201
|
Chlorin e6 Conjugated Interleukin-6 Receptor Aptamers Selectively Kill Target Cells Upon Irradiation. MOLECULAR THERAPY. NUCLEIC ACIDS 2014; 3:e143. [PMID: 24481022 PMCID: PMC3910004 DOI: 10.1038/mtna.2013.70] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/11/2013] [Indexed: 02/01/2023]
Abstract
Photodynamic therapy (PDT) uses the therapeutic properties of light in combination with certain chemicals, called photosensitizers, to successfully treat brain, breast, prostate, and skin cancers. To improve PDT, current research focuses on the development of photosensitizers to specifically target cancer cells. In the past few years, aptamers have been developed to directly deliver cargo molecules into target cells. We conjugated the photosensitizer chlorin e6 (ce6) with a human interleukin-6 receptor (IL-6R) binding RNA aptamer, AIR-3A yielding AIR-3A-ce6 for application in high efficient PDT. AIR-3A-ce6 was rapidly and specifically internalized by IL-6R presenting (IL-6R(+)) cells. Upon light irradiation, targeted cells were selectively killed, while free ce6 did not show any toxic effect. Cells lacking the IL-6R were also not affected by AIR-3A-ce6. With this approach, we improved the target specificity of ce6-mediated PDT. In the future, other tumor-specific aptamers might be used to selectively localize photosensitizers into cells of interest and improve the efficacy and specificity of PDT in cancer and other diseases.Molecular Therapy-Nucleic Acids (2014) 3, e143; doi:10.1038/mtna.2013.70; published online 21 January 2014.
Collapse
|
202
|
Meyer C, Berg K, Eydeler-Haeder K, Lorenzen I, Grötzinger J, Rose-John S, Hahn U. Stabilized Interleukin-6 receptor binding RNA aptamers. RNA Biol 2013; 11:57-65. [PMID: 24440854 DOI: 10.4161/rna.27447] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Interleukin-6 (IL-6) is a multifunctional cytokine that is involved in the progression of various inflammatory diseases, such as rheumatoid arthritis and certain cancers; for example, multiple myeloma or hepatocellular carcinoma. To interfere with IL-6-dependent diseases, targeting IL-6 receptor (IL-6R)-presenting tumor cells using aptamers might be a valuable strategy to broaden established IL-6- or IL-6R-directed treatment regimens. Recently, we reported on the in vitro selection of RNA aptamers binding to the human IL-6 receptor (IL-6R) with nanomolar affinity. One aptamer, namely AIR-3A, was 19 nt in size and able to deliver bulky cargos into IL-6R-presenting cells. As AIR-3A is a natural RNA molecule, its use for in vivo applications might be limited due to its susceptibility to ubiquitous ribonucleases. Aiming at more robust RNA aptamers targeting IL-6R, we now report on the generation of stabilized RNA aptamers for potential in vivo applications. The new 2'-F-modified RNA aptamers bind to IL-6R via its extracellular portion with low nanomolar affinity comparable to the previously identified unmodified counterpart. Aptamers do not interfere with the IL-6 receptor complex formation. The work described here represents one further step to potentially apply stabilized IL-6R-binding RNA aptamers in IL-6R-connected diseases, like multiple myeloma and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Cindy Meyer
- Laboratory of RNA Molecular Biology; Howard Hughes Medical Institute; The Rockefeller University; New York, NY USA
| | - Katharina Berg
- Institute of Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Katja Eydeler-Haeder
- Institute of Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Inken Lorenzen
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Ulrich Hahn
- Institute of Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| |
Collapse
|
203
|
Scheller J, Garbers C, Rose-John S. Interleukin-6: from basic biology to selective blockade of pro-inflammatory activities. Semin Immunol 2013; 26:2-12. [PMID: 24325804 DOI: 10.1016/j.smim.2013.11.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/13/2013] [Indexed: 12/16/2022]
Abstract
Cytokines receptors exist in membrane bound and soluble form. A soluble form of the human IL-6R is generated by limited proteolysis and alternative splicing. The complex of IL-6 and soluble IL-6R stimulates target cells not stimulated by IL-6 alone, since they do not express the membrane bound IL-6R. We have named this process trans-signaling. Soluble gp130 is the natural inhibitor of IL-6/soluble IL-6R complex responses. Recombinant soluble gp130 protein is a molecular tool to discriminate between gp130 responses via membrane bound and soluble IL-6R responses. Neutralizing monoclonal antibodies for global blockade of IL-6 signaling and the sgp130Fc protein for selective blockade of IL-6 trans-signaling have been used in several animal models of human diseases. Using the sgp130Fc protein or sgp130Fc transgenic mice we demonstrate in models of inflammatory bowel disease, peritonitis, rheumatoid arthritis, atherosclerosis pancreatitis, colon cancer, ovarian cancer and pancreatic cancer, that IL-6 trans-signaling via the soluble IL-6R is the crucial step in the development and the progression of the disease. Therefore, sgp130Fc is a novel therapeutic agent for the treatment of chronic inflammatory diseases and cancer and it undergoes phase I clinical trials as an anti-inflammatory drug since June 2013.
Collapse
Affiliation(s)
- Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Christoph Garbers
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany.
| |
Collapse
|
204
|
Trilling M, Le VTK, Rashidi-Alavijeh J, Katschinski B, Scheller J, Rose-John S, Androsiac GE, Jonjić S, Poli V, Pfeffer K, Hengel H. “Activated” STAT Proteins: A Paradoxical Consequence of Inhibited JAK-STAT Signaling in Cytomegalovirus-Infected Cells. THE JOURNAL OF IMMUNOLOGY 2013; 192:447-58. [DOI: 10.4049/jimmunol.1203516] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
205
|
Mikucki ME, Fisher DT, Ku AW, Appenheimer MM, Muhitch JB, Evans SS. Preconditioning thermal therapy: flipping the switch on IL-6 for anti-tumour immunity. Int J Hyperthermia 2013; 29:464-73. [PMID: 23862980 DOI: 10.3109/02656736.2013.807440] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy aims to generate long-lived, tumour-specific adaptive immunity to limit dysregulated tumour progression and metastasis. Tumour vasculature has emerged as a critical checkpoint controlling the efficacy of immunotherapy since it is the main access point for cytotoxic T cells to reach tumour cell targets. Therapeutic success has been particularly challenging to achieve because of the local, cytokine-rich inflammatory milieu that drives a pro-tumourigenic programme supporting the growth and survival of malignant cells. Here, we focus on recent evidence that systemic thermal therapy can switch the activities of the inflammatory cytokine, interleukin-6 (IL-6), to a predominantly anti-tumourigenic function that promotes anti-tumour immunity by mobilising T cell trafficking in the recalcitrant tumour microenvironment.
Collapse
Affiliation(s)
- Maryann E Mikucki
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | |
Collapse
|
206
|
|
207
|
Sörensen-Zender I, Rong S, Susnik N, Lange J, Gueler F, Degen JL, Melk A, Haller H, Schmitt R. Role of fibrinogen in acute ischemic kidney injury. Am J Physiol Renal Physiol 2013; 305:F777-85. [PMID: 23804451 DOI: 10.1152/ajprenal.00418.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal ischemia-reperfusion (I/R) is associated with activation of the coagulation system and accumulation of blood clotting factors in the kidney. The aim of the present study was to examine the functional impact of fibrinogen on renal inflammation, damage, and repair in the context of I/R injury. In this study, we found that I/R was associated with a significant increase in the renal deposition of circulating fibrinogen. In parallel, I/R stress induced the de novo expression of fibrinogen in tubular epithelial cells, as reflected by RT-PCR, immunofluorescence, and in situ hybridization. In vitro, fibrinogen expression was induced by oncostatin M and hyper-IL-6 in primary tubular epithelial cells, and fibrinogen-containing medium had an inhibitory effect on tubular epithelial cell adhesion and migration. Fibrinogen(+/-) mice showed similar survival as wild-type mice but better preservation in early postischemic renal function. In fibrinogen(-/-) mice, renal function and survival were significantly worse than in fibrinogen(+/-) mice. Renal transplant experiments revealed reduced expression of tubular damage markers and attenuated proinflammatory cytokine expression but increased inflammatory cell infiltrates and transforming growth factor-β expression in fibrinogen(-/-) isografts. These data point to heterogeneous effects of fibrinogen in renal I/R injury. While a complete lack of fibrinogen may be detrimental, partial reduction of fibrinogen in heterozygous mice can improve renal function and overall outcome.
Collapse
Affiliation(s)
- I Sörensen-Zender
- Department of Nephrology and Hypertension, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Floss DM, Mrotzek S, Klöcker T, Schröder J, Grötzinger J, Rose-John S, Scheller J. Identification of canonical tyrosine-dependent and non-canonical tyrosine-independent STAT3 activation sites in the intracellular domain of the interleukin 23 receptor. J Biol Chem 2013; 288:19386-400. [PMID: 23673666 DOI: 10.1074/jbc.m112.432153] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Signaling of interleukin 23 (IL-23) via the IL-23 receptor (IL-23R) and the shared IL-12 receptor β1 (IL-12Rβ1) controls innate and adaptive immune responses and is involved in the differentiation and expansion of IL-17-producing CD4(+) T helper (TH17) cells. Activation of signal transducer and activator of transcription 3 (STAT3) appears to be the major signaling pathway of IL-23, and STAT binding sites were predicted in the IL-23R but not in the IL-12Rβ1 chain. Using site-directed mutagenesis and deletion variants of the murine and human IL-23R, we showed that the predicted STAT binding sites (pYXXQ; including Tyr-504 and Tyr-626 in murine IL-23R and Tyr-484 and Tyr-611 in human IL-23R) mediated STAT3 activation. Furthermore, we identified two uncommon STAT3 binding/activation sites within the murine IL-23R. First, the murine IL-23R carried the Y(542)PNFQ sequence, which acts as an unusual Src homology 2 (SH2) domain-binding protein activation site of STAT3. Second, we identified a non-canonical, phosphotyrosine-independent STAT3 activation motif within the IL-23R. A third predicted site, Tyr-416 in murine and Tyr-397 in human IL-23R, is involved in the activation of PI3K/Akt and the MAPK pathway leading to STAT3-independent proliferation of Ba/F3 cells upon stimulation with IL-23. In contrast to IL-6-induced short term STAT3 phosphorylation, cellular activation by IL-23 resulted in a slower but long term STAT3 phosphorylation, indicating that the IL-23R might not be a major target of negative feedback inhibition by suppressor of cytokine signaling (SOCS) proteins. In summary, we characterized IL-23-dependent signal transduction with a focus on STAT3 phosphorylation and identified canonical tyrosine-dependent and non-canonical tyrosine-independent STAT3 activation sites in the IL-23R.
Collapse
Affiliation(s)
- Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | | | | | | | | | | | | |
Collapse
|
209
|
Leibinger M, Müller A, Gobrecht P, Diekmann H, Andreadaki A, Fischer D. Interleukin-6 contributes to CNS axon regeneration upon inflammatory stimulation. Cell Death Dis 2013; 4:e609. [PMID: 23618907 PMCID: PMC3641349 DOI: 10.1038/cddis.2013.126] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mature retinal ganglion cells (RGCs) do not normally regenerate injured axons and undergo apoptosis after axotomy. Inflammatory stimulation (IS) in the eye mediates neuroprotection and induces axon regeneration into the injured optic nerve. Ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF) have been identified as key mediators of these effects. Here, we investigated the role of interleukin-6 (IL-6), another member of the glycoprotein 130-activating cytokine family, as additonal contributing factor. Expression of IL-6 was markedly induced in the retina upon optic nerve injury and IS, and mature RGCs expressed the IL-6 receptor. Treatment of cultured RGCs with IL-6 or specific IL-6 receptor agonist, significantly increased neurite outgrowth janus kinase/signal transducers and activators of transcription-3 (JAK/STAT3) and phosphatidylinositide 3-kinase/protein kinase B (PI3K/Akt) dependently. Moreover, IL-6 reduced myelin, but not neurocan-mediated growth inhibition mammalian target of rapamycin (mTOR) dependently in cultured RGCs. In vivo, intravitreal application of IL-6 transformed RGCs into a regenerative state, enabling axon regeneration beyond the lesion site of the optic nerve. On the other hand, genetic ablation of IL-6 in mice significantly reduced IS-mediated myelin disinhibition and axon regeneration in the optic nerve. Therefore, IL-6 contributes to the beneficial effects of IS and its disinhibitory effect adds an important feature to the effects of so far identified IS-mediating factors. Consequently, application of IL-6 or activation of its receptor might provide suitable strategies for enhancing optic nerve regeneration.
Collapse
Affiliation(s)
- M Leibinger
- Department of Neurology, Heinrich-Heine-University of Düsseldorf, Merowingerplatz 1a, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
210
|
Baran P, Nitz R, Grötzinger J, Scheller J, Garbers C. Minimal interleukin 6 (IL-6) receptor stalk composition for IL-6 receptor shedding and IL-6 classic signaling. J Biol Chem 2013; 288:14756-68. [PMID: 23564454 DOI: 10.1074/jbc.m113.466169] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Signaling of the pleiotropic cytokine Interleukin-6 (IL-6) is coordinated by membrane-bound and soluble forms of the IL-6 receptor (IL-6R) in processes called classic and trans-signaling, respectively. The soluble IL-6R is mainly generated by ADAM10- and ADAM17-mediated ectodomain shedding. Little is known about the role of the 52-amino acid-residue-long IL-6R stalk region in shedding and signal transduction. Therefore, we generated and analyzed IL-6R stalk region deletion variants for cleavability and biological activity. Deletion of 10 amino acids of the stalk region surrounding the ADAM17 cleavage site substantially blocked IL-6R proteolysis by ADAM17 but only slightly affected proteolysis by ADAM10. Interestingly, additional deletion of the remaining five juxtamembrane-located amino acids also abrogated ADAM10-mediated IL-6R shedding. Larger deletions within the stalk region, that do not necessarily include the ADAM17 cleavage site, also reduced ADAM10 and ADAM17-mediated IL-6R shedding, questioning the importance of cleavage site recognition. Furthermore, we show that a 22-amino acid-long stalk region is minimally required for IL-6 classic signaling. The gp130 cytokine binding sites are separated from the plasma membrane by ~96 Å. 22 amino acid residues, however, span maximally 83.6 Å (3.8 Å/amino acid), indicating that the three juxtamembrane fibronectin domains of gp130 are not necessarily elongated but somehow flexed to allow IL-6 classic signaling. Our findings underline a dual role of the IL-6R stalk region in IL-6 signaling. In IL-6 trans-signaling, it regulates proper proteolysis by ADAM10 and ADAM17. In IL-6 classic-signaling, it acts as a spacer to ensure IL-6·IL-6R·gp130 signal complex formation.
Collapse
Affiliation(s)
- Paul Baran
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
211
|
Wang K, Grivennikov SI, Karin M. Implications of anti-cytokine therapy in colorectal cancer and autoimmune diseases. Ann Rheum Dis 2013; 72 Suppl 2:ii100-3. [PMID: 23253923 DOI: 10.1136/annrheumdis-2012-202201] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up to 20% of all cancers have been linked to chronic inflammation and persistent infections. However, almost all solid tumours contain immune infiltrates, and tumour-associated inflammatory cells play broad roles in different stages of tumour development and malignant progression. Cytokines are important mediators of the inflammatory effect on tumorigenesis both in inflammation-induced cancer and in the inflammation that follows tumour development. We have shown interleukin (IL)-6 to be an important tumour promoter in early colitis-associated cancer (CAC). IL-6 is mainly produced by tumour-infiltrating myeloid cells under the control of NF-κB. IL-6 promotes proliferation of tumour-initiating cells derived from the intestinal epithelium and protects them from apoptotic elimination. These pro-survival and proliferative effects of IL-6 are mainly mediated by STAT3, whose ablation in intestinal epithelial cells significantly reduces CAC tumorigenesis. More recently, we found a critical role for IL-23 and its downstream cytokines IL-17 and IL-22 in the development of CAC. These findings suggest that such cytokines or the cells that produce them may provide new therapeutic or preventive targets in forms of colorectal cancer that are linked to inflammation.
Collapse
Affiliation(s)
- Kepeng Wang
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology and Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0723, USA
| | | | | |
Collapse
|
212
|
Zhang H, Neuhöfer P, Song L, Rabe B, Lesina M, Kurkowski MU, Treiber M, Wartmann T, Regnér S, Thorlacius H, Saur D, Weirich G, Yoshimura A, Halangk W, Mizgerd JP, Schmid RM, Rose-John S, Algül H. IL-6 trans-signaling promotes pancreatitis-associated lung injury and lethality. J Clin Invest 2013; 123:1019-1031. [PMID: 23426178 PMCID: PMC3582130 DOI: 10.1172/jci64931] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 12/17/2012] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) is an inflammatory disease with a high mortality rate. Although typically seen in individuals with sepsis, ALI is also a major complication in severe acute pancreatitis (SAP). The pathophysiology of SAP-associated ALI is poorly understood, but elevated serum levels of IL-6 is a reliable marker for disease severity. Here, we used a mouse model of acute pancreatitis-associated (AP-associated) ALI to determine the role of IL-6 in ALI lethality. Il6-deficient mice had a lower death rate compared with wild-type mice with AP, while mice injected with IL-6 were more likely to develop lethal ALI. We found that inflammation-associated NF-κB induced myeloid cell secretion of IL-6, and the effects of secreted IL-6 were mediated by complexation with soluble IL-6 receptor, a process known as trans-signaling. IL-6 trans-signaling stimulated phosphorylation of STAT3 and production of the neutrophil attractant CXCL1 in pancreatic acinar cells. Examination of human samples revealed expression of IL-6 in combination with soluble IL-6 receptor was a reliable predictor of ALI in SAP. These results demonstrate that IL-6 trans-signaling is an essential mediator of ALI in SAP across species and suggest that therapeutic inhibition of IL-6 may prevent SAP-associated ALI.
Collapse
Affiliation(s)
- Hong Zhang
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Patrick Neuhöfer
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Liang Song
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Björn Rabe
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Marina Lesina
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Magdalena U. Kurkowski
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Matthias Treiber
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thomas Wartmann
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sara Regnér
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Henrik Thorlacius
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Dieter Saur
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Gregor Weirich
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Akihiko Yoshimura
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Walter Halangk
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Joseph P. Mizgerd
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Roland M. Schmid
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Stefan Rose-John
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Hana Algül
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
Department of Surgery, Division of Experimental Surgery, Otto-von-Guericke University, Magdeburg, Germany.
Department of Clinical Sciences, Malmö, Section for Surgery, Lund University, Malmö, Sweden.
Pathologisches Institut, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany.
Department of Microbiology and Immunology, Keio University School of Tokyo, and Japan Science and Technology Agency (JST), CREST, Chiyoda-ku, Tokyo, Japan.
Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
213
|
Abstract
Activation of the IL-6 (interleukin 6) receptor subunit gp130 (glycoprotein 130) has been linked to the formation of complexes with IL-6 and the IL-6 receptor, as well as to gp130 dimerization. However, it has been shown that gp130 is present as a pre-formed dimer, indicating that its activation is not solely dependent on dimerization. Therefore the detailed mechanism of gp130 activation still remains to be deciphered. Recently, deletion mutations of gp130 have been found in inflammatory hepatocellular adenoma. The mutations clustered around one IL-6-binding epitope of gp130 and resulted in a ligand-independent constitutively active gp130. We therefore hypothesized that conformational changes of this particular IL-6-binding epitope precedes gp130 activation. Using a rational structure-based approach we identified for the first time amino acids critical for gp130 activation. We can show that gp130 D2–D3 interdomain connectivity by hydrophobic residues stabilizes inactive gp130 conformation. Conformational destabilization of the EF loop present in domain D2 and disruption of D2–D3 hydrophobic interactions resulted in ligand-independent gp130 activation. Furthermore we show that the N-terminal amino acid residues of domain D1 participate in the activation of the gp130 deletion mutants. Taken together we present novel insights into the molecular basis of the activation of a cytokine receptor signalling subunit.
Collapse
|
214
|
Wu J, Rostami MR, Tzanakakis ES. Stem cell modeling: From gene networks to cell populations. Curr Opin Chem Eng 2013; 2:17-25. [PMID: 23914346 DOI: 10.1016/j.coche.2013.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Despite rapid advances in the field of stem/progenitor cells through experimental studies, relevant modeling approaches have not progressed with a similar pace. Various models have focused on particular aspects of stem cell physiology including gene regulatory networks, gene expression noise and signaling cascades activated by exogenous factors. However, the self-renewal and differentiation of stem cells is driven by the coordinated regulation of events at the subcellular, intercellular and milieu levels. Such events also span multiple time domains from the fast molecular reactions governing gene expression to the slower cell cycle and division. Thus, the development of multiscale computational frameworks for stem cell populations is highly desirable. Multiscale models are expected to aid the design of efficient differentiation strategies and bioprocesses for the generation of therapeutically useful stem cell progeny. Yet, challenges in making these models tractable and pairing those to sufficient experimental data prevent their wide adoption by the stem cell community. Here, we review modeling approaches reported for stem cell populations and associated hurdles.
Collapse
Affiliation(s)
- Jincheng Wu
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, NY 14260
| | | | | |
Collapse
|
215
|
Magbanua E, Zivkovic T, Hansen B, Beschorner N, Meyer C, Lorenzen I, Grötzinger J, Hauber J, Torda AE, Mayer G, Rose-John S, Hahn U. d(GGGT) 4 and r(GGGU) 4 are both HIV-1 inhibitors and interleukin-6 receptor aptamers. RNA Biol 2013; 10:216-27. [PMID: 23235494 PMCID: PMC3594281 DOI: 10.4161/rna.22951] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aptamers are oligonucleotides that bind targets with high specificity and affinity. They have become important tools for biosensing, target detection, drug delivery and therapy. We selected the quadruplex-forming 16-mer DNA aptamer AID-1 [d(GGGT) 4] with affinity for the interleukin-6 receptor (IL-6R) and identified single nucleotide variants that showed no significant loss of binding ability. The RNA counterpart of AID-1 [r(GGGU) 4] also bound IL-6R as quadruplex structure. AID-1 is identical to the well-known HIV inhibitor T30923, which inhibits both HIV infection and HIV-1 integrase. We also demonstrated that IL-6R specific RNA aptamers not only bind HIV-1 integrase and inhibit its 3' processing activity in vitro, but also are capable of preventing HIV de novo infection with the same efficacy as the established inhibitor T30175. All these aptamer target interactions are highly dependent on formation of quadruplex structure.
Collapse
Affiliation(s)
- Eileen Magbanua
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Tijana Zivkovic
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Björn Hansen
- Centre for Bioinformatics; Hamburg University; Hamburg, Germany
| | - Niklas Beschorner
- Heinrich Pette Institute; Leibnitz Institute for Experimental Virology; Hamburg, Germany
| | - Cindy Meyer
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Inken Lorenzen
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Joachim Hauber
- Heinrich Pette Institute; Leibnitz Institute for Experimental Virology; Hamburg, Germany
| | - Andrew E. Torda
- Centre for Bioinformatics; Hamburg University; Hamburg, Germany
| | - Günter Mayer
- Life and Medical Sciences Institute; University of Bonn; Bonn, Germany
| | - Stefan Rose-John
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Ulrich Hahn
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
- Correspondence to: Ulrich Hahn,
| |
Collapse
|
216
|
Thiem S, Pierce TP, Palmieri M, Putoczki TL, Buchert M, Preaudet A, Farid RO, Love C, Catimel B, Lei Z, Rozen S, Gopalakrishnan V, Schaper F, Hallek M, Boussioutas A, Tan P, Jarnicki A, Ernst M. mTORC1 inhibition restricts inflammation-associated gastrointestinal tumorigenesis in mice. J Clin Invest 2013; 123:767-81. [PMID: 23321674 DOI: 10.1172/jci65086] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/27/2012] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal cancers are frequently associated with chronic inflammation and excessive secretion of IL-6 family cytokines, which promote tumorigenesis through persistent activation of the GP130/JAK/STAT3 pathway. Although tumor progression can be prevented by genetic ablation of Stat3 in mice, this transcription factor remains a challenging therapeutic target with a paucity of clinically approved inhibitors. Here, we uncovered parallel and excessive activation of mTOR complex 1 (mTORC1) alongside STAT3 in human intestinal-type gastric cancers (IGCs). Furthermore, in a preclinical mouse model of IGC, GP130 ligand administration simultaneously activated mTORC1/S6 kinase and STAT3 signaling. We therefore investigated whether mTORC1 activation was required for inflammation-associated gastrointestinal tumorigenesis. Strikingly, the mTORC1-specific inhibitor RAD001 potently suppressed initiation and progression of both murine IGC and colitis-associated colon cancer. The therapeutic effect of RAD001 was associated with reduced tumor vascularization and cell proliferation but occurred independently of STAT3 activity. We analyzed the mechanism of GP130-mediated mTORC1 activation in cells and mice and revealed a requirement for JAK and PI3K activity but not for GP130 tyrosine phosphorylation or STAT3. Our results suggest that GP130-dependent activation of the druggable PI3K/mTORC1 pathway is required for inflammation-associated gastrointestinal tumorigenesis. These findings advocate clinical application of PI3K/mTORC1 inhibitors for the treatment of corresponding human malignancies.
Collapse
Affiliation(s)
- Stefan Thiem
- Ludwig Institute for Cancer Research, Melbourne-Parkville Branch, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Abstract
Interleukin-6 (IL-6) is a cytokine which is involved in many inflammatory processes and in the development of cancer. In addition, IL-6 has been shown to be important for the induction of hepatic acute-phase proteins, for the regeneration of the liver and for the stimulation of B-cells. IL-6 binds to a transmembrane IL-6 receptor (IL-6R), which is present on hepatocytes and some leukocytes. The complex of IL-6 and IL-6R associates with a second protein, gp130, which is expressed on all cells of the body. Since neither IL-6 nor IL-6R has a measurable affinity for gp130, cells, which do not express IL-6R, are not responsive to the cytokine IL-6. It could be shown, however, that a naturally occurring soluble IL-6R (sIL-6R) in complex with IL-6 can bind to gp130 on cells with no IL-6R expression. Therefore, cells shedding the sIL-6R render cells, which only express gp130, responsive to the cytokine. This process has been called trans-signaling. In the present chapter, we summarize the known activities of IL-6 with a special emphasis on regenerative activities, which often depend on the sIL-6R. A designer cytokine called Hyper-IL-6, which is a fusion protein of IL-6 and the sIL-6R, can mimic IL-6 trans-signaling responses in vitro and in vivo with considerably higher efficacy than the combination of the natural proteins IL-6 and sIL-6R. We present recent examples from animal models in which the therapeutic potential of Hyper-IL-6 has been evaluated. We propose that Hyper-IL-6 can be used to induce potent regeneration responses in liver, kidney, and other tissues and therefore will be a novel therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Eithan Galun
- Goldyne Savad Inst. of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | |
Collapse
|
218
|
Brunssen SH, Moy SS, Toews AD, McPherson CA, Harry GJ. Interleukin-6 (IL-6) receptor/IL-6 fusion protein (Hyper IL-6) effects on the neonatal mouse brain: possible role for IL-6 trans-signaling in brain development and functional neurobehavioral outcomes. Brain Behav Immun 2013; 27:42-53. [PMID: 22985907 PMCID: PMC3565379 DOI: 10.1016/j.bbi.2012.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 08/17/2012] [Accepted: 08/30/2012] [Indexed: 12/30/2022] Open
Abstract
Adverse neurodevelopmental outcomes are linked to perinatal production of inflammatory mediators, including interleukin 6 (IL-6). While a pivotal role for maternal elevation in IL-6 has been established in determining neurobehavioral outcomes in the offspring and considered the primary target mediating the fetal inflammatory response, questions remain as to the specific actions of IL-6 on the developing brain. CD-1 male mice received a subdural injection of the bioactive fusion protein, hyper IL-6 (HIL-6) on postnatal-day (PND)4 and assessed from preweaning until adulthood. Immunohistochemical evaluation of astrocytes and microglia and mRNA levels for pro-inflammatory cytokines and host response genes indicated no evidence of an acute neuroinflammatory injury response. HIL-6 accelerated motor development and increased reactivity to stimulation and number of entries in a light/dark chamber, decreased ability to learn to withhold a response in passive avoidance, and effected deficits in social novelty behavior. No changes were observed in motor activity, pre-pulse startle inhibition, or learning and memory in the Morris water maze or radial arm maze, as have been reported for models of more severe developmental neuroinflammation. In young animals, mRNA levels for MBP and PLP/DM20 decreased and less complexity of MBP processes in the cortex was evident by immunohistochemistry. The non-hydroxy cerebroside fraction of cerebral lipids was increased. These results provide evidence for selective effects of IL-6 signaling, particularly trans-signaling, in the developing brain in the absence of a general neuroinflammatory response. These data contribute to our further understanding of the multiple aspects of IL-6 signaling in the developing brain.
Collapse
Affiliation(s)
- Susan H. Brunssen
- School of Nursing, University of North Carolina, North Carolina
- Carolina Institute for Developmental Disabilities, University of North Carolina, North Carolina
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Sheryl S. Moy
- Carolina Institute for Developmental Disabilities, University of North Carolina, North Carolina
| | - Arrel D. Toews
- Department of Cellular and Molecular Biology, University of North Carolina, North Carolina
| | - Christopher A. McPherson
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - G. Jean Harry
- National Toxicology Program Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| |
Collapse
|
219
|
Garbers C, Spudy B, Aparicio-Siegmund S, Waetzig GH, Sommer J, Hölscher C, Rose-John S, Grötzinger J, Lorenzen I, Scheller J. An interleukin-6 receptor-dependent molecular switch mediates signal transduction of the IL-27 cytokine subunit p28 (IL-30) via a gp130 protein receptor homodimer. J Biol Chem 2012; 288:4346-54. [PMID: 23209286 DOI: 10.1074/jbc.m112.432955] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IL-27 consists of the cytokine subunit p28 and the non-signaling α-receptor EBI3. p28 was shown to additionally act via the non-signaling membrane-bound IL-6 α-receptor (IL-6R) as an agonistic cytokine but also as a gp130 β-receptor antagonist, leading to inhibition of IL-6 signaling. Here, we developed a strategy for bacterial expression, purification, and refolding of murine p28. We show that p28 did not interfere with IL-6- or IL-27-induced signaling, indicating that p28 has no antagonistic properties. Moreover, we demonstrate that murine p28 acts as an agonistic cytokine via the murine and human IL-6R, indicating that p28 exhibits no species specificity. p28 was able to induce p28-trans-signaling via the soluble IL-6R (sIL-6R), a characteristic property that was initially described for trans-signaling of IL-6 via the sIL-6R. Of notice, p28/sIL-6R trans-signaling was inhibited by the IL-6 trans-signaling antagonist, soluble gp130. At higher concentrations, p28 but not IL-6 was able to induce signaling even in the absence of IL-6R or EBI3. Although IL-27 signals via a heterodimer of the β-receptor chains gp130 and Wsx-1, p28/IL-6R specifically recruits two gp130 β-receptor chains for signal transduction. The binding of p28 to a gp130/Wsx-1 heterodimer or a gp130 homodimer is highly selective and controlled by a novel molecular switch induced by EBI3 or IL-6R, respectively.
Collapse
Affiliation(s)
- Christoph Garbers
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Rose-John S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int J Biol Sci 2012; 8:1237-47. [PMID: 23136552 PMCID: PMC3491447 DOI: 10.7150/ijbs.4989] [Citation(s) in RCA: 750] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 08/15/2012] [Indexed: 02/06/2023] Open
Abstract
Interleukin-6 (IL-6) is a cytokine with many activities. It has functions in the regulation of the immune system and the nervous system. Furthermore, IL-6 is involved in liver regeneration and in the metabolic control of the body. On target cells, IL-6 binds to an 80 kDa IL-6 receptor (IL-6R). The complex of IL-6 and IL-6R associates with a second protein, gp130, which thereupon dimerizes and initiates intracellular signaling. Whereas gp130 is expressed on all cells, IL-6R is only present on few cells in the body including hepatocytes and some leukocytes. Cells, which do not express IL-6R cannot respond to the cytokine, since gp130 alone has no measurable affinity for IL-6. Interestingly, a soluble form of IL-6R (sIL-6R) comprising the extracellular portion of the receptor can bind IL-6 with a similar affinity as the membrane bound IL-6R. The complex of IL-6 and sIL-6R can bind to gp130 on cells, which do not express the IL-6R, and which are unresponsive to IL-6. This process has been called trans-signaling. Here I will review published evidence that IL-6 trans-signaling is pro-inflammatory whereas classic IL-6 signaling via the membrane bound IL-6R is needed for regenerative or anti-inflammatory activities of the cytokine. Furthermore, the detailed knowledge of IL-6 biology has important consequences for therapeutic strategies aimed at the blockade of the cytokine IL-6.
Collapse
Affiliation(s)
- Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
| |
Collapse
|
221
|
Veverka V, Baker T, Redpath NT, Carrington B, Muskett FW, Taylor RJ, Lawson ADG, Henry AJ, Carr MD. Conservation of functional sites on interleukin-6 and implications for evolution of signaling complex assembly and therapeutic intervention. J Biol Chem 2012; 287:40043-50. [PMID: 23027872 DOI: 10.1074/jbc.m112.405597] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A number of secreted cytokines, such as interleukin-6 (IL-6), are attractive targets for the treatment of inflammatory diseases. We have determined the solution structure of mouse IL-6 to assess the functional significance of apparent differences in the receptor interaction sites (IL-6Rα and gp130) suggested by the fairly low degree of sequence similarity with human IL-6. Structure-based sequence alignment of mouse IL-6 and human IL-6 revealed surprising differences in the conservation of the two distinct gp130 binding sites (IIa and IIIa), which suggests a primacy for site III-mediated interactions in driving initial assembly of the IL-6/IL-6Rα/gp130 ternary complex. This is further supported by a series of direct binding experiments, which clearly demonstrate a high affinity IL-6/IL-6Rα-gp130 interaction via site III but only weak binding via site II. Collectively, our findings suggest a pathway for the evolution of the hexameric, IL-6/IL-6Rα/gp130 signaling complex and strategies for therapeutic targeting. We propose that the signaling complex originally involved specific interactions between IL-6 and IL-6Rα (site I) and between the D1 domain of gp130 and IL-6/IL-6Rα (site III), with the later inclusion of interactions between the D2 and D3 domains of gp130 and IL-6/IL-6Rα (site II) through serendipity. It seems likely that IL-6 signaling benefited from the evolution of a multipurpose, nonspecific protein interaction surface on gp130, now known as the cytokine binding homology region (site II contact surface), which fortuitously contributes to stabilization of the IL-6/IL-6Rα/gp130 signaling complex.
Collapse
Affiliation(s)
- Vaclav Veverka
- Department of Biochemistry, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Sommer J, Effenberger T, Volpi E, Waetzig GH, Bernhardt M, Suthaus J, Garbers C, Rose-John S, Floss DM, Scheller J. Constitutively active mutant gp130 receptor protein from inflammatory hepatocellular adenoma is inhibited by an anti-gp130 antibody that specifically neutralizes interleukin 11 signaling. J Biol Chem 2012; 287:13743-51. [PMID: 22523320 DOI: 10.1074/jbc.m111.349167] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ligand-independent constitutively active gp130 mutants were described to be responsible for the development of inflammatory hepatocellular adenomas (IHCAs). These variants had gain-of-function somatic mutations within the extracellular domain 2 (D2) of the gp130 receptor chain. Cytokine-dependent Ba/F3 cells were transduced with the constitutively active variant of gp130 featuring a deletion in the domain 2 from Tyr-186 to Tyr-190 (gp130ΔYY). These cells showed constitutive phosphorylation of signal transducer and activator of transcription-3 (STAT3) and cytokine-independent proliferation. Deletion of the Ig-like domain 1 (D1) of gp130, but not anti-gp130 mAbs directed against D1, abolished constitutive activation of gp130ΔYY, highlighting that this domain is involved in ligand-independent activation of gp130ΔYY. Moreover, soluble variants of gp130 were not able to inhibit the constitutive activation of gp130ΔYY. However, the inhibition of constitutive activation of gp130ΔYY was achieved by the anti-gp130 mAb B-P4, which specifically inhibits gp130 signaling by IL-11 but not by other IL-6 type cytokines. IL-11 but not IL-6 levels were found previously to be up-regulated in IHCAs, suggesting that mutations in gp130 are leading to IL-11-like signaling. The mAb B-P4 might be a valuable tool to inhibit the constitutive activation of naturally occurring gp130 mutants in IHCAs and rare cases of gp130-associated hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jan Sommer
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
223
|
O'Connor RA, Floess S, Huehn J, Jones SA, Anderton SM. Foxp3⁺ Treg cells in the inflamed CNS are insensitive to IL-6-driven IL-17 production. Eur J Immunol 2012; 42:1174-9. [PMID: 22539291 DOI: 10.1002/eji.201142216] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Foxp3(+) T regulatory (Treg) cells can be induced to produce interleukin (IL)-17 by in vitro exposure to proinflammatory cytokines, drawing into question their functional stability at sites of inflammation. Unlike their splenic counterparts, Treg cells from the inflamed central nervous system (CNS-Treg cells) during EAE resisted conversion to IL-17 production when exposed to IL-6. We show that the highly activated phenotype of CNS-Treg cells includes elevated expression of the Th1-associated molecules CXCR3 and T-bet, but reduced expression of the IL-6 receptor α chain (CD126) and the signaling chain gp130. We found a lack of IL-6 receptor on all CNS CD4(+) T cells, which was reflected by an absence of both classical and trans-IL-6 signaling in CNS CD4(+) cells, compared with their splenic counterparts. We propose that extinguished responsiveness to IL-6 (via down-regulation of CD126 and gp130) stabilizes the regulatory phenotype of activated Treg cells at sites of autoimmune inflammation.
Collapse
Affiliation(s)
- Richard A O'Connor
- Centre for Multiple Sclerosis Research and Centre for Immunology Infection and Evolution, Queen's Medical Research Institute, Medical Research Council/University of Edinburgh Centre for Inflammation Research, Edinburgh, UK
| | | | | | | | | |
Collapse
|
224
|
O'Donoghue RJJ, Knight DA, Richards CD, Prêle CM, Lau HL, Jarnicki AG, Jones J, Bozinovski S, Vlahos R, Thiem S, McKenzie BS, Wang B, Stumbles P, Laurent GJ, McAnulty RJ, Rose-John S, Zhu HJ, Anderson GP, Ernst MR, Mutsaers SE. Genetic partitioning of interleukin-6 signalling in mice dissociates Stat3 from Smad3-mediated lung fibrosis. EMBO Mol Med 2012; 4:939-51. [PMID: 22684844 PMCID: PMC3491826 DOI: 10.1002/emmm.201100604] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 04/16/2012] [Accepted: 05/09/2012] [Indexed: 12/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease that is unresponsive to current therapies and characterized by excessive collagen deposition and subsequent fibrosis. While inflammatory cytokines, including interleukin (IL)-6, are elevated in IPF, the molecular mechanisms that underlie this disease are incompletely understood, although the development of fibrosis is believed to depend on canonical transforming growth factor (TGF)-β signalling. We examined bleomycin-induced inflammation and fibrosis in mice carrying a mutation in the shared IL-6 family receptor gp130. Using genetic complementation, we directly correlate the extent of IL-6-mediated, excessive Stat3 activity with inflammatory infiltrates in the lung and the severity of fibrosis in corresponding gp130757F mice. The extent of fibrosis was attenuated in B lymphocyte-deficient gp130757F;µMT−/− compound mutant mice, but fibrosis still occurred in their Smad3−/− counterparts consistent with the capacity of excessive Stat3 activity to induce collagen 1α1 gene transcription independently of canonical TGF-β/Smad3 signalling. These findings are of therapeutic relevance, since we confirmed abundant STAT3 activation in fibrotic lungs from IPF patients and showed that genetic reduction of Stat3 protected mice from bleomycin-induced lung fibrosis.
Collapse
Affiliation(s)
- Robert J J O'Donoghue
- Ludwig Institute for Cancer Research, Melbourne - Parkville Branch, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Ventura Ferreira MS, Labude N, Walenda G, Adamzyk C, Wagner W, Piroth D, Müller AM, Knüchel R, Hieronymus T, Zenke M, Jahnen-Dechent W, Neuss S. Ex vivoexpansion of cord blood-CD34+cells using IGFBP2and Angptl-5 impairs short-term lymphoid repopulationin vivo. J Tissue Eng Regen Med 2012; 7:944-54. [DOI: 10.1002/term.1486] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 01/19/2012] [Indexed: 12/15/2022]
Affiliation(s)
| | - Norina Labude
- Institute of Pathology; RWTH Aachen University; Germany
| | - Gudrun Walenda
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering Group; RWTH Aachen University; Germany
| | | | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering Group; RWTH Aachen University; Germany
| | - Daniela Piroth
- Department for Gynecology; RWTH Aachen University; Germany
| | - Albrecht M. Müller
- Institute for Medical Radiation and Cell Research; University of Würzburg; Germany
| | - Ruth Knüchel
- Institute of Pathology; RWTH Aachen University; Germany
| | - Thomas Hieronymus
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering Group; RWTH Aachen University; Germany
- Institute for Biomedical Engineering, Department of Cell Biology; RWTH Aachen University; Germany
| | - Martin Zenke
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering Group; RWTH Aachen University; Germany
- Institute for Biomedical Engineering, Department of Cell Biology; RWTH Aachen University; Germany
| | - Willi Jahnen-Dechent
- Helmholtz Institute for Biomedical Engineering, Biointerface Group; RWTH Aachen University; Germany
| | - Sabine Neuss
- Institute of Pathology; RWTH Aachen University; Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Group; RWTH Aachen University; Germany
| |
Collapse
|
226
|
Seré KM, Lin Q, Felker P, Rehage N, Klisch T, Ortseifer I, Hieronymus T, Rose-John S, Zenke M. Dendritic cell lineage commitment is instructed by distinct cytokine signals. Eur J Cell Biol 2012; 91:515-23. [DOI: 10.1016/j.ejcb.2011.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/05/2011] [Accepted: 09/13/2011] [Indexed: 10/15/2022] Open
|
227
|
Mackiewicz A, Mackiewicz J, Wysocki PJ, Wiznerowicz M, Kapcinska M, Laciak M, Rose-John S, Izycki D, Burzykowski T, Karczewska-Dzionk A. Long-term survival of high-risk melanoma patients immunized with a Hyper-IL-6-modified allogeneic whole-cell vaccine after complete resection. Expert Opin Investig Drugs 2012; 21:773-83. [DOI: 10.1517/13543784.2012.684753] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
228
|
Sommer J, Effenberger T, Volpi E, Waetzig GH, Bernhardt M, Suthaus J, Garbers C, Rose-John S, Floss DM, Scheller J. Constitutively Active Mutant gp130 Receptor Protein from Inflammatory Hepatocellular Adenoma Is Inhibited by an Anti-gp130 Antibody That Specifically Neutralizes Interleukin 11 Signaling. J Biol Chem 2012. [DOI: 10.1074/jbc.m112.349167] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
229
|
Dams-Kozlowska H, Gryska K, Kwiatkowska-Borowczyk E, Izycki D, Rose-John S, Mackiewicz A. A designer hyper interleukin 11 (H11) is a biologically active cytokine. BMC Biotechnol 2012; 12:8. [PMID: 22433466 PMCID: PMC3382428 DOI: 10.1186/1472-6750-12-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 03/21/2012] [Indexed: 11/10/2022] Open
Abstract
Background Interleukin 11 (IL-11) is a pleiotropic cytokine with anti-apoptotic, anti-inflammatory and hematopoietic potential. The IL-11 activity is determined by the expression of the IL-11R receptor alpha (IL-11Rα) and the signal transducing subunit β (gp130) on the cell membrane. A recombinant soluble form of the IL-11Rα (sIL-11Rα) in combination with IL-11 acts as an agonist on cells expressing the gp130 molecule. We constructed a designer cytokine Hyper IL-11 (H11), which is exclusively composed of naturally existing components. It contains the full length sIL-11Rα connected with the mature IL-11 protein using their natural sequences only. Such a construct has two major advantages: (i) its components are as close as possible to the natural forms of both proteins and (ii) it lacks an artificial linker what should avoid induction of antibody production. Results The H11 construct was generated, the protein was produced in a baculovirus expression system and was then purified by using ion exchange chromatography. The H11 protein displayed activity in three independent bioassays, (i) it induced acute phase proteins production in HepG2 cells expressing IL-11, IL-11Rα and gp130, (ii) it stimulated the proliferation of B9 cells (cells expressing IL-11Rα and gp130) and (iii) proliferation of Baf/3-gp130 cells (cells not expressing IL-11 and IL-11Rα but gp130). Moreover, the preliminary data indicated that H11 was functionally distinct from Hyper-IL-6, a molecule which utilizes the same homodimer of signal transducing receptor (gp130). Conclusions The biologically active H11 may be potentially useful for treatment of thrombocytopenia, infertility, multiple sclerosis, cardiovascular diseases or inflammatory disorders.
Collapse
Affiliation(s)
- Hanna Dams-Kozlowska
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866 Poznan, Poland.
| | | | | | | | | | | |
Collapse
|
230
|
Sturm JB, Hess M, Weibel S, Chen NG, Yu YA, Zhang Q, Donat U, Reiss C, Gambaryan S, Krohne G, Stritzker J, Szalay AA. Functional hyper-IL-6 from vaccinia virus-colonized tumors triggers platelet formation and helps to alleviate toxicity of mitomycin C enhanced virus therapy. J Transl Med 2012; 10:9. [PMID: 22236378 PMCID: PMC3268093 DOI: 10.1186/1479-5876-10-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/11/2012] [Indexed: 01/08/2023] Open
Abstract
Background Combination of oncolytic vaccinia virus therapy with conventional chemotherapy has shown promise for tumor therapy. However, side effects of chemotherapy including thrombocytopenia, still remain problematic. Methods Here, we describe a novel approach to optimize combination therapy of oncolytic virus and chemotherapy utilizing virus-encoding hyper-IL-6, GLV-1h90, to reduce chemotherapy-associated side effects. Results We showed that the hyper-IL-6 cytokine was successfully produced by GLV-1h90 and was functional both in cell culture as well as in tumor-bearing animals, in which the cytokine-producing vaccinia virus strain was well tolerated. When combined with the chemotherapeutic mitomycin C, the anti-tumor effect of the oncolytic virotherapy was significantly enhanced. Moreover, hyper-IL-6 expression greatly reduced the time interval during which the mice suffered from chemotherapy-induced thrombocytopenia. Conclusion Therefore, future clinical application would benefit from careful investigation of additional cytokine treatment to reduce chemotherapy-induced side effects.
Collapse
Affiliation(s)
- Julia B Sturm
- Department of Biochemistry, University of Würzburg, 97074 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Meyer C, Eydeler K, Magbanua E, Zivkovic T, Piganeau N, Lorenzen I, Grötzinger J, Mayer G, Rose-John S, Hahn U. Interleukin-6 receptor specific RNA aptamers for cargo delivery into target cells. RNA Biol 2012; 9:67-80. [PMID: 22258147 PMCID: PMC3342945 DOI: 10.4161/rna.9.1.18062] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aptamers represent an emerging strategy to deliver cargo molecules, including dyes, drugs, proteins or even genes, into specific target cells. Upon binding to specific cell surface receptors aptamers can be internalized, for example by macropinocytosis or receptor mediated endocytosis. Here we report the in vitro selection and characterization of RNA aptamers with high affinity (Kd = 20 nM) and specificity for the human IL-6 receptor (IL-6R). Importantly, these aptamers trigger uptake without compromising the interaction of IL-6R with its natural ligands the cytokine IL-6 and glycoprotein 130 (gp130). We further optimized the aptamers to obtain a shortened, only 19-nt RNA oligonucleotide retaining all necessary characteristics for high affinity and selective recognition of IL-6R on cell surfaces. Upon incubation with IL-6R presenting cells this aptamer was rapidly internalized. Importantly, we could use our aptamer, to deliver bulky cargos, exemplified by fluorescently labeled streptavidin, into IL-6R presenting cells, thereby setting the stage for an aptamer-mediated escort of drug molecules to diseased cell populations or tissues.
Collapse
Affiliation(s)
- Cindy Meyer
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Katja Eydeler
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Eileen Magbanua
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Tijana Zivkovic
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Nicolas Piganeau
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| | - Inken Lorenzen
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Joachim Grötzinger
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Günter Mayer
- Life and Medical Sciences Institute; University of Bonn; Bonn, Germany
| | - Stefan Rose-John
- Institute of Biochemistry; Medical Faculty; Christian-Albrechts-University; Kiel, Germany
| | - Ulrich Hahn
- Institute for Biochemistry and Molecular Biology; Chemistry Department; MIN-Faculty; Hamburg University; Hamburg, Germany
| |
Collapse
|
232
|
Ding X, Lin Q, Ensenat-Waser R, Rose-John S, Zenke M. Polycomb Group Protein Bmi1 Promotes Hematopoietic Cell Development from Embryonic Stem Cells. Stem Cells Dev 2012; 21:121-32. [DOI: 10.1089/scd.2010.0539] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Xiaolei Ding
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Qiong Lin
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Roberto Ensenat-Waser
- Joint Research Centre, Institute for Health and Consumer Protection, In Vitro Methods Unit, European Commission, Ispra, Italy
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
233
|
Schuett H, Oestreich R, Waetzig GH, Annema W, Luchtefeld M, Hillmer A, Bavendiek U, von Felden J, Divchev D, Kempf T, Wollert KC, Seegert D, Rose-John S, Tietge UJF, Schieffer B, Grote K. Transsignaling of interleukin-6 crucially contributes to atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2011; 32:281-90. [PMID: 22075248 DOI: 10.1161/atvbaha.111.229435] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Transsignaling of interleukin (IL)-6 is a central pathway in the pathogenesis of disorders associated with chronic inflammation, such as Crohn disease, rheumatoid arthritis, and inflammatory colon cancer. Notably, IL-6 also represents an independent risk factor for coronary artery disease (CAD) in humans and is crucially involved in vascular inflammatory processes. METHODS AND RESULTS In the present study, we showed that treatment with a fusion protein of the natural IL-6 transsignaling inhibitor soluble glycoprotein 130 (sgp130) and IgG1-Fc (sgp130Fc) dramatically reduced atherosclerosis in hypercholesterolemic Ldlr(-/-) mice without affecting weight gain and serum lipid levels. Moreover, sgp130Fc treatment even led to a significant regression of advanced atherosclerosis. Mechanistically, endothelial activation and intimal smooth muscle cell infiltration were decreased in sgp130Fc-treated mice, resulting in a marked reduction of monocyte recruitment and subsequent atherosclerotic plaque progression. Of note, patients with CAD exhibited significantly lower plasma levels of endogenous sgp130, suggesting that a compromised counterbalancing of IL-6 transsignaling may contribute to atherogenesis in humans. CONCLUSIONS These data clarify, for the first time, the critical involvement of, in particular, the transsignaling of IL-6 in CAD and warrant further investigation of sgp130Fc as a novel therapeutic for the treatment of CAD and related diseases.
Collapse
Affiliation(s)
- Harald Schuett
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
März-Weiss P, Kunz D, Bimmler D, Berkemeier C, Özbek S, Dimitriades-Schmutz B, Haybaeck J, Otten U, Graf R. Expression of pancreatitis-associated protein after traumatic brain injury: a mechanism potentially contributing to neuroprotection in human brain. Cell Mol Neurobiol 2011; 31:1141-9. [PMID: 21643999 PMCID: PMC11498531 DOI: 10.1007/s10571-011-9715-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 05/20/2011] [Indexed: 12/20/2022]
Abstract
Neuronal cell death after severe traumatic brain injury (TBI) is caused by a complex interplay of pathological mechanisms including excitotoxicity, oxidative stress, mitochondrial dysfunction, extensive neuroinflammation, and ischemia-reperfusion injury. Pancreatitis-associated protein I (PAP I/reg2) was reported to be a survival factor for peripheral neurons, particularly sensory and motor neurons. In rat brains, by experimental TBI as well as by kainic acid induced brain seizure, PAP I and PAP III were found to be up-regulated in central neurons. In this study, we performed immunohistochemical staining in postmortem human brain from patients who died after severe TBI to demonstrate PAP expression on protein level in cerebellar Purkinje cells, pyramidal and granular neurons in cerebral cortex, and cortical neurons in the fore- and mid-brain. In primary cultures of rat brain cortical, hippocampal, and cerebellar neurons, we found neuroprotective effects for PAP I on H(2)O(2)-induced oxidative stress. Moreover, serum K(+)-deprivation induces apoptotic cell death in 55% of cerebellar granule neurons (CGN), whereas upon treatment with PAP I only 32% of CGN are apoptotic. Using Western blot analyses, we compared protein phosphorylation in neuronal signaling pathways activated by PAP I versus Interleukin-6 (IL-6). We found a rapid activation of Akt-kinase phosphorylation by PAP I with a peak at 15 min, whereas IL-6 induces Akt-phosphorylation lasting longer than 30 min. Phosphorylation of MAP-42/44 kinases is stimulated in a comparable fashion. Both, IL-6 and PAP I increase phosphorylation of NFκB for activation of gene transcription, whereas only IL-6 recruits STAT3 phosphorylation, indicating that STAT3 is not a target of PAP I transcription activation in brain neurons. Application of the Akt-inhibitor Wortmanin reveals only a partial inhibition of PAP I-dependent protection of CGN from H(2)O(2)-induced oxidative stress. Based on our findings, we suggest that PAP I is a long lasting neurotrophic signal for central neurons. The neuroprotective effects parallel those that have been described for effects of PAP I in ciliary neurotrophic factor (CNTF)-mediated survival of sensory and motor neurons. PAP I may act in autocrine and/or paracrine fashion and thus may contribute to endogenous protective mechanisms relevant under harmful conditions like oxidative stress, brain injury, or neurodegeneration.
Collapse
Affiliation(s)
- Pia März-Weiss
- Translational Research, Hoffmann-LaRoche Ltd, Grenzacherstrasse, 4070 Basel, Switzerland
| | - Dieter Kunz
- Institute of Physiology, University of Basel, Pestalozzistr. 20, 4056 Basel, Switzerland
| | - Daniel Bimmler
- Pancreatitis Research Laboratory, Department of Surgery, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Caroline Berkemeier
- Institute of Physiology, University of Basel, Pestalozzistr. 20, 4056 Basel, Switzerland
| | - Suat Özbek
- Institute for Molecular Evolution and Genomics, University of Heidelberg, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | | | - Johannes Haybaeck
- Institute of Neuropathology, University Hospital Zürich, 8091 Zürich, Switzerland
- Institute of Pathology, Medical University Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - Uwe Otten
- Institute of Physiology, University of Basel, Pestalozzistr. 20, 4056 Basel, Switzerland
| | - Rolf Graf
- Pancreatitis Research Laboratory, Department of Surgery, University Hospital Zürich, 8091 Zürich, Switzerland
| |
Collapse
|
235
|
Suchorska WM, Dams-Kozlowska H, Kazimierczak U, Wysocki PJ, Mackiewicz A. Hyper-interleukin-11 novel designer molecular adjuvant targeting gp130 for whole cell cancer vaccines. Expert Opin Biol Ther 2011; 11:1555-67. [PMID: 21995459 DOI: 10.1517/14712598.2011.627852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Hyper-IL-11 (H11) is a fusion protein comprising IL-11 and soluble IL-11 receptor directly targeting gp130. We evaluated efficacy of H11 as a molecular adjuvant in therapeutic whole tumor cell vaccine formulation. METHODS H11 was tested in ectopic and orthotopic murine renal cell carcinoma (RENCA) models. H11 cDNA was transduced into RENCA cells (RENCA-H11). Mice were immunized with RENCA-H11 or control vaccine (RENCA-IRR) in prophylactic, adjuvant and therapeutic settings. Tumor formation, survival and immune mechanisms activated by H11 were studied. RESULTS Biologically active H11 was secreted by RENCA-H11 cells. Immunization with RENCA-H11 resulted in mounting specific anti-RENCA response. Treatment of tumor bearing mice in adjuvant setting prevented disease recurrence in therapeutic setting eradicated tumors. In induction phase H11 inhibited T-regulatory cell formation and activated recruitment and maturation of dendritic cells. Downstream of immunization tumors were densely infiltrated by CD8(+), CD4(+), NK cells, cells expressing CD8(+)CD69(+) and CD4(+)CD62L(low). CONCLUSIONS H11 is a good candidate for adjuvant of whole tumor cell vaccines. Direct targeting of gp130 leads to induction of specific and long lasting anticancer immune response. Enhancement of tumor antigen presentation, abrogation of immune tolerance, and activation of NK cells and generation of memory cells lead to eradication of existing tumors.
Collapse
|
236
|
Garbers C, Thaiss W, Jones GW, Waetzig GH, Lorenzen I, Guilhot F, Lissilaa R, Ferlin WG, Grötzinger J, Jones SA, Rose-John S, Scheller J. Inhibition of classic signaling is a novel function of soluble glycoprotein 130 (sgp130), which is controlled by the ratio of interleukin 6 and soluble interleukin 6 receptor. J Biol Chem 2011; 286:42959-70. [PMID: 21990364 DOI: 10.1074/jbc.m111.295758] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
IL-6 trans-signaling via the soluble IL-6 receptor (sIL-6R) plays a critical role in chronic inflammation and cancer. Soluble gp130 (sgp130) specifically inhibits IL-6 trans-signaling but was described to not interfere with classic signaling via the membrane-bound IL-6R. Physiological and most pathophysiological conditions are characterized by a molar excess of serum sIL-6R over IL-6 characterized by free IL-6 and IL-6 found in IL-6·sIL-6R complexes allowing both classic and trans-signaling. Surprisingly, under these conditions, sgp130 was able to trap all free IL-6 molecules in IL-6·sIL-6R·sgp130 complexes, resulting in inhibition of classic signaling. Because a significant fraction of IL-6 molecules did not form complexes with sIL-6R, our results demonstrate that compared with the anti-IL-6R antibody tocilizumab or the anti-trans-signaling monoclonal antibody 25F10, much lower concentrations of the dimeric sgp130Fc were sufficient to block trans-signaling. In vivo, sgp130Fc blocked IL-6 signaling in the colon but not in liver and lung, indicating that the colon is a prominent target of IL-6 trans-signaling. Our results point to a so far unanticipated role of sgp130 in the blockade of classic signaling and indicate that in vivo only low therapeutic concentrations of sgp130Fc guarantee blockade of IL-6 trans-signaling without affecting IL-6 classic signaling.
Collapse
Affiliation(s)
- Christoph Garbers
- Institute of Biochemistry and Molecular Biology II, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Quarta S, Vogl C, Constantin CE, Üçeyler N, Sommer C, Kress M. Genetic evidence for an essential role of neuronally expressed IL-6 signal transducer gp130 in the induction and maintenance of experimentally induced mechanical hypersensitivity in vivo and in vitro. Mol Pain 2011; 7:73. [PMID: 21951917 PMCID: PMC3197546 DOI: 10.1186/1744-8069-7-73] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 09/27/2011] [Indexed: 11/29/2022] Open
Abstract
Tenderness and mechanical allodynia are key symptoms of malignant tumor, inflammation and neuropathy. The proinflammatory cytokine interleukin-6 (IL-6) is causally involved in all three pathologies. IL-6 not only regulates innate immunity and inflammation but also causes nociceptor sensitization and hyperalgesia. In general and in most cell types including immune cells and sensory neurons, IL-6 binds soluble μ receptor subunits which heteromerizes with membrane bound IL-6 signal transducer gp130. In the present study, we used a conditional knock-out strategy to investigate the importance of signal transducer gp130 expressed in C nociceptors for the generation and maintenance of mechanical hypersensitivity. Nociceptors were sensitized to mechanical stimuli by experimental tumor and this nociceptor sensitization was preserved at later stages of the pathology in control mice. However, in mice with a conditional deletion of gp130 in Nav1.8 expressing nociceptors mechanical hypersensitivity by experimental tumor, nerve injury or inflammation recovery was not preserved in the maintenance phase and nociceptors exhibited normal mechanical thresholds comparable to untreated mice. Together, the results argue for IL-6 signal transducer gp130 as an essential prerequisite in nociceptors for long-term mechanical hypersensitivity associated with cancer, inflammation and nerve injury.
Collapse
Affiliation(s)
- Serena Quarta
- Div. Physiology, DPMP, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
238
|
Fisher DT, Chen Q, Skitzki JJ, Muhitch JB, Zhou L, Appenheimer MM, Vardam TD, Weis EL, Passanese J, Wang WC, Gollnick SO, Dewhirst MW, Rose-John S, Repasky EA, Baumann H, Evans SS. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest 2011; 121:3846-59. [PMID: 21926464 DOI: 10.1172/jci44952] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 08/03/2011] [Indexed: 12/26/2022] Open
Abstract
Immune cells are key regulators of neoplastic progression, which is often mediated through their release of cytokines. Inflammatory cytokines such as IL-6 exert tumor-promoting activities by driving growth and survival of neoplastic cells. However, whether these cytokines also have a role in recruiting mediators of adaptive anticancer immunity has not been investigated. Here, we report that homeostatic trafficking of tumor-reactive CD8+ T cells across microvascular checkpoints is limited in tumors despite the presence of inflammatory cytokines. Intravital imaging in tumor-bearing mice revealed that systemic thermal therapy (core temperature elevated to 39.5°C ± 0.5°C for 6 hours) activated an IL-6 trans-signaling program in the tumor blood vessels that modified the vasculature such that it could support enhanced trafficking of CD8+ effector/memory T cells (Tems) into tumors. A concomitant decrease in tumor infiltration by Tregs during systemic thermal therapy resulted in substantial enhancement of Tem/Treg ratios. Mechanistically, IL-6 produced by nonhematopoietic stromal cells acted cooperatively with soluble IL-6 receptor-α and thermally induced gp130 to promote E/P-selectin- and ICAM-1-dependent extravasation of cytotoxic T cells in tumors. Parallel increases in vascular adhesion were induced by IL-6/soluble IL-6 receptor-α fusion protein in mouse tumors and patient tumor explants. Finally, a causal link was established between IL-6-dependent licensing of tumor vessels for Tem trafficking and apoptosis of tumor targets. These findings suggest that the unique IL-6-rich tumor microenvironment can be exploited to create a therapeutic window to boost T cell-mediated antitumor immunity and immunotherapy.
Collapse
Affiliation(s)
- Daniel T Fisher
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Abstract
Quantitative approaches are essential for the advancement of strategies to manipulate stem cells or their derivatives for therapeutic applications. Predictive models of stem cell systems would provide the means to pose and validate non-intuitive hypotheses and could thus serve as an important tool for discerning underlying regulatory mechanisms governing stem cell fate decisions. In this paper we review the development of computational models that attempt to describe mammalian adult and embryonic stem (ES) cell responses. Early stochastic models that relied exclusively on statistical distributions to describe the in vitro or in vivo output of stem cells are being revised to incorporate the contributions of exogenous and endogenous parameters on specific stem cell fate processes. Recent models utilize cell specific data (for example, cell-surface receptor distributions, transcription factor half-lives, cell-cycle status, etc.) to provide mechanistic descriptions that are consistent with biologically observed phenomena. Ultimately, the goal of these computational models is to, a priori, predict stem cell output given an initial set of conditions. Our efforts to develop a predictive model of ES cell fate are discussed. The quantitative studies presented in this review represent an important step in developing bioengineering approaches to characterize and predict stem cell behavior. Ongoing efforts to incorporate genetic and signaling network data into computational models should accelerate our understanding of fundamental principles governing stem cell fate decisions.
Collapse
Affiliation(s)
- Sowmya Viswanathan
- Institute of Biomaterials and Biomedical Engineering and Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
240
|
Nechemia-Arbely Y, Shriki A, Denz U, Drucker C, Scheller J, Raub J, Pappo O, Rose-John S, Galun E, Axelrod JH. Early hepatocyte DNA synthetic response posthepatectomy is modulated by IL-6 trans-signaling and PI3K/AKT activation. J Hepatol 2011; 54:922-9. [PMID: 21145830 DOI: 10.1016/j.jhep.2010.08.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 07/05/2010] [Accepted: 08/03/2010] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Interleukin-6 (IL-6) is a crucial factor in liver regeneration following partial hepatectomy (PH); however, the role of IL-6 and IL-6 trans-signaling in particular, in hepatocyte mitosis remains controversial. IL-6 trans-signaling relies upon the release of the soluble IL-6R (sIL-6R), which binds IL-6 to form an agonistic IL-6/sIL-6R complex. Herein we have examined the hypothesis that IL-6 trans-signaling plays a crucial and distinct role in liver regeneration following PH. METHODS The specific IL-6/sIL-6R antagonist, sgp130Fc, was expressed in mice and analyzed for its effect on hepatocyte mitosis following PH. Alternatively, we examined the effect of the IL-6/sIL-6R super-agonist, Hyper-IL-6, or IL-6 expressed either alone or in combination with hepatocyte growth factor (HGF) on hepatocyte mitosis in the absence of PH. RESULTS Following PH, the dramatic rise of circulating IL-6 levels is accompanied by a concurrent ∼2-fold increase in circulating sIL-6R levels. Ectopic expression of sgp130Fc reduced hepatocyte mitosis by about 40% at early times following PH, while substantially reducing AKT, but not STAT3, activation. But, ectopic Hyper-IL-6 expression in mice without PH was not mitogenic to hepatocytes in vivo. Rather, Hyper-IL-6, but not IL-6, markedly increased HGF-induced hepatocyte mitosis. This cooperative effect correlated with greater resistance of HIL-6 than IL-6 to HGF-mediated reduction of AKT activation, rather than changes in STAT3 or MAPK signaling, and was completely blocked by PI3K inhibition. CONCLUSIONS Following PH, IL-6/sIL-6R cooperates with growth factors, through a PI3K/AKT-dependent mechanism to promote entry of hepatocytes into the cell cycle.
Collapse
Affiliation(s)
- Yael Nechemia-Arbely
- The Goldyne Savad Institute of Gene Therapy, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Garbers C, Jänner N, Chalaris A, Moss ML, Floss DM, Meyer D, Koch-Nolte F, Rose-John S, Scheller J. Species specificity of ADAM10 and ADAM17 proteins in interleukin-6 (IL-6) trans-signaling and novel role of ADAM10 in inducible IL-6 receptor shedding. J Biol Chem 2011; 286:14804-11. [PMID: 21454673 PMCID: PMC3083187 DOI: 10.1074/jbc.m111.229393] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 03/08/2011] [Indexed: 01/11/2023] Open
Abstract
Hypomorphic ADAM17(ex/ex) mice showed defects in mucosal regeneration due to inefficient enhanced GFR shedding. ADAM17 is the main sheddase of interleukin-6 receptor (IL-6R) to induce IL-6 trans-signaling. However, serum levels of soluble murine IL-6R were not reduced in ADAM17(ex/ex) mice, and murine ADAM17 was not the major sheddase of murine IL-6R. Shedding of murine IL-6R by murine ADAM17 was rescued in chimeric murine IL-6R proteins containing any extracellular domain but not the transmembrane and intracellular domain of human IL-6R. Apoptosis is a physiological stimulus of ADAM17-mediated shedding of human IL-6R. Even though apoptosis induced IL-6R shedding in mice, the responsible protease was identified as ADAM10. ADAM10 also was identified as protease responsible for ionomycin-induced shedding of murine and human IL-6R. However, in ADAM10-deficient murine embryonic fibroblasts, compensatory shedding of human IL-6R was mediated by ADAM17, but loss of ADAM10-mediated shedding of murine IL-6R was compensated by an as-yet-unidentified protease. Finally, we identified physiological purinergic P2X7 receptor stimulation as a novel inducer of murine and human IL-6R shedding solely mediated by ADAM10. In conclusion, we describe an unexpected species specificity of ADAM10 and ADAM17 and identified ADAM10 as novel inducible sheddase of IL-6R in mice and humans, which might have consequences for the interpretation of phenotypes from ADAM17- and ADAM10-deficient mice.
Collapse
Affiliation(s)
- Christoph Garbers
- From the Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Nathalie Jänner
- From the Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Athena Chalaris
- From the Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | | | - Doreen M. Floss
- the Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Universitätsstrasse 1, Düsseldorf, Germany, and
| | - Dörte Meyer
- From the Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Friedrich Koch-Nolte
- the Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Germany
| | - Stefan Rose-John
- From the Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Jürgen Scheller
- the Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Universitätsstrasse 1, Düsseldorf, Germany, and
| |
Collapse
|
242
|
Schiechl G, Bauer B, Fuss I, Lang SA, Moser C, Ruemmele P, Rose-John S, Neurath MF, Geissler EK, Schlitt HJ, Strober W, Fichtner-Feigl S. Tumor development in murine ulcerative colitis depends on MyD88 signaling of colonic F4/80+CD11b(high)Gr1(low) macrophages. J Clin Invest 2011; 121:1692-708. [PMID: 21519141 DOI: 10.1172/jci42540] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 03/11/2011] [Indexed: 12/24/2022] Open
Abstract
Patients with prolonged ulcerative colitis (UC) frequently develop colorectal adenocarcinoma for reasons that are not fully clear. To analyze inflammation-associated colonic tumorigenesis, we developed a chronic form of oxazolone-induced colitis in mice that, similar to UC, was distinguished by the presence of IL-13-producing NKT cells. In this model, the induction of tumors using azoxymethane was accompanied by the coappearance of F4/80+CD11b(high)Gr1(low) M2 macrophages, cells that undergo polarization by IL-13 and are absent in tumors that lack high level IL-13 production. Importantly, this subset of macrophages was a source of tumor-promoting factors, including IL-6. Similar to dextran sodium sulfate-induced colitis, F4/80+CD11b(high)Gr1(intermediate) macrophages were present in the mouse model of chronic oxazolone-induced colitis and may influence tumor development through production of TGF-β1, a cytokine that inhibits tumor immunosurveillance. Finally, while robust chronic oxazolone-induced colitis developed in myeloid differentiation primary response gene 88-deficient (Myd88-/-) mice, these mice did not support tumor development. The inhibition of tumor development in Myd88-/- mice correlated with cessation of IL-6 and TGF-β1 production by M2 and F4/80+CD11b(high)Gr1(intermediate) macrophages, respectively, and was reversed by exogenous IL-6. These data show that an UC-like inflammation may facilitate tumor development by providing a milieu favoring development of MyD88-dependent tumor-supporting macrophages.
Collapse
Affiliation(s)
- Gabriela Schiechl
- Department of Surgery, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Kamiya S, Okumura M, Chiba Y, Fukawa T, Nakamura C, Nimura N, Mizuguchi J, Wada S, Yoshimoto T. IL-27 suppresses RANKL expression in CD4+ T cells in part through STAT3. Immunol Lett 2011; 138:47-53. [PMID: 21453726 DOI: 10.1016/j.imlet.2011.02.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/14/2011] [Accepted: 02/24/2011] [Indexed: 01/06/2023]
Abstract
The receptor activator of NF-κB ligand (RANKL), which is expressed by not only osteoblasts but also activated T cells, plays an important role in bone-destructive diseases such as rheumatoid arthritis. IL-27, a member of the IL-6/IL-12 family cytokines, activates STAT1 and STAT3, promotes early helper T (Th)1 differentiation and generation of IL-10-producing type 1 regulatory T (Tr1) cells, and suppresses the production of inflammatory cytokines and inhibits Th2 differentiation. In addition, IL-27 was recently demonstrated to not only inhibit Th17 differentiation but also directly act on osteoclast precursor cells and suppress RANKL-mediated osteoclastogenesis through STAT1-dependent inhibition of c-Fos, leading to amelioration of the inflammatory bone destruction. In the present study, we investigated the effect of IL-27 on the expression of RANKL in CD4(+) T cells. We found that IL-27 greatly inhibits cell surface expression of RANKL on naive CD4(+) T cells activated by T cell receptor ligation and secretion of its soluble RANKL as well. The inhibitory effect was mediated in part by STAT3 but not by STAT1 or IL-10. In contrast, in differentiated Th17 cells, IL-27 much less efficiently inhibited the RANKL expression after restimulation. Taken together, these results indicate that IL-27 greatly inhibits primary RANKL expression in CD4(+) T cells, which could contribute to the suppressive effects of IL-27 on the inflammatory bone destruction.
Collapse
Affiliation(s)
- Sadahiro Kamiya
- Department of Clinical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Malchow S, Thaiss W, Jänner N, Waetzig GH, Gewiese-Rabsch J, Garbers C, Yamamoto K, Rose-John S, Scheller J. Essential role of neutrophil mobilization in concanavalin A-induced hepatitis is based on classic IL-6 signaling but not on IL-6 trans-signaling. Biochim Biophys Acta Mol Basis Dis 2011; 1812:290-301. [DOI: 10.1016/j.bbadis.2010.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 11/18/2010] [Accepted: 11/23/2010] [Indexed: 10/18/2022]
|
245
|
The pro- and anti-inflammatory properties of the cytokine interleukin-6. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:878-88. [PMID: 21296109 DOI: 10.1016/j.bbamcr.2011.01.034] [Citation(s) in RCA: 2277] [Impact Index Per Article: 162.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 01/23/2011] [Accepted: 01/27/2011] [Indexed: 02/06/2023]
Abstract
Interleukin-6 is a cytokine not only involved in inflammation and infection responses but also in the regulation of metabolic, regenerative, and neural processes. In classic signaling, interleukin-6 stimulates target cells via a membrane bound interleukin-6 receptor, which upon ligand binding associates with the signaling receptor protein gp130. Gp130 dimerizes, leading to the activation of Janus kinases and subsequent phosphorylation of tyrosine residues within the cytoplasmic portion of gp130. This leads to the engagement of phosphatase Src homology domains containing tyrosin phosphatase-2 (SHP-2) and activation of the ras/raf/Mitogen-activated protein (MAP) kinase (MAPK) pathway. In addition, signal transducer and activator of transcription factors are recruited, which are phosphorylated, and consequently dimerize whereupon they translocate into the nucleus and activate target genes. Interestingly, only few cells express membrane bound interleukin-6 receptor whereas all cells display gp130 on the cell surface. While cells, which only express gp130, are not responsive to interleukin-6 alone, they can respond to a complex of interleukin-6 bound to a naturally occurring soluble form of the interleukin-6 receptor. Therefore, the generation of soluble form of the interleukin-6 receptor dramatically enlarges the spectrum of interleukin-6 target cells. This process has been named trans-signaling. Here, we review the involvement of both signaling modes in the biology of interleukin-6. It turns out that regenerative or anti-inflammatory activities of interleukin-6 are mediated by classic signaling whereas pro-inflammatory responses of interleukin-6 are rather mediated by trans-signaling. This is important since therapeutic blockade of interleukin-6 by the neutralizing anti-interleukin-6 receptor monoclonal antibody tocilizumab has recently been approved for the treatment of inflammatory diseases. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.
Collapse
|
246
|
Yoshimoto T, Morishima N, Okumura M, Chiba Y, Xu M, Mizuguchi J. Interleukins and cancer immunotherapy. Immunotherapy 2011; 1:825-44. [PMID: 20636026 DOI: 10.2217/imt.09.46] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cancer is a complex disease with interactions between normal and neoplastic cells. Since current therapies for cancer largely rely on drugs or radiation that kill dividing cells or block cell division, these treatments may have severe side effects on normal proliferating cells in patients with cancer. Therefore, the potential for treatment of cancer patients by immunologic approaches, which may be specific for tumors and will not injure most normal cells, has great promise. Cancer immunotherapy aims to augment the weak host immune response to developing tumors. One strategy is to utilize cytokines such as IL-2. More recently, several exciting new interleukins have been characterized that have considerable promise for future immunotherapy. The promise of cancer immunotherapy largely depends upon the identification of these novel interleukins. This review provides an overview of the antitumor effects of relatively new interleukins as potential therapeutic agents applicable for cancer immunotherapy.
Collapse
Affiliation(s)
- Takayuki Yoshimoto
- Intractable Disease Research Center, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan.
| | | | | | | | | | | |
Collapse
|
247
|
Regulation of T cell activation by TLR ligands. Eur J Cell Biol 2011; 90:582-92. [PMID: 21292344 DOI: 10.1016/j.ejcb.2010.11.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/18/2010] [Accepted: 11/23/2010] [Indexed: 11/23/2022] Open
Abstract
Regulatory T cells (Treg) maintain peripheral tolerance and play a critical role in the control of the immune response in infection, tumor defense, organ transplantation and allergy. CD4(+)CD25(high) Treg suppress the proliferation and cytokine production of CD4(+)CD25(-) responder T cells. The suppression requires cell-cell-contact and/or production of inhibitory cytokines like IL-10 or TGF-β. The current knowledge about the regulation of Treg suppressive function is limited. Toll-like receptors (TLR) are widely expressed in the innate immune system. They recognize conserved microbial ligands such as lipopolysaccharide, bacterial lipopeptides or viral and bacterial RNA and DNA. TLR play an essential role in innate immune responses and in the initiation of adaptive immune responses. However, certain TLR are also expressed in T lymphocytes, and the respective ligands can directly modulate T cell function. TLR2, TLR3, TLR5 and TLR9 act as costimulatory receptors to enhance proliferation and/or cytokine production of T-cell receptor-stimulated T lymphocytes. In addition, TLR2, TLR5 and TLR8 modulate the suppressive activity of naturally occurring CD4(+)CD25(high) Treg. The direct responsiveness of T lymphocytes to TLR ligands offers new perspectives for the immunotherapeutic manipulation of T cell responses. In this article we will discuss the regulation of Treg and other T cell subsets by TLR ligands.
Collapse
|
248
|
Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S. Interleukin-6, a mental cytokine. ACTA ACUST UNITED AC 2011; 67:157-83. [PMID: 21238488 DOI: 10.1016/j.brainresrev.2011.01.002] [Citation(s) in RCA: 279] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/21/2010] [Accepted: 01/08/2011] [Indexed: 12/18/2022]
Abstract
Almost a quarter of a century ago, interleukin-6 (IL-6) was discovered as an inflammatory cytokine involved in B cell differentiation. Today, IL-6 is recognized to be a highly versatile cytokine, with pleiotropic actions not only in immune cells, but also in other cell types, such as cells of the central nervous system (CNS). The first evidence implicating IL-6 in brain-related processes originated from its dysregulated expression in several neurological disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. In addition, IL-6 was shown to be involved in multiple physiological CNS processes such as neuron homeostasis, astrogliogenesis and neuronal differentiation. The molecular mechanisms underlying IL-6 functions in the brain have only recently started to emerge. In this review, an overview of the latest discoveries concerning the actions of IL-6 in the nervous system is provided. The central position of IL-6 in the neuroinflammatory reaction pattern, and more specifically, the role of IL-6 in specific neurodegenerative processes, which accompany Alzheimer's disease, multiple sclerosis and excitotoxicity, are discussed. It is evident that IL-6 has a dichotomic action in the CNS, displaying neurotrophic properties on the one hand, and detrimental actions on the other. This is in agreement with its central role in neuroinflammation, which evolved as a beneficial process, aimed at maintaining tissue homeostasis, but which can become malignant when exaggerated. In this perspective, it is not surprising that 'well-meant' actions of IL-6 are often causing harm instead of leading to recovery.
Collapse
Affiliation(s)
- Anneleen Spooren
- Laboratory of Eukaryotic Signal Transduction and Gene Expression, University of Ghent, K.L. Ledeganckstraat 35, 9000 Gent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
249
|
Lupardus PJ, Skiniotis G, Rice AJ, Thomas C, Fischer S, Walz T, Garcia KC. Structural snapshots of full-length Jak1, a transmembrane gp130/IL-6/IL-6Rα cytokine receptor complex, and the receptor-Jak1 holocomplex. Structure 2011; 19:45-55. [PMID: 21220115 PMCID: PMC3052743 DOI: 10.1016/j.str.2010.10.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/25/2010] [Accepted: 10/31/2010] [Indexed: 11/28/2022]
Abstract
The shared cytokine receptor gp130 signals as a homodimer or heterodimer through activation of Janus kinases (Jaks) associated with the receptor intracellular domains. Here, we reconstitute, in parts and whole, the full-length gp130 homodimer in complex with the cytokine interleukin-6 (IL-6), its alpha receptor (IL-6Rα) and Jak1, for electron microscopy imaging. We find that the full-length gp130 homodimer complex has intimate interactions between the trans- and juxtamembrane segments of the two receptors, appearing to form a continuous connection between the extra- and intracellular regions. 2D averages and 3D reconstructions of full-length Jak1 reveal a three lobed structure comprising FERM-SH2, pseudokinase, and kinase modules possessing extensive intersegmental flexibility that likely facilitates allosteric activation. Single-particle imaging of the gp130/IL-6/IL-6Rα/Jak1 holocomplex shows Jak1 associated with the membrane proximal intracellular regions of gp130, abutting the would-be inner leaflet of the cell membrane. Jak1 association with gp130 is enhanced by the presence of a membrane environment.
Collapse
Affiliation(s)
- Patrick J Lupardus
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | |
Collapse
|
250
|
Oh J, McCloskey MA, Blong CC, Bendickson L, Nilsen-Hamilton M, Sakaguchi DS. Astrocyte-derived interleukin-6 promotes specific neuronal differentiation of neural progenitor cells from adult hippocampus. J Neurosci Res 2010; 88:2798-809. [PMID: 20568291 DOI: 10.1002/jnr.22447] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to investigate the ability of astrocyte-derived factors to influence neural progenitor cell differentiation. We previously demonstrated that rat adult hippocampal progenitor cells (AHPCs) immunoreactive for the neuronal marker class III beta-tubulin (TUJ1) were significantly increased in the presence of astrocyte-derived soluble factors under noncontact coculture conditions. Using whole-cell patch-clamp analysis, we observed that the cocultured AHPCs displayed two prominent voltage-gated conductances, tetraethyl ammonium (TEA)-sensitive outward currents and fast transient inward currents. The outward and inward current densities of the cocultured AHPCs were approximately 2.5-fold and 1.7-fold greater, respectively, than those of cells cultured alone. These results suggest that astrocyte-derived soluble factors induce neuronal commitment of AHPCs. To investigate further the activity of a candidate neurogenic factor on AHPC differentiation, we cultured AHPCs in the presence or absence of purified rat recombinant interleukin-6 (IL-6). We also confirmed that the astrocytes used in this study produced IL-6 by ELISA and RT-qPCR. When AHPCs were cultured with IL-6 for 6-7 days, the TUJ1-immunoreactive AHPCs and the average length of TUJ1-immunoreactive neurites were significantly increased compared with the cells cultured without IL-6. Moreover, IL-6 increased the inward current density to an extent comparable to that of coculture with astrocytes, with no significant differences in the outward current density, apparent resting potential, or cell capacitance. These results suggest that astrocyte-derived IL-6 may facilitate AHPC neuronal differentiation. Our findings have important implications for understanding injury-induced neurogenesis and developing cell-based therapeutic strategies using neural progenitors.
Collapse
Affiliation(s)
- Jisun Oh
- Neuroscience Program, Iowa State University, Ames, Iowa 50011, USA
| | | | | | | | | | | |
Collapse
|