1
|
Kwon J, Arsenis C, Suessmilch M, McColl A, Cavanagh J, Morris BJ. Differential Effects of Toll-Like Receptor Activation and Differential Mediation by MAP Kinases of Immune Responses in Microglial Cells. Cell Mol Neurobiol 2021; 42:2655-2671. [PMID: 34297254 PMCID: PMC9560989 DOI: 10.1007/s10571-021-01127-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/10/2021] [Indexed: 10/26/2022]
Abstract
Microglial activation is believed to play a role in many psychiatric and neurodegenerative diseases. Based largely on evidence from other cell types, it is widely thought that MAP kinase (ERK, JNK and p38) signalling pathways contribute strongly to microglial activation following immune stimuli acting on toll-like receptor (TLR) 3 or TLR4. We report here that exposure of SimA9 mouse microglial cell line to immune mimetics stimulating TLR4 (lipopolysaccharide-LPS) or TLR7/8 (resiquimod/R848), results in marked MAP kinase activation, followed by induction of nitric oxide synthase, and various cytokines/chemokines. However, in contrast to TLR4 or TLR7/8 stimulation, very few effects of TLR3 stimulation by poly-inosine/cytidine (polyI:C) were detected. Induction of chemokines/cytokines at the mRNA level by LPS and resiquimod were, in general, only marginally affected by MAP kinase inhibition, and expression of TNF, Ccl2 and Ccl5 mRNAs, along with nitrite production, were enhanced by p38 inhibition in a stimulus-specific manner. Selective JNK inhibition enhanced Ccl2 and Ccl5 release. Many distinct responses to stimulation of TLR4 and TLR7 were observed, with JNK mediating TNF protein induction by the latter but not the former, and suppressing Ccl5 release by the former but not the latter. These data reveal complex modulation by MAP kinases of microglial responses to immune challenge, including a dampening of some responses. They demonstrate that abnormal levels of JNK or p38 signalling in microglial cells will perturb their profile of cytokine and chemokine release, potentially contributing to abnormal inflammatory patterns in CNS disease states.
Collapse
Affiliation(s)
- Jaedeok Kwon
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK.,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Christos Arsenis
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK
| | - Maria Suessmilch
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Alison McColl
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, West Medical Building, Glasgow, G12 8QQ, UK.
| |
Collapse
|
2
|
Le A, Azouz A, Thomas S, Istaces N, Nguyen M, Goriely S. JNK1 Signaling Downstream of the EGFR Pathway Contributes to Aldara ®-Induced Skin Inflammation. Front Immunol 2021; 11:604785. [PMID: 33613525 PMCID: PMC7892463 DOI: 10.3389/fimmu.2020.604785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/10/2020] [Indexed: 01/12/2023] Open
Abstract
c-Jun N-terminal protein kinase 1 (JNK1) is involved in multiple biological processes but its implication in inflammatory skin diseases is still poorly defined. Herein, we studied the role of JNK1 in the context of Aldara®-induced skin inflammation. We observed that constitutive ablation of JNK1 reduced Aldara®-induced acanthosis and expression of inflammatory markers. Conditional deletion of JNK1 in myeloid cells led to reduced skin inflammation, a finding that was associated with impaired Aldara®-induced inflammasome activation in vitro. Next, we evaluated the specific role of JNK1 in epidermal cells. We observed reduced Aldara®-induced acanthosis despite similar levels of inflammatory markers. Transcriptomic and epigenomic analysis of keratinocytes revealed the potential involvement of JNK1 in the EGFR signaling pathway. Finally, we show that inhibition of the EGFR pathway reduced Aldara®-induced acanthosis. Taken together, these data indicate that JNK1 plays a dual role in the context of psoriasis by regulating the production of inflammatory cytokines by myeloid cells and the sensitivity of keratinocytes to EGFR ligands. These results suggest that JNK1 could represent a valuable therapeutic target in the context of psoriasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Stanislas Goriely
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles, Gosselies, Belgium
| |
Collapse
|
3
|
Activation of c-Jun N-Terminal Kinase, a Potential Therapeutic Target in Autoimmune Arthritis. Cells 2020; 9:cells9112466. [PMID: 33198301 PMCID: PMC7696795 DOI: 10.3390/cells9112466] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
The c-Jun-N-terminal kinase (JNK) is a critical mediator involved in various physiological processes, such as immune responses, and the pathogenesis of various diseases, including autoimmune disorders. JNK is one of the crucial downstream signaling molecules of various immune triggers, mainly proinflammatory cytokines, in autoimmune arthritic conditions, mainly including rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis. The activation of JNK is regulated in a complex manner by upstream kinases and phosphatases. Noticeably, different subtypes of JNKs behave differentially in immune responses. Furthermore, aside from biologics targeting proinflammatory cytokines, small-molecule inhibitors targeting signaling molecules such as Janus kinases can act as very powerful therapeutics in autoimmune arthritis patients unresponsiveness to conventional synthetic antirheumatic drugs. Nevertheless, despite these encouraging therapies, a population of patients with an inadequate therapeutic response to all currently available medications still remains. These findings identify the critical signaling molecule JNK as an attractive target for investigation of the immunopathogenesis of autoimmune disorders and for consideration as a potential therapeutic target for patients with autoimmune arthritis to achieve better disease control. This review provides a useful overview of the roles of JNK, how JNK is regulated in immunopathogenic responses, and the potential of therapeutically targeting JNK in patients with autoimmune arthritis.
Collapse
|
4
|
Bagnoud M, Briner M, Remlinger J, Meli I, Schuetz S, Pistor M, Salmen A, Chan A, Hoepner R. c-Jun N-Terminal Kinase as a Therapeutic Target in Experimental Autoimmune Encephalomyelitis. Cells 2020; 9:cells9102154. [PMID: 32977663 PMCID: PMC7598244 DOI: 10.3390/cells9102154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
c-Jun N-terminal kinase (JNK) is upregulated during multiple sclerosis relapses and at the peak of experimental autoimmune encephalomyelitis (EAE). We aim to investigate the effects of pharmacological pan-JNK inhibition on the course of myelin oligodendrocyte glycoprotein (MOG35-55) EAE disease using in vivo and in vitro experimental models. EAE was induced in female C57BL/6JRj wild type mice using MOG35-55. SP600125 (SP), a reversible adenosine triphosphate competitive pan-JNK inhibitor, was then given orally after disease onset. Positive correlation between SP plasma and brain concentration was observed. Nine, but not three, consecutive days of SP treatment led to a significant dose-dependent decrease of mean cumulative MOG35-55 EAE severity that was associated with increased mRNA expression of interferon gamma (INF-γ) and tumor necrosis factor alpha (TNF-α) in the spinal cord. On a histological level, reduced spinal cord immune cell-infiltration predominantly of CD3+ T cells as well as increased activity of Iba1+ cells were observed in treated animals. In addition, in vitro incubation of murine and human CD3+ T cells with SP resulted in reduced T cell apoptosis and proliferation. In conclusion, our study demonstrates that pharmacological pan-JNK inhibition might be a treatment strategy for autoimmune central nervous system demyelination.
Collapse
Affiliation(s)
- Maud Bagnoud
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010 Bern, Switzerland
- Correspondence: ; Tel.: +41-31-6323076
| | - Myriam Briner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Jana Remlinger
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3010 Bern, Switzerland
| | - Ivo Meli
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Sara Schuetz
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Maximilian Pistor
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.B.); (J.R.); (I.M.); (S.S.); (M.P.); (A.S.); (A.C.); (R.H.)
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
5
|
Lombard CK, Davis AL, Inukai T, Maly DJ. Allosteric Modulation of JNK Docking Site Interactions with ATP-Competitive Inhibitors. Biochemistry 2018; 57:5897-5909. [PMID: 30211540 DOI: 10.1021/acs.biochem.8b00776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The c-Jun N-terminal kinases (JNKs) play a wide variety of roles in cellular signaling processes, dictating important, and even divergent, cellular fates. These essential kinases possess docking surfaces distal to their active sites that interact with diverse binding partners, including upstream activators, downstream substrates, and protein scaffolds. Prior studies have suggested that the interactions of certain protein-binding partners with one such JNK docking surface, termed the D-recruitment site (DRS), can allosterically influence the conformational state of the ATP-binding pocket of JNKs. To further explore the allosteric relationship between the ATP-binding pockets and DRSs of JNKs, we investigated how the interactions of the scaffolding protein JIP1, as well as the upstream activators MKK4 and MKK7, are allosterically influenced by the ATP-binding site occupancy of the JNKs. We show that the affinity of the JNKs for JIP1 can be divergently modulated with ATP-competitive inhibitors, with a >50-fold difference in dissociation constant observed between the lowest- and highest-affinity JNK1-inhibitor complexes. Furthermore, we found that we could promote or attenuate phosphorylation of JNK1's activation loop by MKK4 and MKK7, by varying the ATP-binding site occupancy. Given that JIP1, MKK4, and MKK7 all interact with JNK DRSs, these results demonstrate that there is functional allostery between the ATP-binding sites and DRSs of these kinases. Furthermore, our studies suggest that ATP-competitive inhibitors can allosterically influence the intracellular binding partners of the JNKs.
Collapse
Affiliation(s)
- Chloe K Lombard
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States
| | - Audrey L Davis
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States
| | - Takayuki Inukai
- Medicinal Chemistry Research Laboratories , Ono Pharmaceutical Company, Ltd. , 3-1-1 Sakurai , Shimamoto, Mishima, Osaka 618-8585 , Japan
| | - Dustin J Maly
- Department of Chemistry , University of Washington , Seattle , Washington 98117 , United States.,Department of Biochemistry , University of Washington , Seattle , Washington 98117 , United States
| |
Collapse
|
6
|
c-Jun N-terminal kinase 1 defective CD4+CD25+FoxP3+ cells prolong islet allograft survival in diabetic mice. Sci Rep 2018; 8:3310. [PMID: 29459675 PMCID: PMC5818514 DOI: 10.1038/s41598-018-21477-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 02/06/2023] Open
Abstract
CD4+CD25+FoxP3+ cells (Tregs) inhibit inflammatory immune responses to allografts. Here, we found that co-transplantation of allogeneic pancreatic islets with Tregs that are defective in c-Jun N-terminal kinase 1 (JNK1) signaling prolongs islet allograft survival in the liver parenchyma of chemically induced diabetic mice (CDM). Adoptively transferred JNK1−/− but not wild-type (WT) Tregs survive longer in the liver parenchyma of CDM. JNK1−/− Tregs are resistant to apoptosis and express anti-apoptotic molecules. JNK1−/− Tregs express higher levels of lymphocyte activation gene-3 molecule (LAG-3) on their surface and produce higher amounts of the anti-inflammatory cytokine interleukin (IL)-10 compared with WT Tregs. JNK1−/− Tregs inhibit liver alloimmune responses more efficiently than WT Tregs. JNK1−/− but not WT Tregs are able to inhibit IL-17 and IL-21 production through enhanced LAG-3 expression and IL-10 production. Our study identifies a novel role of JNK1 signaling in Tregs that enhances islet allograft survival in the liver parenchyma of CDM.
Collapse
|
7
|
Fatty acid transport protein 1 enhances the macrophage inflammatory response by coupling with ceramide and c-Jun N-terminal kinase signaling. Int Immunopharmacol 2017; 55:205-215. [PMID: 29272817 DOI: 10.1016/j.intimp.2017.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 11/17/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023]
Abstract
Macrophages are important cells that need to be controlled at the site of inflammation. Several factors are involved in chronic inflammation and its timely resolution. Free fatty acids drive the inflammatory response in macrophages and contribute to the vicious cycle of the inflammatory response. However, the identity of the uptake pathways of fatty acids is not fully clear in macrophages and how the inflammatory responses are regulated by the uptake of fatty acids remain poorly understood. We investigated the relationship between fatty acid transport protein (FATP) and the inflammatory response signaling pathway in macrophages as the first report. The FATP family has composed six isoforms, FATP1-6. We found that FATP1 is the most highly expressed isoform in macrophages. Forced expression of FATP1 enhanced production of inflammatory cytokines, such as TNFα and IL-6 concomitant with the increased uptake of fatty acids, increased level of ceramide, and increased phosphorylation of c-Jun N-terminal kinase (JNK). The enhancement by FATP1 was abolished by treatment with a JNK inhibitor, NF-κB inhibitor, or ceramide synthesis inhibitor. siRNA-mediated knockdown of FATP1 strongly inhibited the production of TNFα and IL-6. Similarly, an inhibitor of FATP1 inhibited the production of TNFα and IL-6. Finally, an inhibitor of FATP1 attenuated the production of inflammatory cytokines in bronchoalveolar lavage fluid in an LPS-induced acute lung injury in vivo mouse model. In summary, we propose that FATP1 is an important regulator of inflammatory response signaling in macrophages. Our findings suggest that ceramide-JNK signaling is important to terminate or sustain inflammation.
Collapse
|
8
|
Molnarfi N, Prod'homme T, Schulze-Topphoff U, Spencer CM, Weber MS, Patarroyo JC, Lalive PH, Zamvil SS. Glatiramer acetate treatment negatively regulates type I interferon signaling. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e179. [PMID: 26601118 PMCID: PMC4645172 DOI: 10.1212/nxi.0000000000000179] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023]
Abstract
Objective: Glatiramer acetate (GA; Copaxone), a disease-modifying therapy for multiple sclerosis (MS), promotes development of anti-inflammatory (M2, type II) monocytes that can direct differentiation of regulatory T cells. We investigated the innate immune signaling pathways that participate in GA-mediated M2 monocyte polarization. Methods: Monocytes were isolated from myeloid differentiation primary response gene 88 (MyD88)–deficient, Toll-IL-1 receptor domain–containing adaptor inducing interferon (IFN)–β (TRIF)–deficient, IFN-α/β receptor subunit 1 (IFNAR1)–deficient, and wild-type (WT) mice and human peripheral blood. GA-treated monocytes were stimulated with Toll-like receptor ligands, then evaluated for activation of kinases and transcription factors involved in innate immunity, and secretion of proinflammatory cytokines. GA-treated mice were evaluated for cytokine secretion and susceptibility to experimental autoimmune encephalomyelitis. Results: GA-mediated inhibition of proinflammatory cytokine production by monocytes occurred independently of MyD88 and nuclear factor–κB, but was blocked by TRIF deficiency. Furthermore, GA did not provide clinical benefit in TRIF-deficient mice. GA inhibited activation of p38 mitogen-activated protein kinase, an upstream regulator of activating transcription factor (ATF)–2, and c-Jun N-terminal kinase 1, which regulates IFN regulatory factor 3 (IRF3). Consequently, nuclear translocation of ATF-2 and IRF3, components of the IFN-β enhanceosome, was impaired. Consistent with these observations, GA inhibited production of IFN-β in vivo in WT mice, but did not modulate proinflammatory cytokine production by monocytes from IFNAR1-deficient mice. Conclusion: Our results demonstrate that GA inhibits the type I IFN pathway in M2 polarization of monocytes independently of MyD88, providing an important mechanism connecting innate and adaptive immune modulation in GA therapy and valuable insight regarding its potential use with other MS treatments.
Collapse
Affiliation(s)
- Nicolas Molnarfi
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Thomas Prod'homme
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Ulf Schulze-Topphoff
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Collin M Spencer
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Martin S Weber
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Juan C Patarroyo
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Patrice H Lalive
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology (N.M., T.P., U.S.-T., C.M.S., J.C.P., S.S.Z.), University of California, San Francisco; the Institute of Neuropathology and Department of Neurology (M.S.W.), University Medical Center, Georg-August University, Göttingen, Germany; the Department of Pathology and Immunology (P.H.L.), Faculty of Medicine, University of Geneva; and the Department of Neurosciences (P.H.L.), Division of Neurology, University Hospital of Geneva, Switzerland. N.M. is currently affiliated with the Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, and Department of Neurosciences, Division of Neurology, University Hospital of Geneva, Switzerland. T.P. is currently affiliated with Momenta Pharmaceuticals, Cambridge, MA. U.S.-T. is currently affiliated with Silence Therapeutics GmbH, Berlin, Germany. J.C.P. is currently affiliated with Vedanta Biosciences, Inc., Cambridge, MA
| |
Collapse
|
9
|
Dai MM, Wu H, Li H, Chen J, Chen JY, Hu SL, Shen C. Effects and mechanisms of Geniposide on rats with adjuvant arthritis. Int Immunopharmacol 2014; 20:46-53. [PMID: 24583144 DOI: 10.1016/j.intimp.2014.02.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/18/2014] [Accepted: 02/11/2014] [Indexed: 01/14/2023]
Abstract
Geniposide (GE), an iridoid glycoside compound, is the major active ingredient of Gardenia jasminoides Ellis (GJ) fruit which has anti-inflammatory and other important therapeutic activities. The aim of this study was to investigate the effects of GE on adjuvant arthritis (AA) rats and its possible mechanisms. AA was induced by injecting with Freund's complete adjuvant (FCA). Male SD rats were subjected to treatment with GE at 30, 60 and 120mg/kg from days 18 to 24 after immunization. Lymphocyte proliferation was assessed by MTT. Interleukin (IL)-6, IL-17, IL-4 and transforming growth factor-beta 1 (TGF-β1) were determined by ELISA. c-Jun N-terminal kinase (JNK) and phospho-JNK (p-JNK) were detected by Western blot. GE (60, 120mg/kg) significantly relieved the secondary hind paw swelling and arthritis index, along with decreased Th17-cells cytokines and increased Treg-cell cytokines in mesenteric lymph node lymphocytes (MLNL) and peripheral blood lymphocytes (PBL) of AA rats. In addition, GE decreased the expression of p-JNK in MLNL and PBL of AA rats. In vivo study, it was also observed that GE attenuated histopathologic changes of MLN in AA rats. Collectively, GE might exert its anti-inflammatory and immunoregulatory effects through inducing Th17 cell immune tolerance and enhancing Treg cell-mediated activities by down-regulating the expression of p-JNK. The mechanisms of GE on JNK signaling in MLNL and PBL may play critical roles in the pathogenesis of rheumatoid arthritis.
Collapse
Affiliation(s)
- Miao-Miao Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China.
| | - Hui Li
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| | - Jian Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| | - Jin-Yun Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| | - Shun-Li Hu
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| | - Chen Shen
- College of Pharmacy, Anhui University of Chinese Medicine, Key Laboratory of Modernized Chinese Medicine in Anhui Province, Hefei 230031, Anhui Province, China
| |
Collapse
|
10
|
Abstract
JNK is involved in a broad range of physiological processes. Several inflammatory and neurodegenerative diseases, such as multiple sclerosis, Alzheimer's and Parkinson's disease have been linked with the dysregulated JNK pathway. Research on disease models using the relevant knockout mice has highlighted the importance of specific JNK isoformsin-particular disorders and has stimulated further efforts in the drug-discovery area. However, most of the experimental evidence for the efficacy of JNK inhibition in animal models is from studies using JNK inhibitors, which are not isoform selective. Some of the more recent compounds exhibit good oral bioavailability, CNS penetration and selectivity against the rest of the kinome. Efforts to design isoform-selective inhibitors have produced a number of examples with various selectivity profiles. This article presents recent progress in this area and comment on the role of isoform selectivity for efficacy.
Collapse
|
11
|
Feng Y, Chambers JW, Iqbal S, Koenig M, Park H, Cherry L, Hernandez P, Figuera-Losada M, LoGrasso PV. A small molecule bidentate-binding dual inhibitor probe of the LRRK2 and JNK kinases. ACS Chem Biol 2013; 8:1747-54. [PMID: 23751758 DOI: 10.1021/cb3006165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Both JNK and LRRK2 are associated with Parkinson's disease (PD). Here we report a reasonably selective and potent kinase inhibitor (compound 6) that bound to both JNK and LRRK2 (a dual inhibitor). A bidentate-binding strategy that simultaneously utilized the ATP hinge binding and a unique protein surface site outside of the ATP pocket was applied to the design and identification of this kind of inhibitor. Compound 6 was a potent JNK3 and modest LRRK2 dual inhibitor with an enzyme IC50 value of 12 nM and 99 nM (LRRK2-G2019S), respectively. Compound 6 also exhibited good cell potency, inhibited LRRK2:G2019S-induced mitochondrial dysfunction in SHSY5Y cells, and was demonstrated to be reasonably selective against a panel of 116 kinases from representative kinase families. Design of such a probe molecule may help enable testing if dual JNK and LRRK2 inhibitions have added or synergistic efficacy in protecting against neurodegeneration in PD.
Collapse
Affiliation(s)
- Yangbo Feng
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Jeremy W. Chambers
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Sarah Iqbal
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Marcel Koenig
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - HaJeung Park
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Lisa Cherry
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Pamela Hernandez
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Mariana Figuera-Losada
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| | - Philip V. LoGrasso
- Medicinal Chemistry, ‡Discovery Biology, §Modeling/Crystallography
Facility, Translational Research Institute, and ∥Department of Molecular Therapeutics, The Scripps Research Institute, Florida,
Jupiter, Florida 33458, United States
| |
Collapse
|
12
|
Karasinska JM, de Haan W, Franciosi S, Ruddle P, Fan J, Kruit JK, Stukas S, Lütjohann D, Gutmann DH, Wellington CL, Hayden MR. ABCA1 influences neuroinflammation and neuronal death. Neurobiol Dis 2013; 54:445-55. [DOI: 10.1016/j.nbd.2013.01.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 01/04/2013] [Accepted: 01/17/2013] [Indexed: 11/29/2022] Open
|
13
|
Guma M, Firestein GS. c-Jun N-Terminal Kinase in Inflammation and Rheumatic Diseases. Open Rheumatol J 2012; 6:220-31. [PMID: 23028407 PMCID: PMC3460413 DOI: 10.2174/1874312901206010220] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Revised: 01/03/2011] [Accepted: 07/13/2011] [Indexed: 01/24/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are members of the mitogen-activated protein kinase (MAPK) family and are activated by environmental stress. JNK is also activated by proinflammatory cytokines, such as TNF and IL-1, and Toll-like receptor ligands. This pathway, therefore, can act as a critical convergence point in immune system signaling for both adaptive and innate responses. Like other MAPKs, the JNKs are activated via the sequential activation of protein kinases that includes two dual-specificity MAP kinase kinases (MKK4 and MKK7) and multiple MAP kinase kinase kinases. MAPKs, including JNKs, can be deactivated by a specialized group of phosphatases, called MAP kinase phosphatases. JNK phosphorylates and regulates the activity of transcription factors other than c-Jun, including ATF2, Elk-1, p53 and c-Myc and non-transcription factors, such as members of the Bcl-2 family. The pathway plays a critical role in cell proliferation, apoptosis, angiogenesis and migration. In this review, an overview of the functions that are related to rheumatic diseases is presented. In addition, some diseases in which JNK participates will be highlighted.
Collapse
Affiliation(s)
- Monica Guma
- Division of Rheumatology, Allergy and Immunology, UC San Diego School of Medicine, La Jolla, CA, USA
| | | |
Collapse
|
14
|
Integration of MicroRNA databases to study MicroRNAs associated with multiple sclerosis. Mol Neurobiol 2012; 45:520-35. [PMID: 22549745 DOI: 10.1007/s12035-012-8270-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 04/13/2012] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs which regulate many genes post-transcriptionally. In various contexts of medical science, miRNAs gained increasing attention over the last few years. Analyzing the functions, interactions and cellular effects of miRNAs is a very complex and challenging task. Many miRNA databases with diverse data contents have been developed. Here, we demonstrate how to integrate their information in a reasonable way on a set of miRNAs that were found to be dysregulated in the blood of patients with multiple sclerosis (MS). Using the miR2Disease database, we retrieved 16 miRNAs associated with MS according to four different studies. We studied the predicted and experimentally validated target genes of these miRNAs, their expression profiles in different blood cell types and brain tissues, the pathways and biological processes affected by these miRNAs as well as their regulation by transcription factors. Only miRNA-mRNA interactions that were predicted by at least seven different prediction algorithms were considered. This resulted in a network of 1,498 target genes. In this network, the MS-associated miRNAs hsa-miR-20a-5p and hsa-miR-20b-5p occurred as central hubs regulating about 500 genes each. Strikingly, many of the putative target genes play a role in T cell activation and signaling, and many have transcription factor activity. The latter suggests that miRNAs often act as regulators of regulators with many secondary effects on gene expression. Our present work provides a guideline on how information of different databases can be integrated in the analysis of miRNAs. Future investigations of miRNAs shall help to better understand the mechanisms underlying different diseases and their treatments.
Collapse
|
15
|
Denninger K, Rasmussen S, Larsen JM, Orskov C, Seier Poulsen S, Sørensen P, Christensen JP, Illges H, Odum N, Labuda T. JNK1, but not JNK2, is required in two mechanistically distinct models of inflammatory arthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:1884-93. [PMID: 21839715 DOI: 10.1016/j.ajpath.2011.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 06/09/2011] [Accepted: 06/20/2011] [Indexed: 11/29/2022]
Abstract
The roles of the c-Jun N-terminal kinases (JNKs) in inflammatory arthritis have been investigated; however, the roles of each isotype (ie, JNK1 and JNK2) in rheumatoid arthritis and conclusions about whether inhibition of one or both is necessary for amelioration of disease are unclear. By using JNK1- or JNK2-deficient mice in the collagen-induced arthritis and the KRN T-cell receptor transgenic mouse on C57BL/6 nonobese diabetic (K/BxN) serum transfer arthritis models, we demonstrate that JNK1 deficiency results in protection from arthritis, as judged by clinical score and histological evaluation in both models of inflammatory arthritis. In contrast, abrogation of JNK2 exacerbates disease. In collagen-induced arthritis, the distinct roles of the JNK isotypes can, at least in part, be explained by altered regulation of CD86 expression in JNK1- or JNK2-deficient macrophages in response to microbial products, thereby affecting T-cell-mediated immunity. The protection from K/BxN serum-induced arthritis in Jnk1(-/-) mice can also be explained by inept macrophage function because adoptive transfer of wild-type macrophages to Jnk1(-/-) mice restored disease susceptibility. Thus, our results provide a possible explanation for the modest therapeutic effects of broad JNK inhibitors and suggest that future therapies should selectively target the JNK1 isoform.
Collapse
Affiliation(s)
- Katja Denninger
- Department of Pharmacology and Pharmacotherapy, Institute of Molecular Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Guma M, Ronacher LM, Firestein GS, Karin M, Corr M. JNK-1 deficiency limits macrophage-mediated antigen-induced arthritis. ARTHRITIS AND RHEUMATISM 2011; 63:1603-12. [PMID: 21305529 PMCID: PMC3106119 DOI: 10.1002/art.30271] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To elucidate the nonredundant roles of JNK-1 and JNK-2 in antigen-induced arthritis (AIA). METHODS Mice that were genetically disrupted in Jnk1 or Jnk2 were primed by injection of methylated bovine serum albumin (mBSA) in Freund's complete adjuvant and then challenged on day 21 by intraarticular injection of mBSA into the right knee. Bone marrow chimeras were generated and similarly treated. Joints were harvested and prepared for histologic assessment. T cell responses were verified by cytokine and proliferation responses, and relative immunoglobulin responses were measured by enzyme-linked immunosorbent assay. Cytokine messenger RNA expression levels were measured by quantitative polymerase chain reaction analysis. Thioglycollate-elicited and zymosan A-elicited macrophage recruitment was tested in vivo, and cell migration was tested in vitro. The peptide inhibitor D-JNKi was injected daily starting 4 days after intraarticular injection of mBSA into wild-type (WT) mice, and inflammation was scored histologically. RESULTS JNK-1-deficient, but not JNK-2-deficient, mice had a reduction in inflammatory cell infiltration and joint damage. This effect was primarily restricted to hematopoietic cells, but B and T cell responses were preserved in mBSA-injected mice. JNK-1-deficient macrophages produced cytokines and chemokines at a level comparable to that in their WT counterparts. However, macrophage migration was impaired in vivo and in vitro. Targeting JNK with the peptide inhibitor D-JNKi dramatically reduced inflammation and joint destruction in WT mice. CONCLUSION AIA is dependent on JNK-1, but not JNK-2. JNK-1 is a promising molecular target for reducing autoimmune inflammation, since its inhibition impairs macrophage migration.
Collapse
Affiliation(s)
- Monica Guma
- Laboratory of Gene Regulation and Signal Transduction, University of California at San Diego, La Jolla, California
- Department of Pharmacology, University of California at San Diego, La Jolla, California
- Department of Pathology, University of California at San Diego, La Jolla, California
| | - Lisa M. Ronacher
- Division of Rheumatology, Allergy and Immunology, University of California at San Diego, La Jolla, California
| | - Gary S. Firestein
- Division of Rheumatology, Allergy and Immunology, University of California at San Diego, La Jolla, California
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, University of California at San Diego, La Jolla, California
- Department of Pharmacology, University of California at San Diego, La Jolla, California
- Department of Pathology, University of California at San Diego, La Jolla, California
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California at San Diego, La Jolla, California
| |
Collapse
|
17
|
Transcription factor Smad-independent T helper 17 cell induction by transforming-growth factor-β is mediated by suppression of eomesodermin. Immunity 2011; 34:741-54. [PMID: 21600798 DOI: 10.1016/j.immuni.2011.02.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 01/05/2011] [Accepted: 02/04/2011] [Indexed: 12/29/2022]
Abstract
Transforming growth factor-β (TGF-β) has been shown to be required for Th17 cell differentiation via Smad-independent mechanisms. The molecular mechanism underlying this pathway remains to be clarified, however. We searched for genes regulated by TGF-β through the Smad-independent pathway by using Smad2 and Smad3 double-deficient T cells and identified the transcription factor Eomesodermin (Eomes), whose expression was suppressed by TGF-β via the c-Jun N-terminal kinase (JNK)-c-Jun signaling pathway. Inhibition of JNK strongly suppressed disease in an in vivo EAE model as well as in vitro Th17 cell induction. Overexpression of Eomes substantially suppressed Th17 cell differentiation, whereas ablation of Eomes expression could substitute for TGF-β in Th17 cell induction in primary T cells. Eomes suppressed Rorc and Il17a promoters by directly binding to the proximal region of these promoters. In conclusion, the suppression of Eomes by TGF-β via the JNK pathway is an important mechanism for Smad-independent Th17 cell differentiation.
Collapse
|
18
|
Hedrich CM, Bream JH. Cell type-specific regulation of IL-10 expression in inflammation and disease. Immunol Res 2010; 47:185-206. [PMID: 20087682 DOI: 10.1007/s12026-009-8150-5] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IL-10 plays an essential part in controlling inflammation and instructing adaptive immune responses. Consequently, dysregulation of IL-10 is linked with susceptibility to numerous infectious and autoimmune diseases in mouse models and in humans. It has become increasingly clear that appropriate temporal/spatial expression of IL-10 may be the key to how IL-10 contributes to the delicate balance between inflammation and immunoregulation. The mechanisms that govern the cell type- and receptor-specific induction of IL-10, however, remain unclear. This is due largely to the wide distribution of cellular sources that express IL-10 under diverse stimulation conditions and in a variety of tissue compartments. Further complicating the issue is the fact that human IL-10 expression patterns appear to be under genetic influence resulting in differential expression and disease susceptibility. In this review, we discuss the cellular sources of IL-10, their link to disease phenotypes and the molecular mechanisms implicated in IL-10 regulation.
Collapse
Affiliation(s)
- Christian M Hedrich
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Room E5624, Baltimore, MD 21205-1901, USA
| | | |
Collapse
|
19
|
Azuma YT, Matsuo Y, Kuwamura M, Yancopoulos GD, Valenzuela DM, Murphy AJ, Nakajima H, Karow M, Takeuchi T. Interleukin-19 protects mice from innate-mediated colonic inflammation. Inflamm Bowel Dis 2010; 16:1017-28. [PMID: 19834971 DOI: 10.1002/ibd.21151] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) results from the chronic dysregulation of the mucosal immune system and the aberrant activation of both the innate and the adaptive immune responses. We used two complementary models of colonic inflammation to examine the roles of interleukin (IL)-19 in colonic inflammation and thus its possible role in IBD. METHODS Using gene-targeting, we generated IL-19-deficient mice. To study the activation of the innate immune response during colonic inflammation we characterized an innate immune-mediated model of colitis induced by dextran sulfate sodium (DSS). DSS can induce not only acute colitis but also chronic colitis. In addition to the acute DSS-induced colitis model, we used a chronic DSS-induced colitis model that is associated with the activation of both Th1 and Th2 cytokines as well as innate immune response in the colon. RESULTS We show that IL-19-deficient mice are more susceptible to experimental acute colitis induced by DSS, and this increased susceptibility is correlated with the accumulation of macrophages and the increased production of IFN-gamma, IL-1beta, IL-6, IL-12, TNF-alpha, and KC. Additionally, cytokine production in IL-19-deficient macrophages was enhanced on stimulation of lipopolysaccharide (LPS) through reduced phosphorylation of STAT1 and STAT3. Moreover, our results clearly demonstrate that IL-19 is required for B-cell infiltration during chronic DSS-induced colitis, which may be mediated by IL-13 and IL-6. CONCLUSIONS The finding that IL-19 drives pathogenic innate immune responses in the colon suggests that the selective targeting of IL-19 may be an effective therapeutic approach in the treatment of human IBD.
Collapse
Affiliation(s)
- Yasu-Taka Azuma
- Laboratory of Veterinary Pharmacology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Science, Izumisano, Osaka 598-8531, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
The development of Th1 lymphocytes is essential for cell-mediated immunity and resistance against intracellular pathogens. However, if left unregulated, the same response can cause serious damage to host tissues and lead to mortality. A number of different paracrine regulatory mechanisms involving distinct myeloid and lymphoid subpopulations have been implicated in controlling excessive secretion of inflammatory cytokines by Th1 cells. Much of this work has focused on interleukin (IL)-10, a cytokine with broad anti-inflammatory properties, one of which is to counteract the function of Th1 lymphocytes. While studying the role of IL-10 in regulating immunopathology during infection with the intracellular parasite Toxoplasma gondii, we discovered that the host-protective IL-10 derives in an autocrine manner from conventional interferon-gamma (IFN-gamma)-producing T-bet(+) Foxp3(neg) Th1 cells. In the following review, we will discuss these findings that support the general concept that production of IL-10 is an important self-regulatory function of CD4(+) T lymphocytes.
Collapse
Affiliation(s)
- D Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
21
|
Cohen SJ, Cohen IR, Nussbaum G. IL-10 mediates resistance to adoptive transfer experimental autoimmune encephalomyelitis in MyD88(-/-) mice. THE JOURNAL OF IMMUNOLOGY 2009; 184:212-21. [PMID: 19949074 DOI: 10.4049/jimmunol.0900296] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
MyD88 is an adaptor molecule that functions in the innate signaling induced by proinflammatory adjuvants that interact with TLRs. Mice lacking MyD88, for example, resist active experimental autoimmune encephalomyelitis (EAE) induced by immunization with an encephalitogenic myelin oligodendrocyte glycoprotein (MOG) peptide in CFA. We reasoned that MyD88(-/-) mice, nevertheless, should be susceptible to EAE mediated by adoptive transfer of activated encephalitogenic T cell lines, which do not require adjuvant signaling for their effector functions. We now report, however, that mice lacking MyD88 also resist adoptive EAE mediated by an anti-MOG T cell line that is strongly encephalitogenic in wild-type (WT) mice. The transferred anti-MOG T cells proliferated, secreted INF-gamma, and migrated to the CNS in the MyD88(-/-) mice, as they did in WT mice, but inflammatory infiltrates did not progress and clinical EAE did not develop. The resistance of the MyD88(-/-) mice to adoptive EAE mediated by the otherwise encephalitogenic T cells was found to result from the secretion of IL-10 by recipient T cells of two different specificities: those specific for MOG and those responding to the T cell clone itself-both anticlonotypic and antiergotypic T regulators were detected. IL-10-producing anti-MOG T cells isolated from immunized MyD88(-/-) mice suppressed the induction of active EAE in WT recipients. Moreover, the absence of IL-10 production in MyD88/IL-10 double-knockout mice rendered the mice susceptible to adoptive transfer of EAE. Thus, MyD88 signaling appears to be a key factor in determining the cytokine phenotype of T cells involved in autoimmune inflammation and regulation.
Collapse
Affiliation(s)
- Shmuel J Cohen
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine, Jerusalem, Israel
| | | | | |
Collapse
|
22
|
Jin D, Zhang L, Zheng J, Zhao Y. The inflammatory Th 17 subset in immunity against self and non-self antigens. Autoimmunity 2009; 41:154-62. [DOI: 10.1080/08916930701776605] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
23
|
Chan MMP, Cheung BKW, Li JCB, Chan LLY, Lau ASY. A role for glycogen synthase kinase-3 in antagonizing mycobacterial immune evasion by negatively regulating IL-10 induction. J Leukoc Biol 2009; 86:283-91. [PMID: 19401395 DOI: 10.1189/jlb.0708442] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mtb dysregulates monocyte/macrophage functions to produce a large amount of the immunosuppressive cytokine IL-10. An important function of IL-10 in promoting Mtb survival is the suppression of antigen presentation of monocytes/macrophages to T cells. This dampens the host immune responses and provides an opportunity for immune evasion. GSK3 has been shown to control the balance between pro- and anti-inflammatory cytokine productions. Here, we investigated whether GSK3 regulates IL-10 expression and mediates a protective role upon live mycobacterial challenge using BCG as a model. Our results showed that BCG increased Akt phosphorylation and inhibited GSK3 activity, resulting in increased IL-10 production. We confirmed further that suppression of GSK3 activities by a specific chemical inhibitor strongly enhanced BCG-induced IL-10 production. We also showed that IL-10 secreted by BCG-infected human PBMo was a major suppressor of subsequent IFN-gamma production by PBMC and HLA-DR expression on PBMo in response to BCG. Neutralization of PBMo-secreted IL-10 by anti-IL-10 antibodies restored the IFN-gamma production and HLA-DR surface expression. Taken together, GSK3 negatively regulates mycobacteria-induced IL-10 production in human PBMo. The kinase may play a role in restoring IFN-gamma secretions and subsequent antigen presentation in response to mycobacterial infection. In conclusion, our results suggest a significant role for GSK3 in guarding against mycobacterial evasion of immunity via IL-10 induction in the host.
Collapse
Affiliation(s)
- Mabel M P Chan
- Department of Pediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
24
|
Abstract
Activation of immune cells to mediate an immune response is often triggered by potential 'danger' or 'stress' stimuli that the organism receives. Within the mitogen-activated protein kinases (MAPKs) family, the stress-activated protein kinase (SAPK) group was defined as group of kinases that activated by stimuli that cause cell stress. In the immune cells, SAPKs are activated by antigen receptors (B- or T-cell receptors), Toll-like receptors, cytokine receptors, and physical-chemical changes in the environment among other stimuli. The SAPKs are established to be important mediators of intracellular signaling during adaptive and innate immune responses. Here we summarize what is currently known about the role of two sub-groups of SAPKs - c-Jun NH(2)-terminal kinase and p38 MAPK-in the function of specific components of the immune system and the overall contribution to the immune response.
Collapse
Affiliation(s)
- Mercedes Rincón
- Immunology Program, Department of Medicine, University of Vermont, Burlington, VT 05405, USA.
| | | |
Collapse
|
25
|
Feng H, Yamaki K, Takano H, Inoue K, Yanagisawa R, Yoshino S. Effect of sinomenine on collagen-induced arthritis in mice. Autoimmunity 2008; 40:532-9. [PMID: 17966044 DOI: 10.1080/08916930701615159] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The present study was designed to investigate the effect of sinomenine (SIN), an alkaloid extracted from sinomenium acutum on collagen-induced arthritis (CIA) in mice. For this investigation, mice were s.c. immunized with type II collagen (CII) emulsified with complete Freund's adjuvant (day 0). Varying doses of SIN were orally administered daily commencing on day 0 daily over a period of 55 days. The severity of arthritis was evaluated according to clinical score, the effect of SIN on immune responses were determined by measurement of proliferative responses of spleen cells, antibody levels in serum and cytokine assays. Anti-CII IgG2a and IFN-gamma were measured as indicators of Th1 immune responses and anti-CII IgG1, IgE and IL-5 as those of Th2 responses. IL-10 and TGF-beta were measured as indicators of T cell regulator responses. The results showed that treatment with SIN was followed by decreases in the incidence and severity of CIA, anti-CII IgG and the antigen-specific splenocyte proliferation. Production of all isotypes of antibodies including anti-CII IgG2a, IgG1 and IgE as well as secretion of cytokines such as IFN-gamma and IL-5 were suppressed by SIN. In addition, SIN enhanced the secretion of TGF-beta while it had no obvious effect on production of IL-10. These results suggest that the anti-arthritic effect of SIN may be related to the suppression of both Th1 and Th2 immune responses. TGF-beta may at least in part contribute to the suppression of Th1 as well as Th2 immune responses.
Collapse
Affiliation(s)
- H Feng
- Department of Pharmacology, Kobe Pharmaceutical University, Kobe, Hyogo, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Detrick B, Lee MT, Chin MS, Hooper LC, Chan CC, Hooks JJ. Experimental coronavirus retinopathy (ECOR): retinal degeneration susceptible mice have an augmented interferon and chemokine (CXCL9, CXCL10) response early after virus infection. J Neuroimmunol 2008; 193:28-37. [PMID: 18037505 PMCID: PMC2562577 DOI: 10.1016/j.jneuroim.2007.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 09/05/2007] [Accepted: 09/21/2007] [Indexed: 11/19/2022]
Abstract
Mouse hepatitis virus induces a biphasic disease in BALB/c mice that consists of an acute retinitis followed by progression to a chronic retinal degeneration with autoimmune reactivity. Retinal degeneration resistant CD-1 mice do not develop the late phase. What host factors contribute to the distinct responses to the virus are unknown. Herein, we show that IFN-alpha, IFN-beta and IFN-gamma act in concert as part of the innate immune response to the retinal infection. At day 2, high serum levels of IFN-gamma, CXCL9 and CXCL10, were detected in BALB/c mice. Moreover, elevated levels of CXCL9 and CXCL10 gene expression were detected in retinal tissue. Although IFN-gamma and the chemokines were detected in CD-1 mice, they were at significantly lower levels compared to BALB/c mice. These augmented innate responses observed correlated with the development of autoimmune reactivity and retinal degeneration and thus may contribute to the pathogenic processes.
Collapse
Affiliation(s)
- Barbara Detrick
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD 21287-7065, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Azuma YT, Hagi K, Shintani N, Kuwamura M, Nakajima H, Hashimoto H, Baba A, Takeuchi T. PACAP provides colonic protection against dextran sodium sulfate induced colitis. J Cell Physiol 2008; 216:111-9. [DOI: 10.1002/jcp.21381] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
28
|
c-Jun amino terminal kinase 1 deficient mice are protected from streptozotocin-induced islet injury. Biochem Biophys Res Commun 2007; 366:710-6. [PMID: 18082135 DOI: 10.1016/j.bbrc.2007.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 12/03/2007] [Indexed: 11/24/2022]
Abstract
In vitro studies have implicated the c-Jun amino terminal kinase (JNK) in cytokine-induced pancreatic injury leading to a loss of insulin production and hyperglycemia. We examined the role of JNK1 in the multiple low dose streptozotocin (MLD-STZ) model in which islet injury and hyperglycemia are dependent upon T cell immunity and pro-inflammatory cytokines. MLD-STZ in wild type mice induced islet leukocyte infiltration, cytokine production, beta-cell apoptosis, and hyperglycemia. In contrast, Jnk1-/- mice were substantially protected from a loss of insulin producing cells and hyperglycemia in the MLD-STZ model despite a marked islet T cell and macrophage infiltrate. Based upon several lines of evidence, this protection was attributed to a reduction in TNF-alpha production by infiltrating Jnk1-/- macrophages leading to reduced beta-cell apoptosis. In conclusion, JNK1 signaling plays an essential role in macrophage induced beta-cell apoptosis and the development of hyperglycemia in MLD-STZ induced pancreatic injury.
Collapse
|
29
|
Abstract
One principal aim of research in the signal transduction field is to identify targets for therapeutic intervention, in an attempt to modify disease and curtail human suffering. Diseases such as chronic inflammation, atherosclerosis, diabetes and cancer exact a huge toll on health, in physical, social and financial terms. Defective signaling mechanisms are central to their pathogenesis. One candidate signaling molecule that is presently undergoing intense investigation is the c-Jun N-terminal kinase. With roles described in almost all classes of disease, the main questions are what type of inhibitor to use and when exactly to use it during the disease course?
Collapse
Affiliation(s)
- Baljinder Salh
- University of British Columbia, Department of Medicine, Vancouver, BC, Canada.
| |
Collapse
|
30
|
Weston CR, Davis RJ. The JNK signal transduction pathway. Curr Opin Cell Biol 2007; 19:142-9. [PMID: 17303404 DOI: 10.1016/j.ceb.2007.02.001] [Citation(s) in RCA: 833] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 02/05/2007] [Indexed: 11/18/2022]
Abstract
The c-Jun NH(2)-terminal kinases (JNKs) are an evolutionarily conserved sub-group of mitogen-activated protein (MAP) kinases. Recent studies have improved our understanding of the physiological function of the JNK pathway. Roles of novel molecules that participate in the JNK pathway have been defined and new insight into the role of JNK in survival signaling, cell death, cancer and diabetes has been achieved.
Collapse
Affiliation(s)
- Claire R Weston
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|