compartments, and early N-linked glycan biosynthesis is a central mediator of protein folding and the unfolded protein response (UPR). In the brain, N-linked glycosylated proteins participate in a myriad of processes, from electrical gradients to neurotransmission. However, it is less clear how perturbations in N-linked glycosylation impact and even potentially drive aspects of neurological disorders. In this review, we discuss our current understanding of the metabolic origins of N-linked glycans in the brain, their role in modulating neuronal function, and how aberrant N-linked glycosylation can drive neurological disorders.
Collapse
Affiliation(s)
- Lindsey R Conroy
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40508-0536, USA; Markey Cancer Center, Lexington, KY 40508-0536, USA
| | - Tara R Hawkinson
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40508-0536, USA
| | - Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry University of Kentucky College of Medicine, Lexington, KY 40508-0536, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry University of Kentucky College of Medicine, Lexington, KY 40508-0536, USA
| | - Ramon C Sun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY 40508-0536, USA; Markey Cancer Center, Lexington, KY 40508-0536, USA; Sanders Brown Center for Aging, Lexington, KY 40508-0536, USA.
| |
Collapse
|
41
|
Lokau J, Garbers Y, Grötzinger J, Garbers C. A single aromatic residue in sgp130Fc/olamkicept allows the discrimination between interleukin-6 and interleukin-11 trans-signaling. iScience 2021; 24:103309. [PMID: 34765926 PMCID: PMC8571719 DOI: 10.1016/j.isci.2021.103309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023] Open
Abstract
Blocking the activity of cytokines is an efficient strategy to combat inflammatory diseases. Interleukin-6 (IL-6) fulfills its pro-inflammatory properties via its soluble receptor (IL-6 trans-signaling). The selective trans-signaling inhibitor olamkicept (sgp130Fc) is currently in clinical development. We have previously shown that sgp130Fc can also efficiently block trans-signaling of the closely related cytokine IL-11, which elicits the question how selectivity for one of the two cytokines can be achieved. Using structural information, we show that the interfaces between IL-6R-gp130 and IL-11R-gp130, respectively, within the so-called site III are different between the two cytokines. Modification of an aromatic cluster around Q113 of gp130 within these interfaces allows the discrimination between IL-6 and IL-11 trans-signaling. Using recombinant sgp130Fc variants, we demonstrate that these differences can indeed be exploited to generate a truly selective IL-6 trans-signaling inhibitor. Our data highlight how the selectivity of a clinically relevant designer protein can be further improved.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Yvonne Garbers
- Institute of Psychology, Kiel University, 24118 Kiel, Germany
| | | | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, 39120 Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| |
Collapse
|
42
|
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] [Scholar 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
|
43
|
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] [Scholar 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
|
44
|
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: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar 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
|
45
|
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] [Scholar 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
|
46
|
Tian W, Li D, Zhang N, Bai G, Yuan K, Xiao H, Gao F, Chen Y, Wong CCL, Gao GF. O-glycosylation pattern of the SARS-CoV-2 spike protein reveals an "O-Follow-N" rule. Cell Res 2021; 31:1123-1125. [PMID: 34341488 PMCID: PMC8326647 DOI: 10.1038/s41422-021-00545-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Affiliation(s)
- Wenmin Tian
- grid.11135.370000 0001 2256 9319Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Delin Li
- grid.458513.e0000 0004 1763 3963Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, China
| | - Nan Zhang
- grid.11135.370000 0001 2256 9319Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Guijie Bai
- grid.458513.e0000 0004 1763 3963Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, China ,Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi China
| | - Kai Yuan
- grid.458513.e0000 0004 1763 3963Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, China ,Shanxi Academy of Advanced Research and Innovation, Taiyuan, Shanxi China
| | - Haixia Xiao
- grid.458513.e0000 0004 1763 3963Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, China
| | - Feng Gao
- grid.458513.e0000 0004 1763 3963Laboratory of Protein Engineering and Vaccines, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences (CAS), Tianjin, China
| | - Yang Chen
- grid.11135.370000 0001 2256 9319Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Catherine C. L. Wong
- grid.11135.370000 0001 2256 9319Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China ,grid.452723.50000 0004 7887 9190Peking-Tsinghua Center for Life Sciences, Beijing, China ,grid.411472.50000 0004 1764 1621Peking University First Hospital, Beijing, China ,grid.24696.3f0000 0004 0369 153XAdvanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - George Fu Gao
- grid.458488.d0000 0004 0627 1442CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China ,grid.419468.60000 0004 1757 8183National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, China
| |
Collapse
|
47
|
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] [Scholar 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
|
48
|
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: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
49
|
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] [Scholar 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
|
50
|
Koch L, Kespohl B, Agthe M, Schumertl T, Düsterhöft S, Lemberg MK, Lokau J, Garbers C. Interleukin-11 (IL-11) receptor cleavage by the rhomboid protease RHBDL2 induces IL-11 trans-signaling. FASEB J 2021; 35:e21380. [PMID: 33566379 DOI: 10.1096/fj.202002087r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-11 (IL-11) is a pleiotropic cytokine with both pro- and anti-inflammatory properties. It activates its target cells via binding to the membrane-bound IL-11 receptor (IL-11R), which then recruits a homodimer of the ubiquitously expressed, signal-transducing receptor gp130. Besides this classic signaling pathway, IL-11 can also bind to soluble forms of the IL-11R (sIL-11R), and IL-11/sIL-11R complexes activate cells via the induction of gp130 homodimerization (trans-signaling). We have previously reported that the metalloprotease ADAM10 cleaves the membrane-bound IL-11R and thereby generates sIL-11R. In this study, we identify the rhomboid intramembrane protease RHBDL2 as a so far unrecognized alternative sheddase that can efficiently trigger IL-11R secretion. We determine the cleavage site used by RHBDL2, which is located in the extracellular part of the receptor in close proximity to the plasma membrane, between Ala-370 and Ser-371. Furthermore, we identify critical amino acid residues within the transmembrane helix that are required for IL-11R proteolysis. We also show that ectopically expressed RHBDL2 is able to cleave the IL-11R within the early secretory pathway and not only at the plasma membrane, indicating that its subcellular localization plays a central role in controlling its activity. Moreover, RHBDL2-derived sIL-11R is biologically active and able to perform IL-11 trans-signaling. Finally, we show that the human mutation IL-11R-A370V does not impede IL-11 classic signaling, but prevents RHBDL2-mediated IL-11R cleavage.
Collapse
Affiliation(s)
- Lydia Koch
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Birte Kespohl
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Maria Agthe
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Tim Schumertl
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, RWTH Aachen University, Aachen, Germany
| | - Marius K Lemberg
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Juliane Lokau
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|