1
|
Yu X, Duan R, Jiang L, Wang T, Li Z, Zhang B, Su W, Lin Y. Interleukin-6 in non-infectious uveitis: Biology, experimentalevidence and treatment strategies. Biochem Pharmacol 2024; 230:116605. [PMID: 39491564 DOI: 10.1016/j.bcp.2024.116605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/10/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Uveitis is the leading cause of visual impairment worldwide. Interleukin-6 (IL-6), which is upregulated in response to inflammation, is one of the most important inflammatory cytokines associated with uveitis. Two major IL-6 receptors (IL-6R) mediate the pro-inflammatory and anti-inflammatory biological effects of IL-6. This review summarized multiple perspectives on the mechanism of IL-6-mediated uveitis, based on experimental evidence from clinical and animal models. It includes discussions on the roles of the downstream IL-6 signaling pathway, immunocytes, and the blood-retinal barrier. Therapeutic strategies aimed at blocking the action of IL-6 have progressed in clinical practice. However, due to the adverse events associated with existing biologics including infections, drugs that selectively inhibit intraocular IL-6 still require further development. The novel concept of converting the pro-inflammatory effects of IL-6 into protective effects also requires further research. In addition, the relationship between the trans-presentation of IL-6R and T-helper17 cells in uveitis remains unexplored. This review aims to consolidate our current understanding of the biology, signaling pathways, experimental models, and immune pathogenesis related to IL-6 and uveitis. We also discuss clinical strategies focused on blocking IL-6 as a treatment for uveitis. Targeting IL-6 provides unlimited potential for improving the diagnosis, treatment, and prognosis of uveitis.
Collapse
Affiliation(s)
- Xiaoyang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510060, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Loujing Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Tianfu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Bowen Zhang
- Sun Yat-sen University Zhongshan School of Medicine, Guangzhou 510060, China
| | - Wenru Su
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200000, China.
| | - Ying Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
2
|
Miri N, Köhler N, Dittrich A. Quantification of membrane-bound cytokine receptors by calibrated flow cytometry. STAR Protoc 2023; 4:102511. [PMID: 37581983 PMCID: PMC10457439 DOI: 10.1016/j.xpro.2023.102511] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 08/17/2023] Open
Abstract
We present a protocol for quantifying the expression of the receptor gp130 using a calibrated flow cytometric approach. We describe pitfalls for receptor quantification such as titration of primary antibodies and standardizing cell culture. Receptors are stained with primary antibodies and fluorophore-coupled secondary antibodies. Beads covered with defined numbers of immunoglobulin G stained with fluorophore-coupled secondary antibodies serve as calibrators. In this way, the fluorescence intensity of cells is converted to the number of receptors on the cell surface. For complete details on the use and execution of this protocol, please refer to Reeh et al. (2019).1.
Collapse
Affiliation(s)
- Niloufarsadat Miri
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Nadine Köhler
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany; Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, 39106 Magdeburg, Germany; Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, 39106 Magdeburg, Germany; Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39106 Magdeburg, Germany.
| |
Collapse
|
3
|
Köhler N, Miri N, Dittrich A. Quantification of total and phosphorylated STAT3 by calibrated western blotting. STAR Protoc 2023; 4:102508. [PMID: 37669163 PMCID: PMC10485629 DOI: 10.1016/j.xpro.2023.102508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/23/2023] [Accepted: 07/25/2023] [Indexed: 09/07/2023] Open
Abstract
Quantification of intracellular proteins is essential to understand signaling. Here, we describe quantification of the expression and phosphorylation of the transcription factor STAT3. We present isolation of total and phosphorylated STAT3 from cell lysates by immunoprecipitation, followed by SDS-PAGE and western blot together with known amounts of a calibrator protein that shares an epitope with the precipitated proteins. Finally, we explain how to relate the amount of precipitated protein to the amount of calibrator protein considering the efficiency of immunoprecipitation. For complete details on the use and execution of this protocol, please refer to Dittrich et al. (2012)1 and Reeh et al. (2019).2.
Collapse
Affiliation(s)
- Nadine Köhler
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Niloufarsadat Miri
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, 39106 Magdeburg, Germany; Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, 39106 Magdeburg, Germany; Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, 39106 Magdeburg, Germany; Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke University, 39106 Magdeburg, Germany.
| |
Collapse
|
4
|
The Interleukin-1 (IL-1) Superfamily Cytokines and Their Single Nucleotide Polymorphisms (SNPs). J Immunol Res 2022; 2022:2054431. [PMID: 35378905 PMCID: PMC8976653 DOI: 10.1155/2022/2054431] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 12/19/2022] Open
Abstract
Interleukins (ILs)—which are important members of cytokines—consist of a vast group of molecules, including a wide range of immune mediators that contribute to the immunological responses of many cells and tissues. ILs are immune-glycoproteins, which directly contribute to the growth, activation, adhesion, differentiation, migration, proliferation, and maturation of immune cells; and subsequently, they are involved in the pro and anti-inflammatory responses of the body, by their interaction with a wide range of receptors. Due to the importance of immune system in different organisms, the genes belonging to immune elements, such as ILs, have been studied vigorously. The results of recent investigations showed that the genes pertaining to the immune system undergo progressive evolution with a constant rate. The occurrence of any mutation or polymorphism in IL genes may result in substantial changes in their biology and function and may be associated with a wide range of diseases and disorders. Among these abnormalities, single nucleotide polymorphisms (SNPs) can represent as important disruptive factors. The present review aims at concisely summarizing the current knowledge available on the occurrence, properties, role, and biological consequences of SNPs within the IL-1 family members.
Collapse
|
5
|
Soni B, Singh S. Synthetic Perturbations in IL6 Biological Circuit Induces Dynamical Cellular Response. Molecules 2021; 27:molecules27010124. [PMID: 35011356 PMCID: PMC8746995 DOI: 10.3390/molecules27010124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022] Open
Abstract
Macrophage phenotype plays a crucial role in the pathogenesis of Leishmanial infection. Pro-inflammatory cytokines signals through the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway that functions in parasite killing. Suppression of cytokine signaling (SOCS) is a well-known negative feedback regulator of the JAK/STAT pathway. However, change in the expression levels of SOCSs in correlation with the establishment of infection is not well understood. IL6 is a pleotropic cytokine that induces SOCS1 and SOCS3 expression through JAK-STAT signaling. Mathematical modeling of the TLR2 and IL6 signaling pathway has established the immune axis of SOCS1 and SOCS3 functioning in macrophage polarization during the early stage of Leishmania major infection. The ratio has been quantified both in silico and in vitro as 3:2 which is required to establish infection during the early stage. Furthermore, phosphorylated STAT1 and STAT3 have been established as an immunological cross talk between TLR2 and IL6 signaling pathways. Using synthetic biology approaches, peptide based immuno-regulatory circuits have been designed to target the activity of SOCS1 which can restore pro-inflammatory cytokine expression during infection. In a nutshell, we explored the potential of synthetic biology to address and rewire the immune response from Th2 to Th1 type during the early stage of leishmanial infection governed by SOCS1/SOCS3 immune axis.
Collapse
Affiliation(s)
| | - Shailza Singh
- Correspondence: or ; Tel.: +91-20-2570-8296 (ext. 95); Fax: +91-20-2569-2259
| |
Collapse
|
6
|
Fiebelkow J, Guendel A, Guendel B, Mehwald N, Jetka T, Komorowski M, Waldherr S, Schaper F, Dittrich A. The tyrosine phosphatase SHP2 increases robustness and information transfer within IL-6-induced JAK/STAT signalling. Cell Commun Signal 2021; 19:94. [PMID: 34530865 PMCID: PMC8444181 DOI: 10.1186/s12964-021-00770-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/28/2021] [Indexed: 11/21/2022] Open
Abstract
Background Cell-to-cell heterogeneity is an inherent feature of multicellular organisms and is central in all physiological and pathophysiological processes including cellular signal transduction. The cytokine IL-6 is an essential mediator of pro- and anti-inflammatory processes. Dysregulated IL-6-induced intracellular JAK/STAT signalling is associated with severe inflammatory and proliferative diseases. Under physiological conditions JAK/STAT signalling is rigorously controlled and timely orchestrated by regulatory mechanisms such as expression of the feedback-inhibitor SOCS3 and activation of the protein-tyrosine phosphatase SHP2 (PTPN11). Interestingly, the function of negative regulators seems not to be restricted to controlling the strength and timely orchestration of IL-6-induced STAT3 activation. Exemplarily, SOCS3 increases robustness of late IL-6-induced STAT3 activation against heterogenous STAT3 expression and reduces the amount of information transferred through JAK/STAT signalling. Methods Here we use multiplexed single-cell analyses and information theoretic approaches to clarify whether also SHP2 contributes to robustness of STAT3 activation and whether SHP2 affects the amount of information transferred through IL-6-induced JAK/STAT signalling. Results SHP2 increases robustness of both basal, cytokine-independent STAT3 activation and early IL-6-induced STAT3 activation against differential STAT3 expression. However, SHP2 does not affect robustness of late IL-6-induced STAT3 activation. In contrast to SOCS3, SHP2 increases the amount of information transferred through IL-6-induced JAK/STAT signalling, probably by reducing cytokine-independent STAT3 activation and thereby increasing sensitivity of the cells. These effects are independent of SHP2-dependent MAPK activation. Conclusion In summary, the results of this study extend our knowledge of the functions of SHP2 in IL-6-induced JAK/STAT signalling. SHP2 is not only a repressor of basal and cytokine-induced STAT3 activity, but also ensures robustness and transmission of information.![]() Plain English summary Cells within a multicellular organism communicate with each other to exchange information about the environment. Communication between cells is facilitated by soluble molecules that transmit information from one cell to the other. Cytokines such as interleukin-6 are important soluble mediators that are secreted when an organism is faced with infections or inflammation. Secreted cytokines bind to receptors within the membrane of their target cells. This binding induces activation of an intracellular cascade of reactions called signal transduction, which leads to cellular responses. An important example of intracellular signal transduction is JAK/STAT signalling. In healthy organisms signalling is controlled and timed by regulatory mechanisms, whose activation results in a controlled shutdown of signalling pathways. Interestingly, not all cells within an organism are identical. They differ in the amount of proteins involved in signal transduction, such as STAT3. These differences shape cellular communication and responses to intracellular signalling. Here, we show that an important negative regulatory protein called SHP2 (or PTPN11) is not only responsible for shutting down signalling, but also for steering signalling in heterogeneous cell populations. SHP2 increases robustness of STAT3 activation against variable STAT3 amounts in individual cells. Additionally, it increases the amount of information transferred through JAK/STAT signalling by increasing the dynamic range of pathway activation in heterogeneous cell populations. This is an amazing new function of negative regulatory proteins that contributes to communication in heterogeneous multicellular organisms in health and disease. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00770-7.
Collapse
Affiliation(s)
- Jessica Fiebelkow
- Institute of Biology, Department of Systems Biology, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - André Guendel
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Beate Guendel
- Institute of Biology, Department of Systems Biology, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany.,Karolinska Institutet, Clintec, Huddinge, Sweden
| | - Nora Mehwald
- Institute of Biology, Department of Systems Biology, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
| | - Tomasz Jetka
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong, Hong Kong
| | - Michal Komorowski
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warszawa, Poland
| | | | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany.,Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, Magdeburg, Germany.,Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, Magdeburg, Germany
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany. .,Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, Magdeburg, Germany. .,Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
7
|
Schmidt-Arras D, Rose-John S. Endosomes as Signaling Platforms for IL-6 Family Cytokine Receptors. Front Cell Dev Biol 2021; 9:688314. [PMID: 34141712 PMCID: PMC8204807 DOI: 10.3389/fcell.2021.688314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin-6 (IL-6) is the name-giving cytokine of a family of eleven members, including IL-6, CNTF, LIF, and IL-27. IL-6 was first recognized as a B-cell stimulating factor but we now know that the cytokine plays a pivotal role in the orchestration of inflammatory processes as well as in inflammation associated cancer. Moreover, IL-6 is involved in metabolic regulation and it has been shown to be involved in major neural activities such as neuroprotection, which can help to repair and to reduce brain damage. Receptor complexes of all members formed at the plasma membrane contain one or two molecules of the signaling receptor subunit GP130 and the mechanisms of signal transduction are well understood. IL-6 type cytokines can also signal from endomembranes, in particular the endosome, and situations have been reported in which endocytosis of receptor complexes are a prerequisite of intracellular signaling. Moreover, pathogenic GP130 variants were shown to interfere with spatial activation of downstream signals. We here summarize the molecular mechanisms underlying spatial regulation of IL-6 family cytokine signaling and discuss its relevance for pathogenic processes.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
8
|
Soni B, Singh S. COVID-19 co-infection mathematical model as guided through signaling structural framework. Comput Struct Biotechnol J 2021; 19:1672-1683. [PMID: 33815692 PMCID: PMC7997053 DOI: 10.1016/j.csbj.2021.03.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 and Influenza co-infection turned out to be a huge threat in recent times. The clinical presentation and disease severity is common in both the infection condition. The present paper deals with studying co-infection model system through systems biology approaches. Understanding signaling regulation in COVID-19 and co-infection model systems aid in the development of network-based models thereby suggesting intervention points for therapeutics. This paper highlights the aim of revealing such perturbations to decipher opportune mediating cross talks characterizing the deadly viral disease. The comparative analysis of both the models reveals major signaling protein NFκB and STAT1 playing a crucial role in establishing co-infection. By targeting these proteins at cellular level, it might help modulating the release of potent pro-inflammatory cytokines thereby taming the severity of the disease symptoms. Mathematical models developed here are precisely tailored and serves as a first step towards co-infection model offering flexibility and pitching towards therapeutic investigation.
Collapse
Affiliation(s)
- Bhavnita Soni
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SPPU Campus, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SPPU Campus, Pune 411007, India
| |
Collapse
|
9
|
Zhang J, Zhang Y, Qu B, Yang H, Hu S, Dong X. If small molecules immunotherapy comes, can the prime be far behind? Eur J Med Chem 2021; 218:113356. [PMID: 33773287 DOI: 10.1016/j.ejmech.2021.113356] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/15/2021] [Accepted: 02/28/2021] [Indexed: 02/07/2023]
Abstract
Anti-cancer immunotherapy, which includes cellular immunotherapy, immune checkpoint inhibitors and cancer vaccines, has transformed the treatment strategies of several malignancies in the past decades. Immune checkpoints blockade (ICB) is the most commonly tested therapy and has the potential to induce a durable immune response in different types of cancers. However, all approved immune checkpoint inhibitors (ICIs) are monoclonal antibodies (mAbs), which are fraught with disadvantages including lack of oral bioavailability, prolonged tissue retention and poor membrane permeability. Therefore, the research focus has shifted to developing small molecule inhibitors to obviate the limitations of mAbs. Given the complexity of the tumor micro-environment (TME), the combination of ICIs with various small molecule agonists/inhibitors are currently being tested in clinical trials to improve treatment outcomes and prevent tumor recurrence. In this review, we have summarized the mechanisms and therapeutic potential of several molecular targets, along with the current status of small molecule inhibitors.
Collapse
Affiliation(s)
- Jingyu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu Zhang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Bingxue Qu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Haiyan Yang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), PR China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, PR China
| | - Shengquan Hu
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310058, PR China; Cancer Center of Zhejiang University, Hangzhou, 310058, PR China.
| |
Collapse
|
10
|
Affiliation(s)
- Vladimir Jurisic
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
11
|
Gibbs KD, Washington EJ, Jaslow SL, Bourgeois JS, Foster MW, Guo R, Brennan RG, Ko DC. The Salmonella Secreted Effector SarA/SteE Mimics Cytokine Receptor Signaling to Activate STAT3. Cell Host Microbe 2019; 27:129-139.e4. [PMID: 31901521 DOI: 10.1016/j.chom.2019.11.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/05/2019] [Accepted: 11/21/2019] [Indexed: 01/18/2023]
Abstract
Bacteria masterfully co-opt and subvert host signal transduction. As a paradigmatic example, Salmonella uses two type-3 secretion systems to inject effector proteins that facilitate Salmonella entry, establishment of an intracellular niche, and modulation of immune responses. We previously demonstrated that the Salmonella anti-inflammatory response activator SarA (Stm2585, GogC, PagJ, SteE) activates the host transcription factor STAT3 to drive expression of immunomodulatory STAT3-targets. Here, we demonstrate-by sequence, function, and biochemical measurement-that SarA mimics the cytoplasmic domain of glycoprotein 130 (gp130, IL6ST). SarA is phosphorylated at a YxxQ motif, facilitating binding to STAT3 with greater affinity than gp130. Departing from canonical gp130 signaling, SarA function is JAK-independent but requires GSK-3, a key regulator of metabolism and development. Our results reveal that SarA undergoes host phosphorylation to recruit a STAT3-activating complex, circumventing cytokine receptor activation. Effector mimicry of gp130 suggests GSK-3 can regulate normal cytokine signaling, potentially enabling metabolic and immune crosstalk.
Collapse
Affiliation(s)
- Kyle D Gibbs
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Erica J Washington
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Sarah L Jaslow
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jeffrey S Bourgeois
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; Duke University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Matthew W Foster
- Duke Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, NC 27710, USA
| | - Robyn Guo
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Richard G Brennan
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; Duke University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
12
|
Jak-Stat Signaling Induced by Interleukin-6 Family Cytokines in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11111704. [PMID: 31683891 PMCID: PMC6896168 DOI: 10.3390/cancers11111704] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide. It can be caused by chronic liver cell injury with resulting sustained inflammation, e.g., triggered by infections with hepatitis viruses B (HBV) and C (HCV). Death of hepatocytes leads to the activation of compensatory mechanisms, which can ultimately result in liver fibrosis and cirrhosis. Another common feature is the infiltration of the liver with inflammatory cells, which secrete cytokines and chemokines that act directly on the hepatocytes. Among several secreted proteins, members of the interleukin-6 (IL-6) family of cytokines have emerged as important regulatory proteins that might constitute an attractive target for therapeutic intervention. The IL-6-type cytokines activate multiple intracellular signaling pathways, and especially the Jak/STAT cascade has been shown to be crucial for HCC development. In this review, we give an overview about HCC pathogenesis with respect to IL-6-type cytokines and the Jak/STAT pathway. We highlight the role of mutations in genes encoding several proteins involved in the cytokine/Jak/STAT axis and summarize current knowledge about IL-6 family cytokines in this context. We further discuss possible anti-cytokine therapies for HCC patients in comparison to already established therapies.
Collapse
|
13
|
Reeh H, Rudolph N, Billing U, Christen H, Streif S, Bullinger E, Schliemann-Bullinger M, Findeisen R, Schaper F, Huber HJ, Dittrich A. Response to IL-6 trans- and IL-6 classic signalling is determined by the ratio of the IL-6 receptor α to gp130 expression: fusing experimental insights and dynamic modelling. Cell Commun Signal 2019; 17:46. [PMID: 31101051 PMCID: PMC6525395 DOI: 10.1186/s12964-019-0356-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Interleukin-6 is a pleiotropic cytokine with high clinical relevance and an important mediator of cellular communication, orchestrating both pro- and anti-inflammatory processes. Interleukin-6-induced signalling is initiated by binding of IL-6 to the IL-6 receptor α and subsequent binding to the signal transducing receptor subunit gp130. This active receptor complex initiates signalling through the Janus kinase/signal transducer and activator of transcription pathway. Of note, IL-6 receptor α exists in a soluble and a transmembrane form. Binding of IL-6 to membrane-bound IL-6 receptor α induces anti-inflammatory classic signalling, whereas binding of IL-6 to soluble IL-6 receptor α induces pro-inflammatory trans-signalling. Trans-signalling has been described to be markedly stronger than classic signalling. Understanding the molecular mechanisms that drive differences between trans- and classic signalling is important for the design of trans-signalling-specific therapies. These differences will be addressed here using a combination of dynamic mathematical modelling and molecular biology. METHODS We apply an iterative systems biology approach using set-based modelling and validation approaches combined with quantitative biochemical and cell biological analyses. RESULTS The combination of experimental analyses and dynamic modelling allows to relate the observed differences between IL-6-induced trans- and classic signalling to cell-type specific differences in the expression and ratios of the individual subunits of the IL-6 receptor complex. Canonical intracellular Jak/STAT signalling is indifferent in IL-6-induced trans- and classic signalling. CONCLUSION This study contributes to the understanding of molecular mechanisms of IL-6 signal transduction and underlines the power of combined dynamical modelling, model-based validation and biological experiments. The opposing pro- and anti-inflammatory responses initiated by IL-6 trans- and classic signalling depend solely on the expression ratios of the subunits of the entire receptor complex. By pointing out the importance of the receptor expression ratio for the strength of IL-6 signalling this study lays a foundation for future precision medicine approaches that aim to selectively block pro-inflammatory trans-signalling. Furthermore, the derived models can be used for future therapy design.
Collapse
Affiliation(s)
- Heike Reeh
- Department of Systems Biology, Institute of Biology, Faculty of Natural Sciences, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Nadine Rudolph
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Ulrike Billing
- Department of Systems Biology, Institute of Biology, Faculty of Natural Sciences, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Henrike Christen
- Department of Systems Biology, Institute of Biology, Faculty of Natural Sciences, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Stefan Streif
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany.,Automatic Control and System Dynamics Laboratory, Institute of Automation, Chemnitz University of Technology, Reichenhainer Straße 70, 09107, Chemnitz, Germany
| | - Eric Bullinger
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Monica Schliemann-Bullinger
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Rolf Findeisen
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Fred Schaper
- Department of Systems Biology, Institute of Biology, Faculty of Natural Sciences, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Heinrich J Huber
- Department of Systems Theory and Automatic Control, Institute for Automation Engineering, Faculty of Electrical Engineering and Information Technology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany.,Comuptational Biology, Discovery Research, Boehringer Ingelheim Pharma, Birkendorfer Straße 65, 88400, Biberach, Germany
| | - Anna Dittrich
- Department of Systems Biology, Institute of Biology, Faculty of Natural Sciences, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany.
| |
Collapse
|
14
|
Konjević GM, Vuletić AM, Mirjačić Martinović KM, Larsen AK, Jurišić VB. The role of cytokines in the regulation of NK cells in the tumor environment. Cytokine 2019; 117:30-40. [PMID: 30784898 DOI: 10.1016/j.cyto.2019.02.001] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/29/2019] [Accepted: 02/07/2019] [Indexed: 12/19/2022]
Abstract
Natural killer (NK) cells are innate lymphoid cells that are important effectors in the first line of defense toward transformed cells. This is mediated both by direct cytotoxic mechanisms and by production of immunoregulatory cytokines. Recent evidence has shown that NK cells also display memory, similar to the cells of the adaptive immune system. Cytokines are pivotal for the maturation, activation and survival of NK cells. Interleukins (IL)-2, IL-12, IL-15, IL-18, IL-21 and type I interferons positively regulate NK cell function, either independently or in cooperation, whereas other cytokines, such as IL-23 and IL-27, may enhance or suppress NK cell function depending on the context. In the tumor microenvironment, TGFβ, IL-10 and IL-6 suppress NK cell activity not only directly, but also indirectly, by affecting immunosuppressive cells and by antagonizing the effect of stimulatory cytokines, thereby dampening the antitumor response of NK cells and promoting subsequent tumor evasion and progression. Increased understanding of the NK cell response to cytokines has provided a better understanding of their impaired function in tumors which may aid in the development of novel immunotherapeutic strategies to enhance NK cell responses in cancer patients.
Collapse
Affiliation(s)
- Gordana M Konjević
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, Dr Subotića 8, 11000 Beograd, Serbia
| | - Ana M Vuletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Katarina M Mirjačić Martinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Annette K Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine, INSERM U938 and Sorbonne University, Kourilsky Building 1st Floor, Hôpital Saint-Antoine, 184 rue du Faubourg Saint Antoine, 75571 PARIS Cédex 12 France
| | - Vladimir B Jurišić
- Faculty of Medical Sciences, University of Kragujevac, P.BOX 124, 34000 Kragujevac, Serbia.
| |
Collapse
|
15
|
Billing U, Jetka T, Nortmann L, Wundrack N, Komorowski M, Waldherr S, Schaper F, Dittrich A. Robustness and Information Transfer within IL-6-induced JAK/STAT Signalling. Commun Biol 2019; 2:27. [PMID: 30675525 PMCID: PMC6338669 DOI: 10.1038/s42003-018-0259-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023] Open
Abstract
Cellular communication via intracellular signalling pathways is crucial. Expression and activation of signalling proteins is heterogenous between isogenic cells of the same cell-type. However, mechanisms evolved to enable sufficient communication and to ensure cellular functions. We use information theory to clarify mechanisms facilitating IL-6-induced JAK/STAT signalling despite cell-to-cell variability. We show that different mechanisms enabling robustness against variability complement each other. Early STAT3 activation is robust as long as cytokine concentrations are low. Robustness at high cytokine concentrations is ensured by high STAT3 expression or serine phosphorylation. Later the feedback-inhibitor SOCS3 increases robustness. Channel Capacity of JAK/STAT signalling is limited by cell-to-cell variability in STAT3 expression and is affected by the same mechanisms governing robustness. Increasing STAT3 amount increases Channel Capacity and robustness, whereas increasing STAT3 tyrosine phosphorylation reduces robustness but increases Channel Capacity. In summary, we elucidate mechanisms preventing dysregulated signalling by enabling reliable JAK/STAT signalling despite cell-to-cell heterogeneity.
Collapse
Affiliation(s)
- Ulrike Billing
- Otto-von-Guericke University Magdeburg, Institute of Biology, Department of Systems Biology, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Tomasz Jetka
- Polish Academy of Sciences, Institute of Fundamental Technological Research, Division of Modelling in Biology and Medicine, Pawinskiego 5B, 02- 106, Warszawa, Poland
| | - Lukas Nortmann
- Otto-von-Guericke University Magdeburg, Institute of Biology, Department of Systems Biology, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Nicole Wundrack
- Otto-von-Guericke University Magdeburg, Institute of Biology, Department of Systems Biology, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Michal Komorowski
- Polish Academy of Sciences, Institute of Fundamental Technological Research, Division of Modelling in Biology and Medicine, Pawinskiego 5B, 02- 106, Warszawa, Poland
| | - Steffen Waldherr
- KU Leuven, Department of Chemical Engineering, Celestijnenlaan 200f - box 2424, 3001 Leuven, Belgium
| | - Fred Schaper
- Otto-von-Guericke University Magdeburg, Institute of Biology, Department of Systems Biology, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Anna Dittrich
- Otto-von-Guericke University Magdeburg, Institute of Biology, Department of Systems Biology, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
16
|
Lokau J, Garbers C. Activating mutations of the gp130/JAK/STAT pathway in human diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 116:283-309. [PMID: 31036294 DOI: 10.1016/bs.apcsb.2018.11.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cytokines of the interleukin-6 (IL-6) family are involved in numerous physiological and pathophysiological processes. Dysregulated and increased activities of its members can be found in practically all human inflammatory diseases including cancer. All cytokines activate several intracellular signaling cascades, including the Jak/STAT, MAPK, PI3K, and Src/YAP signaling pathways. Additionally, several mutations in proteins involved in these signaling cascades have been identified in human patients, which render these proteins constitutively active and result in a hyperactivation of the signaling pathway. Interestingly, some of these mutations are associated with or even causative for distinct human diseases, making them interesting targets for therapy. This chapter describes the basic biology of the gp130/Jak/STAT pathway, summarizes what is known about the molecular mechanisms of the activating mutations, and gives an outlook how this knowledge can be exploited for targeted therapy in human diseases.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
17
|
Wu D, Wen X, Wang Y, Han X, Wang S, Shen M, Fan S, Zhuang J, Zhang Z, Shan Q, Li M, Hu B, Sun C, Lu J, Chen G, Zheng Y. Retracted
: Effect of microRNA‐186 on oxidative stress injury of neuron by targeting interleukin 2 through the janus kinase‐signal transducer and activator of transcription pathway in a rat model of Alzheimer’s disease. J Cell Physiol 2018; 233:9488-9502. [DOI: 10.1002/jcp.26843] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/10/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Dong‐Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Yong‐Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Xin‐Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Shao‐Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- School of Environment Science and Spatial Informatics China University of Mining and Technology Xuzhou China
- Jiangsu Key Laboratory for Eco‐Agricultural Biotechnology Around Hongze Lake, School of Life Sciences Huaiyin Normal University Huaian China
| | - Zi‐Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Meng‐Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Chun‐Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| | - Gui‐Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center Nanjing University Nanjing China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center Nanjing University Nanjing China
| | - Yuan‐Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science Jiangsu Normal University Xuzhou China
- College of Health Sciences Jiangsu Normal University Xuzhou Jiangsu China
| |
Collapse
|
18
|
Haeseler FV, Rudolph N, Findeisen R, Huber HJ. Parameter Estimation for Signal Transduction Networks from Experimental Time Series Using Picard Iteration. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.ifacol.2018.09.298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
19
|
Sobotta S, Raue A, Huang X, Vanlier J, Jünger A, Bohl S, Albrecht U, Hahnel MJ, Wolf S, Mueller NS, D'Alessandro LA, Mueller-Bohl S, Boehm ME, Lucarelli P, Bonefas S, Damm G, Seehofer D, Lehmann WD, Rose-John S, van der Hoeven F, Gretz N, Theis FJ, Ehlting C, Bode JG, Timmer J, Schilling M, Klingmüller U. Model Based Targeting of IL-6-Induced Inflammatory Responses in Cultured Primary Hepatocytes to Improve Application of the JAK Inhibitor Ruxolitinib. Front Physiol 2017; 8:775. [PMID: 29062282 PMCID: PMC5640784 DOI: 10.3389/fphys.2017.00775] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/22/2017] [Indexed: 12/12/2022] Open
Abstract
IL-6 is a central mediator of the immediate induction of hepatic acute phase proteins (APP) in the liver during infection and after injury, but increased IL-6 activity has been associated with multiple pathological conditions. In hepatocytes, IL-6 activates JAK1-STAT3 signaling that induces the negative feedback regulator SOCS3 and expression of APPs. While different inhibitors of IL-6-induced JAK1-STAT3-signaling have been developed, understanding their precise impact on signaling dynamics requires a systems biology approach. Here we present a mathematical model of IL-6-induced JAK1-STAT3 signaling that quantitatively links physiological IL-6 concentrations to the dynamics of IL-6-induced signal transduction and expression of target genes in hepatocytes. The mathematical model consists of coupled ordinary differential equations (ODE) and the model parameters were estimated by a maximum likelihood approach, whereas identifiability of the dynamic model parameters was ensured by the Profile Likelihood. Using model simulations coupled with experimental validation we could optimize the long-term impact of the JAK-inhibitor Ruxolitinib, a therapeutic compound that is quickly metabolized. Model-predicted doses and timing of treatments helps to improve the reduction of inflammatory APP gene expression in primary mouse hepatocytes close to levels observed during regenerative conditions. The concept of improved efficacy of the inhibitor through multiple treatments at optimized time intervals was confirmed in primary human hepatocytes. Thus, combining quantitative data generation with mathematical modeling suggests that repetitive treatment with Ruxolitinib is required to effectively target excessive inflammatory responses without exceeding doses recommended by the clinical guidelines.
Collapse
Affiliation(s)
- Svantje Sobotta
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Andreas Raue
- Discovery Division, Merrimack Pharmaceuticals, Cambridge, MA, United States
| | - Xiaoyun Huang
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Joep Vanlier
- Institute of Physics, Albert Ludwigs University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Anja Jünger
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Sebastian Bohl
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Ute Albrecht
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Maximilian J Hahnel
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Stephanie Wolf
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Nikola S Mueller
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lorenza A D'Alessandro
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Stephanie Mueller-Bohl
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Martin E Boehm
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Philippe Lucarelli
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Sandra Bonefas
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, Leipzig University, Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, Leipzig University, Leipzig, Germany
| | - Wolf D Lehmann
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | | | - Frank van der Hoeven
- Transgenic Service, Center for Preclinical Research, German Cancer Research Center, Heidelberg, Germany
| | - Norbert Gretz
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany.,Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Christian Ehlting
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Johannes G Bode
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, University Hospital, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Jens Timmer
- Institute of Physics, Albert Ludwigs University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Marcel Schilling
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
20
|
Pinno J, Bongartz H, Klepsch O, Wundrack N, Poli V, Schaper F, Dittrich A. Interleukin-6 influences stress-signalling by reducing the expression of the mTOR-inhibitor REDD1 in a STAT3-dependent manner. Cell Signal 2016; 28:907-16. [PMID: 27094713 DOI: 10.1016/j.cellsig.2016.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/05/2016] [Accepted: 04/09/2016] [Indexed: 11/30/2022]
Abstract
Interleukin 6 (IL-6) is a pleiotropic cytokine and a strong activator of Mammalian Target of Rapamycin (mTOR). In contrast, mTOR activity is negatively regulated by Regulated in Development and DNA Damage Responses 1 (REDD1). Expression of REDD1 is induced by cellular stressors such as glucocorticoids and DNA damaging agents. We show that the expression of basal as well as stress-induced REDD1 is reduced by IL-6. The reduction of REDD1 expression by IL-6 is independent of proteasomal or caspase-mediated degradation of REDD1 protein. Instead, induction of REDD1 mRNA is reduced by IL-6. The regulation of REDD1 expression by IL-6 is independent of Phosphatidylinositide-3-Kinase (PI3K) and Mitogen-Activated Protein Kinase (MAPK) signalling but depends on the expression and activation of Signal Transducer and Activator of Transcription 3 (STAT3). Furthermore, the reduction of basal REDD1 expression by IL-6 correlates with IL-6-induced activation of mTOR signalling. Inhibition of STAT3 activation blocks IL-6-induced mTOR activation. In summary, we present a novel STAT3-dependent mechanism of both IL-6-induced activation of mTOR and IL-6-dependent reversion of stress-induced inhibition of mTOR activity.
Collapse
Affiliation(s)
- Jessica Pinno
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Hannes Bongartz
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Oliver Klepsch
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Nicole Wundrack
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Valeria Poli
- Molecular Biotechnology Center (MBC), Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126 Turin, Italy.
| | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| | - Anna Dittrich
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Universitätsplatz 2, Gebäude 28, 39106 Magdeburg, Germany.
| |
Collapse
|
21
|
Schaper F, Rose-John S. Interleukin-6: Biology, signaling and strategies of blockade. Cytokine Growth Factor Rev 2015; 26:475-87. [DOI: 10.1016/j.cytogfr.2015.07.004] [Citation(s) in RCA: 287] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023]
|
22
|
Dittrich A, Hessenkemper W, Schaper F. Systems biology of IL-6, IL-12 family cytokines. Cytokine Growth Factor Rev 2015; 26:595-602. [PMID: 26187858 DOI: 10.1016/j.cytogfr.2015.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Interleukin-6-type cytokines play important roles in the communication between cells of multicellular organisms. They are involved in the regulation of complex cellular processes such as proliferation and differentiation and act as key player during inflammation and immune response. A major challenge is to understand how these complex non-linear processes are connected and regulated. Systems biology approaches are used to tackle this challenge in an iterative process of quantitative experimental and mathematical analyses. Here we review quantitative experimental studies and systems biology approaches dealing with the function of Interleukin-6-type cytokines in physiological and pathophysiological conditions. These approaches cover the analyses of signal transduction on a cellular level up to pharmacokinetic and pharmacodynamic studies on a whole organism level.
Collapse
Affiliation(s)
- Anna Dittrich
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Wiebke Hessenkemper
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| | - Fred Schaper
- Institute of Biology, Otto-von-Guericke-University, Universitätsplatz 2, 39106 Magdeburg, Germany.
| |
Collapse
|
23
|
Rinis N, Küster A, Schmitz-Van de Leur H, Mohr A, Müller-Newen G. Intracellular signaling prevents effective blockade of oncogenic gp130 mutants by neutralizing antibodies. Cell Commun Signal 2014; 12:14. [PMID: 24612692 PMCID: PMC4007646 DOI: 10.1186/1478-811x-12-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 02/24/2014] [Indexed: 12/26/2022] Open
Abstract
Background Short in-frame deletions in the second extracellular domain of the cytokine receptor gp130 are the leading cause of inflammatory hepatocellular adenomas (IHCAs). The deletions render gp130 constitutively active. In this study we investigate the intracellular signaling potential of one of the most potent constitutively active gp130 mutants (CAgp130) found in IHCAs. Results Trafficking and signaling of CAgp130 were studied in stably transfected cell lines that allowed the inducible expression of CAgp130 fused to fluorescent proteins such as YFP and mCherry. In contrast to the predominantly highly glycosylated gp130 wild type (WTgp130), CAgp130 is preferentially found in the less glycosylated high-mannose form. Accordingly, the mutated receptor is retained intracellularly and therefore less prominently expressed at the cell surface. CAgp130 persistently activates Stat3 despite the presence of the feedback inhibitor SOCS3 but fails to activate Erk1/2. De novo synthesized CAgp130 signals already from the ER-Golgi compartment before having reached the plasma membrane. Cell surface expressed and endocytosed CAgp130 do not significantly contribute to signaling. As a consequence, Stat3 activation through CAgp130 cannot be inhibited by neutralizing gp130 antibodies but through overexpression of a dominant-negative Stat3 mutant. Conclusion CAgp130 and WTgp130 differ significantly with respect to glycosylation, trafficking and signaling. As a consequence of intracellular signaling pharmacological inhibition of CAgp130 will not be achieved by targeting the receptor extracellularly but by compounds that act from within the cell.
Collapse
Affiliation(s)
| | | | | | | | - Gerhard Müller-Newen
- Institute of Biochemistry and Molecular Biology, RWTH Aachen University, Pauwelsstraße 30, Aachen 52074, Germany.
| |
Collapse
|
24
|
Calçada D, Vianello D, Giampieri E, Sala C, Castellani G, de Graaf A, Kremer B, van Ommen B, Feskens E, Santoro A, Franceschi C, Bouwman J. The role of low-grade inflammation and metabolic flexibility in aging and nutritional modulation thereof: a systems biology approach. Mech Ageing Dev 2014; 136-137:138-47. [PMID: 24462698 DOI: 10.1016/j.mad.2014.01.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 02/07/2023]
Abstract
Aging is a biological process characterized by the progressive functional decline of many interrelated physiological systems. In particular, aging is associated with the development of a systemic state of low-grade chronic inflammation (inflammaging), and with progressive deterioration of metabolic function. Systems biology has helped in identifying the mediators and pathways involved in these phenomena, mainly through the application of high-throughput screening methods, valued for their molecular comprehensiveness. Nevertheless, inflammation and metabolic regulation are dynamical processes whose behavior must be understood at multiple levels of biological organization (molecular, cellular, organ, and system levels) and on multiple time scales. Mathematical modeling of such behavior, with incorporation of mechanistic knowledge on interactions between inflammatory and metabolic mediators, may help in devising nutritional interventions capable of preventing, or ameliorating, the age-associated functional decline of the corresponding systems.
Collapse
Affiliation(s)
- Dulce Calçada
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands; Wageningen University, Department of Human Nutrition, Wageningen, The Netherlands
| | - Dario Vianello
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy
| | - Enrico Giampieri
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Claudia Sala
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Gastone Castellani
- University of Bologna, Department of Physics and Astronomy, 40127 Bologna, Italy
| | - Albert de Graaf
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Bas Kremer
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Ben van Ommen
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands
| | - Edith Feskens
- Wageningen University, Department of Human Nutrition, Wageningen, The Netherlands
| | - Aurelia Santoro
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy
| | - Claudio Franceschi
- University of Bologna, Department of Experimental, Diagnostic and Specialty Medicine, Via San Giacomo 12, 40126 Bologna, Italy; University of Bologna, Interdepartmental Centre "L. Galvani" (CIG), 40126 Bologna, Italy
| | - Jildau Bouwman
- TNO, Microbiology and Systems Biology Group, Utrechtseweg 48, 3704 HE Zeist, The Netherlands.
| |
Collapse
|
25
|
Gu J, Han T, Ma RH, Zhu YL, Jia YN, Du JJ, Chen Y, Jiang XJ, Xie XD, Guo X. SHP2 promotes laryngeal cancer growth through the Ras/Raf/Mek/Erk pathway and serves as a prognostic indicator for laryngeal cancer. Int J Oncol 2013; 44:481-90. [PMID: 24297342 DOI: 10.3892/ijo.2013.2191] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/05/2013] [Indexed: 11/06/2022] Open
Abstract
The overall survival rate and prognosis of patients with laryngeal cancer are not optimistic despite advances in therapeutic techniques. Gene expression prognostic models enable the development of more appropriate treatment strategies. The human gene PTPN11 encoding a non-receptor protein tyrosine phosphatase, Src homology phosphotyrosine phosphatase 2 (SHP2), is a well-documented proto-oncogene in various malignancies. This study investigated the role of SHP2 expression and associated clinical manifestations in laryngeal cancer using a tissue microarray of 112 pairs of laryngeal cancer samples and corresponding adjacent normal mucosae. SHP2 expression increased in laryngeal cancer, and this result was associated with the poor survival rate of laryngeal cancer patients. Moreover, increased SHP2 expression remarkably promoted the growth of laryngeal cancer cells in vitro and tumorigenicity of laryngeal cancer cells in vivo. The Ras/Raf/Mek/Erk pathway was also found to be involved in the SHP2-induced growth of laryngeal cancer cells. Overall, our findings indicated that SHP2 plays an important role in laryngeal cancer tumorigenesis and that its expression is negatively correlated with the prognosis of patients. Thus, SHP2 may be a promising combinational therapeutic target for treatment of laryngeal cancer. The interference of SHP2 expression can serve as a novel strategy for laryngeal cancer treatment.
Collapse
Affiliation(s)
- Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Tao Han
- Department of Oncology, General Hospital of Shenyang Military Region, Shenyang 110016, P.R. China
| | - Rui-Hang Ma
- Department of Operation Room, General Hospital of Shenyang Military Region, Shenyang 110016, P.R. China
| | - Yu-Lin Zhu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Yi-Na Jia
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Jing-Jing Du
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Yu Chen
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Xue-Jun Jiang
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| | - Xiao-Dong Xie
- Department of Oncology, General Hospital of Shenyang Military Region, Shenyang 110016, P.R. China
| | - Xing Guo
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang 110001, P.R. China
| |
Collapse
|
26
|
Stec W, Vidal O, Zeidler MP. Drosophila SOCS36E negatively regulates JAK/STAT pathway signaling via two separable mechanisms. Mol Biol Cell 2013; 24:3000-9. [PMID: 23885117 PMCID: PMC3771960 DOI: 10.1091/mbc.e13-05-0275] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The low-complexity Drosophila genome includes previously uncharacterized SOCS36E, an ancestral SOCS4/5 homologue. It is shown that SOCS36E suppresses JAK/STAT signaling through two separate mechanisms: via receptor stability, mediated by the conserved SOCS-box domain, and via suppression of receptor phosphorylation that requires the N-terminal domain. Conserved from humans to Drosophila, the Janus kinase/signal transducer and activators of transcription (JAK/STAT) signaling cascade is essential for multiple developmental and homeostatic processes, with regulatory molecules controlling pathway activity also highly conserved. We characterize the Drosophila JAK/STAT pathway regulator SOCS36E and show that it functions via two independent mechanisms. First, we show that Drosophila Elongin B/C and Cullin-5 act via the SOCS-box of SOCS36E to reduce pathway activity specifically in response to ligand stimulation—a process that involves endocytic trafficking and lysosomal degradation of the Domeless (Dome) receptor. Second, SOCS36E also suppresses both stimulated and basal pathway activity via an Elongin/Cullin-independent mechanism that is mediated by the N-terminus of SOCS36E, which is required for the physical interaction of SOCS36E with Dome. Although some human SOCS proteins contain N-terminal kinase-inhibitory domains, we do not identify such a region in SOCS36E and propose a model wherein the N-terminal of SOCS36E blocks access to tyrosine residues in Dome. Our biochemical analysis of a SOCS-family regulator from a lower organism highlights the fundamental conserved roles played by regulatory mechanisms in signal transduction.
Collapse
Affiliation(s)
- Wojciech Stec
- MRC Centre for Development and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | | | | |
Collapse
|
27
|
Blätke MA, Dittrich A, Rohr C, Heiner M, Schaper F, Marwan W. JAK/STAT signalling – an executable model assembled from molecule-centred modules demonstrating a module-oriented database concept for systems and synthetic biology. MOLECULAR BIOSYSTEMS 2013; 9:1290-307. [DOI: 10.1039/c3mb25593j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|