1
|
Bansod S, Dodhiawala PB, Geng Y, Bulle A, Liu P, Li L, Townsend R, Grierson PM, Held JM, Adhikari H, Lim KH. The TRIM4 E3 ubiquitin ligase degrades TPL2 and is modulated by oncogenic KRAS. Cell Rep 2024; 43:114667. [PMID: 39178114 PMCID: PMC11472288 DOI: 10.1016/j.celrep.2024.114667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/24/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024] Open
Abstract
Loss-of-function mutations in the C terminus of TPL2 kinase promote oncogenesis by impeding its proteasomal degradation, leading to sustained protein expression. However, the degradation mechanism for TPL2 has remained elusive. Through proximity-dependent biotin identification (BioID), we uncovered tripartite motif-containing 4 (TRIM4) as the E3 ligase that binds and degrades TPL2 by polyubiquitination of lysines 415 and 439. The naturally occurring TPL2 mutants R442H and E188K exhibit impaired TRIM4 binding, enhancing their stability. We further discovered that TRIM4 itself is stabilized by another E3 ligase, TRIM21, which in turn is regulated by KRAS. Mutant KRAS recruits RNF185 to degrade TRIM21 and subsequently TRIM4, thereby stabilizing TPL2. In the presence of mutant KRAS, TPL2 phosphorylates and degrades GSK3β, resulting in β-catenin stabilization and activation of the Wnt pathway. These findings elucidate the physiological mechanisms regulating TPL2 and its exploitation by mutant KRAS, underscoring the need to develop TPL2 inhibitors for KRAS-mutant cancers.
Collapse
Affiliation(s)
- Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yutong Geng
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Peng Liu
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Reid Townsend
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason M Held
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hema Adhikari
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
2
|
Guan J, Fan Y, Wang S, Zhou F. Functions of MAP3Ks in antiviral immunity. Immunol Res 2023; 71:814-832. [PMID: 37286768 PMCID: PMC10247270 DOI: 10.1007/s12026-023-09401-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Immune signal transduction is crucial to the body's defense against viral infection. Recognition of pathogen-associated molecular patterns by pattern recognition receptors (PRRs) activates the transcription of interferon regulators and nuclear factor-κB (NF-κB); this promotes the release of interferons and inflammatory factors. Efficient regulation of type I interferon and NF-κB signaling by members of the mitogen-activated protein (MAP) kinase kinase kinase (MAP3K) family plays an important role in antiviral immunity. Elucidating the specific roles of MAP3K activation during viral infection is essential to develop effective antiviral therapies. In this review, we outline the specific regulatory mechanisms of MAP3Ks in antiviral immunity and discuss the feasibility of targeting MAP3Ks for the treatment of virus-induced diseases.
Collapse
Affiliation(s)
- Jizhong Guan
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Yao Fan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shuai Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
3
|
Wu XJ, Gao J, Mu BJ, Yu LM, Wang ZR, Zheng WB, Gao WW, Zhu XQ, Liu Q. Transcriptomic analysis of LMH cells in response to the overexpression of a protein of Eimeria tenella encoded by the locus ETH_00028350. Front Vet Sci 2022; 9:1053701. [DOI: 10.3389/fvets.2022.1053701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
A protein of Eimeria tenella (encoded by the locus ETH_00028350) homologous to Toxoplasma gondii dense granule protein 9, designated as EtHGRA9 hereafter, was reported to be expressed in all life cycle stages of E. tenella. However, no data are currently available regarding its functional properties. In the present study, a recombinant vector harboring a 741 bp gene segment encoding the mature form of EtHGRA9 was constructed and transfected into leghorn male hepatoma (LMH) cells. Then, transcriptomic analysis of the transfected LMH cells was carried out by using a high-throughput RNA-seq technology. The LMH cells overexpressing EtHGRA9 was validated by means of Western blotting as well as indirect immunofluorescence staining. The results demonstrated that the expression of 547 genes (275 upregulated genes and 272 downregulated genes) was altered by EtHGRA9. The quantitative real-time polymerase chain reaction (qRT-PCR) validation of the ten genes with differential expression between the two groups was consistent with the transcriptome analysis. According to pathway enrichment analysis for the obtained differentially expressed genes, seven pathways were significantly affected by EtHGRA9, such as cytokine-cytokine receptor interaction, MAPK signaling pathway, and protein processing in endoplasmic reticulum. Our data reveal several possible roles of EtHGRA9 in immune or inflammatory responses, which paves the way for a better understanding of the molecular interplay between E. tenella and its host.
Collapse
|
4
|
Sandhu G, Thelma BK. New Druggable Targets for Rheumatoid Arthritis Based on Insights From Synovial Biology. Front Immunol 2022; 13:834247. [PMID: 35265082 PMCID: PMC8899708 DOI: 10.3389/fimmu.2022.834247] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease characterized by chronic inflammation and destruction of multiple small joints which may lead to systemic complications. Altered immunity via pathogenic autoantibodies pre-date clinical symptom development by several years. Incompletely understood range of mechanisms trigger joint-homing, leading to clinically evident articular disease. Advances in therapeutic approaches and understanding pathogenesis have improved prognosis and likely remission. However, partial/non-response to conventional and biologic therapies witnessed in a subset of patients highlights the need for new therapeutics. It is now evident that joint disease chronicity stems from recalcitrant inflammatory synovial environment, majorly maintained by epigenetically and metabolically reprogrammed synoviocytes. Therefore, interference with effector functions of activated cell types seems a rational strategy to reinstate synovial homeostasis and complement existing anti-inflammatory interventions to mitigate chronic RA. Presenting this newer aspect of fibroblast-like synoviocytes and myeloid cells underlying the altered synovial biology in RA and its potential for identification of new druggable targets is attempted in this review. Major leads from i) molecular insights of pathogenic cell types from hypothesis free OMICS approaches; ii) hierarchy of their dysregulated signaling pathways; and iii) knowledge of druggability of molecular nodes in these pathways are highlighted. Development of such synovial biology-directed therapeutics hold promise for an enriched drug repertoire for RA.
Collapse
Affiliation(s)
| | - B. K. Thelma
- Department of Genetics, University of Delhi, New Delhi, India
| |
Collapse
|
5
|
Foot-and-Mouth Disease Virus Structural Protein VP1 Destroys the Stability of TPL2 Trimer by Degradation TPL2 to Evade Host Antiviral Immunity. J Virol 2021; 95:JVI.02149-20. [PMID: 33361430 PMCID: PMC8092693 DOI: 10.1128/jvi.02149-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Tumor progression locus 2 (TPL2) is a serine/threonine kinase that belongs to the mitogen-activated protein 3 kinase (MAP3K) family, and it plays an important role in pathogen infection. The trimer complex of TPL2, p105, and ABIN2 is essential for maintenance of TPL2 steady-state levels and host cell response to pathogens. Foot-and-mouth disease virus (FMDV) is a positive-strand RNA virus of the family Picornaviridae that encodes proteins capable of antagonizing host immune responses to achieve infection. The VP1 protein of FMDV is a multifunctional protein that can bind host cells and induce an immune response as well as cell apoptosis. However, the role and mechanisms of TPL2 in FMDV infection remain unknown. Here, we determined that FMDV infection could inhibit TPL2, p105, and ABIN2 at the transcription and protein levels, while VP1 could only inhibit TPL2, p105 and ABIN2 at protein level. TPL2 inhibited the replication of FMDV in vivo and in vitro, the 268 to 283 amino-acid region in the TPL2 kinase domain was essential for interaction with VP1. Moreover, VP1 promoted K48-linked polyubiquitination of TPL2 and degraded TPL2 by the proteasome pathway. However, VP1-induced degradation of p105 and ABIN2 was independent of proteasome, autophagy, lysosome, and caspase-dependent pathways. Further studies showed that VP1 destroyed the stability of the TPL2-p105-ABIN2 complex. Taken together, these results revealed that VP1 antagonized TPL2-meditated antivirus activity by degrading TPL2 and destroying its complex. These findings may contribute to understand FMDV-host interactions and improve development of a novel vaccine to prevent FMDV infection.Importance Virus-host interactions are critical for virus infection. This study was the first to demonstrate the antiviral effect of host TPL2 during FMDV replication by increasing production of interferons and antiviral cytokines. Both FMDV and VP1 protein can reduce host TPL2, ABIN2 and p105 to destroy TPL2-p105-ABIN2 trimer complex. VP1 interacted with TPL2 and degrade TPL2 via proteasome pathway to repress TPL2-mediated antivirus activity. This study provided new insights into FMDV immune evasion mechanisms, elucidating new informations regarding FMDV counteraction of host antivirus activity.
Collapse
|
6
|
Njunge LW, Estania AP, Guo Y, Liu W, Yang L. Tumor progression locus 2 (TPL2) in tumor-promoting Inflammation, Tumorigenesis and Tumor Immunity. Am J Cancer Res 2020; 10:8343-8364. [PMID: 32724474 PMCID: PMC7381748 DOI: 10.7150/thno.45848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Over the years, tumor progression locus 2 (TPL2) has been identified as an essential modulator of immune responses that conveys inflammatory signals to downstream effectors, subsequently modulating the generation and function of inflammatory cells. TPL2 is also differentially expressed and activated in several cancers, where it is associated with increased inflammation, malignant transformation, angiogenesis, metastasis, poor prognosis and therapy resistance. However, the relationship between TPL2-driven inflammation, tumorigenesis and tumor immunity has not been addressed. Here, we reconcile the function of TPL2-driven inflammation to oncogenic functions such as inflammation, proliferation, apoptosis resistance, angiogenesis, metastasis, immunosuppression and immune evasion. We also address the controversies reported on TPL2 function in tumor-promoting inflammation and tumorigenesis, and highlight the potential role of the TPL2 adaptor function in regulating the mechanisms leading to pro-tumorigenic inflammation and tumor progression. We discuss the therapeutic implications and limitations of targeting TPL2 for cancer treatment. The ideas presented here provide some new insight into cancer pathophysiology that might contribute to the development of more integrative and specific anti-inflammatory and anti-cancer therapeutics.
Collapse
|
7
|
Webb LV, Ventura S, Ley SC. ABIN-2, of the TPL-2 Signaling Complex, Modulates Mammalian Inflammation. Trends Immunol 2019; 40:799-808. [PMID: 31401161 DOI: 10.1016/j.it.2019.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/19/2022]
Abstract
Mammalian TPL-2 kinase (MAP3K8) mediates Toll-like receptor activation of ERK1/2 and p38α MAP kinases and is critical for regulating immune responses to pathogens. TPL-2 also has an important adaptor function, maintaining stability of associated ABIN-2 ubiquitin-binding protein. Consequently, phenotypes detected in Map3k8-/- mice can be caused by lack of TPL-2, ABIN-2, or both proteins. Recent studies show that increased inflammation of Map3k8-/- mice in allergic airway inflammation and colitis results from reduced ABIN-2 signaling, rather than blocked TPL-2 signaling. However, Map3k8-/- mice have been employed extensively to evaluate the potential of TPL-2 as an anti-inflammatory drug target. We posit that Map3k8D270A/D270A mice, expressing catalytically inactive TPL-2 and physiologic ABIN-2, should be used to evaluate the potential effects of TPL-2 inhibitors in disease.
Collapse
|
8
|
Ventura S, Cano F, Kannan Y, Breyer F, Pattison MJ, Wilson MS, Ley SC. A20-binding inhibitor of NF-κB (ABIN) 2 negatively regulates allergic airway inflammation. J Exp Med 2018; 215:2737-2747. [PMID: 30337469 PMCID: PMC6219730 DOI: 10.1084/jem.20170852] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/20/2018] [Accepted: 10/05/2018] [Indexed: 11/29/2022] Open
Abstract
Airway allergic responses are shown to be inhibited by binding of ABIN-2 to A20, a key negative regulator of inflammation. In contrast, the catalytic activity of the ABIN-2–associated kinase TPL-2 does not regulate airway allergic responses, an important consideration for the development of TPL-2 inhibitors to treat inflammatory diseases. TPL-2 MAP 3-kinase promotes inflammation in numerous mouse disease models and is an attractive anti-inflammatory drug target. However, TPL-2–deficient (Map3k8−/−) mice develop exacerbated allergic airway inflammation to house dust mite (HDM) compared with wild type controls. Here, we show that Map3k8D270A/D270A mice expressing kinase dead TPL-2 had an unaltered response to HDM, indicating that the severe airway inflammation observed in Map3k8−/− mice is not due to blockade of TPL-2 signaling and rather reflects a TPL-2 adaptor function. Severe allergic inflammation in TPL-2–deficient mice was likely due to reduced levels of ABIN-2 (TNIP2), whose stability depends on TPL-2 expression. Tnip2E256K knock-in mutation, which reduced ABIN-2 binding to A20, augmented the HDM-induced airway inflammation, but did not affect TPL-2 expression or signaling. These results identify ABIN-2 as a novel negative regulator of allergic airway responses and importantly indicate that TPL-2 inhibitors would not have unwanted allergic comorbidities.
Collapse
Affiliation(s)
| | | | | | - Felix Breyer
- The Francis Crick Institute, London, England, UK
| | | | - Mark S Wilson
- Immunology Discovery, Genentech Inc., South San Francisco, CA
| | - Steven C Ley
- Department of Medicine, Imperial College London, London, England UK
| |
Collapse
|
9
|
The multifaceted allosteric regulation of Aurora kinase A. Biochem J 2018; 475:2025-2042. [PMID: 29946042 PMCID: PMC6018539 DOI: 10.1042/bcj20170771] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
The protein kinase Aurora A (AurA) is essential for the formation of bipolar mitotic spindles in all eukaryotic organisms. During spindle assembly, AurA is activated through two different pathways operating at centrosomes and on spindle microtubules. Recent studies have revealed that these pathways operate quite differently at the molecular level, activating AurA through multifaceted changes to the structure and dynamics of the kinase domain. These advances provide an intimate atomic-level view of the finely tuned regulatory control operating in protein kinases, revealing mechanisms of allosteric cooperativity that provide graded levels of regulatory control, and a previously unanticipated mechanism for kinase activation by phosphorylation on the activation loop. Here, I review these advances in our understanding of AurA function, and discuss their implications for the use of allosteric small molecule inhibitors to address recently discovered roles of AurA in neuroblastoma, prostate cancer and melanoma.
Collapse
|
10
|
Assaying kinase activity of the TPL-2/NF-κB1 p105/ABIN-2 complex using an optimal peptide substrate. Biochem J 2018; 475:329-340. [PMID: 29229763 PMCID: PMC5763956 DOI: 10.1042/bcj20170579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 11/17/2022]
Abstract
The MKK1/2 kinase tumour progression locus 2 (TPL-2) is critical for the production of tumour necrosis factor alpha (TNFα) in innate immune responses and a potential anti-inflammatory drug target. Several earlier pharmaceutical company screens with the isolated TPL-2 kinase domain have identified small-molecule inhibitors that specifically block TPL-2 signalling in cells, but none of these have progressed to clinical development. We have previously shown that TPL-2 catalytic activity regulates TNF production by macrophages while associated with NF-κB1 p105 and ABIN-2, independently of MKK1/2 phosphorylation via an unknown downstream substrate. In the present study, we used a positional scanning peptide library to determine the optimal substrate specificity of a complex of TPL-2, NF-κB1 p105 and ABIN-2. Using an optimal peptide substrate based on this screen and a high-throughput mass spectrometry assay to monitor kinase activity, we found that the TPL-2 complex has significantly altered sensitivities versus existing ATP-competitive TPL-2 inhibitors than the isolated TPL-2 kinase domain. These results imply that screens with the more physiologically relevant TPL-2/NF-κB1 p105/ABIN-2 complex have the potential to deliver novel TPL-2 chemical series; both ATP-competitive and allosteric inhibitors could emerge with significantly improved prospects for development as anti-inflammatory drugs.
Collapse
|
11
|
Xu D, Matsumoto ML, McKenzie BS, Zarrin AA. TPL2 kinase action and control of inflammation. Pharmacol Res 2017; 129:188-193. [PMID: 29183769 DOI: 10.1016/j.phrs.2017.11.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
Tumor progression locus 2 (TPL2, also known as COT or MAP3K8) is a mitogen-activated protein kinase kinase (MAP3K) activated downstream of TNFαR, IL1R, TLR, CD40, IL17R, and some GPCRs. TPL2 regulates the MEK1/2 and ERK1/2 pathways to regulate a cascade of inflammatory responses. In parallel to this, TPL2 also activates p38α and p38δ to drive the production of various inflammatory mediators in neutrophils. We discuss the implications of this finding in the context of various inflammatory diseases.
Collapse
Affiliation(s)
- Daqi Xu
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Marissa L Matsumoto
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Brent S McKenzie
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Ali A Zarrin
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
12
|
Genick CC, Wright SK. Biophysics: for HTS hit validation, chemical lead optimization, and beyond. Expert Opin Drug Discov 2017; 12:897-907. [DOI: 10.1080/17460441.2017.1349096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Christine C. Genick
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Chemical Biology and Therapeutics, Protein Sciences, Basel, Switzerland
- Protein Sciences, Research Parkway Meriden, Cambridge, MA, USA
| | - S. Kirk Wright
- Protein Sciences, Research Parkway Meriden, Cambridge, MA, USA
- Protein Sciences, Novartis Pharma AG, Novartis Institutes for BioMedical Research, Chemical Biology and Therapeutics, Cambridge, MA, USA
| |
Collapse
|
13
|
Glatthar R, Stojanovic A, Troxler T, Mattes H, Möbitz H, Beerli R, Blanz J, Gassmann E, Drückes P, Fendrich G, Gutmann S, Martiny-Baron G, Spence F, Hornfeld J, Peel JE, Sparrer H. Discovery of Imidazoquinolines as a Novel Class of Potent, Selective, and in Vivo Efficacious Cancer Osaka Thyroid (COT) Kinase Inhibitors. J Med Chem 2016; 59:7544-60. [DOI: 10.1021/acs.jmedchem.6b00598] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ralf Glatthar
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Aleksandar Stojanovic
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Thomas Troxler
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Henri Mattes
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Henrik Möbitz
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Rene Beerli
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Joachim Blanz
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Ernst Gassmann
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Peter Drückes
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Gabriele Fendrich
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Sascha Gutmann
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Georg Martiny-Baron
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Fiona Spence
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jeff Hornfeld
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - John Edmonson Peel
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Helmut Sparrer
- Global Discovery Chemistry, ‡Analytical Sciences, §Center for Proteomic
Chemistry, ∥Preclinical Safety, and ⊥Autoimmunity
Transplantation Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
14
|
TLR and TNF-R1 activation of the MKK3/MKK6-p38α axis in macrophages is mediated by TPL-2 kinase. Biochem J 2016; 473:2845-61. [PMID: 27402796 PMCID: PMC5095906 DOI: 10.1042/bcj20160502] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
Abstract
Previous studies suggested that Toll-like receptor (TLR) stimulation of the p38α MAP kinase (MAPK) is mediated by transforming growth factor-β-activated kinase 1 (TAK1) activation of MAPK kinases, MKK3, MKK4 and MKK6. We used quantitative mass spectrometry to monitor tumour progression locus 2 (TPL-2)-dependent protein phosphorylation following TLR4 stimulation with lipopolysaccharide, comparing macrophages from wild-type mice and Map3k8(D270A/D270A) mice expressing catalytically inactive TPL-2 (MAP3K8). In addition to the established TPL-2 substrates MKK1/2, TPL-2 kinase activity was required to phosphorylate the activation loops of MKK3/6, but not of MKK4. MKK3/6 activation required IκB kinase (IKK) phosphorylation of the TPL-2 binding partner nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB1) p105, similar to MKK1/2 activation. Tumour necrosis factor (TNF) stimulation of MKK3/6 phosphorylation was similarly dependent on TPL-2 catalytic activity and IKK phosphorylation of NF-κB1 p105. Owing to redundancy of MKK3/6 with MKK4, Map3k8(D270A) mutation only fractionally decreased lipopolysaccharide activation of p38α. TNF activation of p38α, which is mediated predominantly via MKK3/6, was substantially reduced. TPL-2 catalytic activity was also required for MKK3/6 and p38α activation following macrophage stimulation with Mycobacterium tuberculosis and Listeria monocytogenes Our experiments demonstrate that the IKK/NF-κB1 p105/TPL-2 signalling pathway, downstream of TAK1, regulates MKK3/6 and p38α activation in macrophages in inflammation.
Collapse
|