1
|
Jiang Z, Su W, Yang M, Fu J, Gong T, Li W, Wen C, Wang X, Wang F, Jin M, Wang Y, Lu Z. Integrated multi-omics reveals the Bacillus amyloliquefaciens BA40 against Clostridium perfringens infection in weaned piglets. J Adv Res 2025:S2090-1232(25)00052-9. [PMID: 39855299 DOI: 10.1016/j.jare.2025.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION Clostridium perfringens (C. perfringens) can cause necrotic enteritis and higher mortality rates in piglets, by impairing the intestinal barrier function. Bacillus amyloliquefaciens 40 (BA40) has showed potential ability to reduce C. perfringens infections, but the mechanisms responsible for its effectiveness remain unclear. OBJECTIVES This study aims to evaluate the impact of BA40 on inflammation induced by C. perfringens and to explain the mechanisms underlying its therapeutic effects. We aim to show how BA40 can bolster piglet health by strengthening the intestinal barrier and regulating immune responses. METHODS We used piglets and cellular models, alongside microbiomics, metabolomic, and transcriptomic analyses, to investigate BA40's impact on C. perfringens-induced inflammation. A model of C. perfringens infection was constructed using piglets and cells to investigate the effect of BA40 on its phenotype. Microbiomics, metabolomics, and transcriptomics analyses were subsequently used to investigate the mechanisms of protection and immune response to BA40 on the intestinal barrier of piglets. RESULTS Our study revealed significant improvements in piglet health following BA40 administration. Notably, BA40 strengthened the intestinal mucosal barrier and mitigated the inflammatory response triggered by C. perfringens BA40 decreased harmful bacteria and increased beneficial bacteria. Metabolite profiles improved, showing a reduction in harmful substances. Transscriptomics analysis indicated BA40's role in TNF/NF-κB signaling pathway, hinting at its ability to regulate immune responses and reduce intestinal inflammation. Cellular assays further confirmed BA40's capacity to diminish inflammatory cytokine release and encourage the differentiation of anti-inflammatory macrophages. CONCLUSION Datasets from the present study demonstrate that BA40 modulates gut microbes and metabolites, inhibits inflammation-related signaling pathways, and maintains gut barrier function. Our findings not only deepen our understanding of the therapeutic capacity of BA40 but also provide a theoretical foundation for the development of probiotics and alternative therapies aimed at improving piglet gut health.
Collapse
Affiliation(s)
- Zipeng Jiang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Weifa Su
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Mingzhi Yang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Tao Gong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Wentao Li
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Chaoyue Wen
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Xinxia Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Fengqin Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Mingliang Jin
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China
| | - Yizhen Wang
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China.
| | - Zeqing Lu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Key Laboratory of Animal Nutrition and Feed, Ministry of Agricultural and Rural Affairs, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; National Engineering Research Center of Green Feeds and Healthy Livestock Industry, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China; College of Animal Science, Institute of Feed Science, Zhejiang University, 866 Yuhang Tang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
2
|
Zenke K, Sugimoto R, Watanabe S, Muroi M. NF-κB p105-mediated nuclear translocation of ERK is required for full activation of IFNγ-induced iNOS expression. Cell Signal 2024; 124:111424. [PMID: 39304100 DOI: 10.1016/j.cellsig.2024.111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/30/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Inducible nitric oxidase (iNOS) encoded by Nos2 is a representative IFNγ-inducible effector molecule that plays an important role in both innate and adaptive immunity. In the present study, we demonstrated that full-length NF-κB p105 (p105), which is a precursor of NF-κB p50 (p50), is required for full activation of IFNγ-induced iNOS expression in the RAW264.7 mouse macrophage cell line. In comparison to wild-type (WT) RAW264.7 cells, p105 KO RAW264.7 (p105 KO) cells completely lost IFNγ-induced iNOS expression. Despite the limited effect of exogenous expression of p50 in p105 KO cells on IFNγ-induced Nos2 promoter activity, p105 expression fully restored IFNγ-induced Nos2 promoter activity to a level comparable to that of WT cells, suggesting an important role for full-length p105 in IFNγ-induced iNOS expression. While the expression and phosphorylation of JAK1 and STAT1 were rather enhanced in p105 KO cells, the phosphorylation of c-Jun downstream of MAPK signaling was decreased. IFNγ-induced phosphorylation of ERK, a kinase for IFNγ-induced c-Jun phosphorylation, was not significantly reduced in p105 KO cells, although the nuclear activity of ERK was significantly decreased due to its reduced translocation to the nucleus. Expression of iNOS, nuclear translocation of ERK, and phosphorylation of c-Jun were restored by stable supplementation of p105 in p105 KO cells. These results suggest that p105 is required for the nuclear translocation of ERK and the subsequent phosphorylation of c-Jun, which are necessary for full activation of IFNγ-induced iNOS expression. Reduced nuclear translocation of ERK in p105 KO cells was also observed in the activation of ERK following serum starvation, further suggesting that the involvement of p105 in ERK nuclear translocation is not limited to IFNγ-stimulated cells but is a more general function of p105.
Collapse
Affiliation(s)
- Kosuke Zenke
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Rino Sugimoto
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Sachiko Watanabe
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Masashi Muroi
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| |
Collapse
|
3
|
Herb M, Schatz V, Hadrian K, Hos D, Holoborodko B, Jantsch J, Brigo N. Macrophage variants in laboratory research: most are well done, but some are RAW. Front Cell Infect Microbiol 2024; 14:1457323. [PMID: 39445217 PMCID: PMC11496307 DOI: 10.3389/fcimb.2024.1457323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages play a pivotal role in the innate immune response. While their most characteristic function is phagocytosis, it is important not to solely characterize macrophages by this activity. Their crucial roles in body development, homeostasis, repair, and immune responses against pathogens necessitate a broader understanding. Macrophages exhibit remarkable plasticity, allowing them to modify their functional characteristics in response to the tissue microenvironment (tissue type, presence of pathogens or inflammation, and specific signals from neighboring cells) swiftly. While there is no single defined "macrophage" entity, there is a diverse array of macrophage types because macrophage ontogeny involves the differentiation of progenitor cells into tissue-resident macrophages, as well as the recruitment and differentiation of circulating monocytes in response to tissue-specific cues. In addition, macrophages continuously sense and respond to environmental cues and tissue conditions, adjusting their functional and metabolic states accordingly. Consequently, it is of paramount importance to comprehend the heterogeneous origins and functions of macrophages employed in in vitro studies, as each available in vitro macrophage model is associated with specific sets of strengths and limitations. This review centers its attention on a comprehensive comparison between immortalized mouse macrophage cell lines and primary mouse macrophages. It provides a detailed analysis of the strengths and weaknesses inherent in these in vitro models. Finally, it explores the subtle distinctions between diverse macrophage cell lines, offering insights into numerous factors beyond the model type that can profoundly influence macrophage function.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Valentin Schatz
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bohdan Holoborodko
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Natascha Brigo
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Ni R, Jiang L, Zhang C, Liu M, Luo Y, Hu Z, Mou X, Zhu Y. Biologic Mechanisms of Macrophage Phenotypes Responding to Infection and the Novel Therapies to Moderate Inflammation. Int J Mol Sci 2023; 24:ijms24098358. [PMID: 37176064 PMCID: PMC10179618 DOI: 10.3390/ijms24098358] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Pro-inflammatory and anti-inflammatory types are the main phenotypes of the macrophage, which are commonly notified as M1 and M2, respectively. The alteration of macrophage phenotypes and the progression of inflammation are intimately associated; both phenotypes usually coexist throughout the whole inflammation stage, involving the transduction of intracellular signals and the secretion of extracellular cytokines. This paper aims to address the interaction of macrophages and surrounding cells and tissues with inflammation-related diseases and clarify the crosstalk of signal pathways relevant to the phenotypic metamorphosis of macrophages. On these bases, some novel therapeutic methods are proposed for regulating inflammation through monitoring the transition of macrophage phenotypes so as to prevent the negative effects of antibiotic drugs utilized in the long term in the clinic. This information will be quite beneficial for the diagnosis and treatment of inflammation-related diseases like pneumonia and other disorders involving macrophages.
Collapse
Affiliation(s)
- Renhao Ni
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Lingjing Jiang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Chaohai Zhang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Mujie Liu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yang Luo
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zeming Hu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Xianbo Mou
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
5
|
Gutierrez AH, Mazariegos MS, Alemany S, Nevzorova YA, Cubero FJ, Sanz-García C. Tumor progression locus 2 (TPL2): A Cot-plicated progression from inflammation to chronic liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166660. [PMID: 36764206 DOI: 10.1016/j.bbadis.2023.166660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
The cytoplasmic protein tumor progression locus 2 (TPL2), also known as cancer Osaka thyroid (Cot), or MAP3K8, is thought to have a significant role in a variety of cancers and illnesses and it is a key component in the activation pathway for the expression of inflammatory mediators. Despite the tight connection between inflammation and TPL2, its function has not been extensively studied in chronic liver disease (CLD), a major cause of morbidity and mortality worldwide. Here, we analyze more in detail the significance of TPL2 in CLD to shed light on the pathological and molecular transduction pattern of TPL2 during the progression of CLD. This might result in important advancements and enable progress in the diagnosis and treatment of CLD.
Collapse
Affiliation(s)
- Alejandro H Gutierrez
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Marina S Mazariegos
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Susana Alemany
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, 35001 Las Palmas, Spain
| | - Yulia A Nevzorova
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Carlos Sanz-García
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain.
| |
Collapse
|
6
|
Vibrio cholerae Porin OmpU Activates Dendritic Cells via TLR2 and the NLRP3 Inflammasome. Infect Immun 2023; 91:e0033222. [PMID: 36794951 PMCID: PMC9933687 DOI: 10.1128/iai.00332-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OmpU is one of the major porins of Vibrio cholerae, a Gram-negative human pathogen. Previously, we showed that OmpU stimulates host monocytes and macrophages and induces the production of proinflammatory mediators via activation of the Toll-like receptor 1/2 (TLR1/2)-MyD88-dependent pathways. In the present study, we show that OmpU activates murine dendritic cells (DCs) via activation of the TLR2-mediated pathway and the NLRP3 inflammasome, leading to the production of proinflammatory cytokines and DC maturation. Our data reveal that although TLR2 plays an important role in providing both priming and the activation signal for the NLRP3 inflammasome in OmpU-activated DCs, OmpU is capable of activating the NLRP3 inflammasome, even in the absence of TLR2, if a priming signal is given. Furthermore, we show that the OmpU-mediated interleukin-1β (IL-1β) production in DCs depends on calcium flux and mitochondrial reactive oxygen species (mitoROS) generation. Interestingly, both OmpU translocation to the mitochondria of DCs as well as calcium signaling contribute to mitoROS production and prompt NLRP3 inflammasome activation. We also demonstrate that OmpU induces downstream signaling via activation of phosphoinositide-3-kinase (PI3K)-AKT, protein kinase C (PKC), mitogen-activated protein kinases (MAPKs), and transcription factor NF-κB. Furthermore, our data reveal that OmpU-mediated activation of TLR2 induces signaling via PKC, MAPKs p38 and extracellular signal-regulated kinase (ERK), and transcription factor NF-κB; however, PI3K and MAPK Jun N-terminal protein kinase (JNK) are activated in TLR2 independent manner.
Collapse
|
7
|
Nagao T, Yoshifuji K, Sadato D, Motomura Y, Saito M, Yamamoto K, Yamamoto K, Nogami A. Establishment and characterization of a new activated B-cell-like DLBCL cell line, TMD12. Exp Hematol 2022; 116:37-49. [PMID: 36191884 DOI: 10.1016/j.exphem.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 12/29/2022]
Abstract
We report the establishment of a novel activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) cell line, designated as TMD12, from a patient with highly refractory DLBCL. ABC-DLBCL is a subtype with a relatively unfavorable prognosis that was originally categorized using gene expression profiling according to its cell of origin. TMD12 cells were isolated from the pleural effusion of the patient at relapse and passaged continuously in vitro for >4 years. The cells displayed cluster of differentiation (CD)19, CD20, CD22, CD38, human leukocyte antigen-DR isotype, and κ positivity and CD5, CD10, CD23, and λ negativity, as detected using flow cytometric analysis. The chromosomal karyotypic analysis, including the spectral karyotyping method, confirmed t(1;19)(q21:q13.1), del(6q23), gain of chromosome 18, and other abnormalities. Mutation analyses, including whole-exome sequencing, revealed that TMD12 cells harbored mutations in MYD88 and CD79B, indicating an ABC subtype. TMD12 cells exhibited chronic active B-cell receptor signaling and constitutive activation of the nuclear factor κB pathway, which is typically associated with sensitivity to a specific Bruton tyrosine kinase inhibitor, ibrutinib. Intriguingly, TMD12 cells displayed moderate resistance to ibrutinib and lacked activation of Janus kinase/signal transducers and activators of transcription 3 signaling, another hallmark of this DLBCL subtype. Treatment with an inhibitor against tumor progression locus 2 (TPL2), a multifunctional intracellular kinase that is activated particularly downstream of Toll-like receptors or MYD88 and IκB kinase α/β (IKKα/β), suppressed the proliferation of TMD12 cells, implying the possible involvement of the TPL2-p105 pathway in the tumorigenesis of ABC-DLBCL. Because only a limited number of ABC-DLBCL cell lines are currently available, TMD12 cells might provide a useful tool in the search for novel druggable targets for this intractable lymphoma.
Collapse
Affiliation(s)
- Toshikage Nagao
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Kota Yoshifuji
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daichi Sadato
- Clinical Research Support Center, Tokyo Metropolitan Center and Infection Disease Center, Komagome Hospital, Tokyo, Japan
| | - Yotaro Motomura
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Makiko Saito
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kurara Yamamoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Ayako Nogami
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Hou Y, Wei D, Bossila EA, Zhang Z, Li S, Bao J, Xu H, Zhang L, Zhao Y. FABP5 Deficiency Impaired Macrophage Inflammation by Regulating AMPK/NF-κB Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2181-2191. [PMID: 36426981 DOI: 10.4049/jimmunol.2200182] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022]
Abstract
Fatty acid binding protein 5 (FABP5) is mainly involved in the uptake, transport, and metabolism of fatty acid in the cytoplasm, and its role in immune cells has been recognized in recent years. However, the role of FABP5 in macrophage inflammation and its underlying mechanisms were not fully addressed. In our study, the acute liver injury and sepsis mouse models were induced by i.p. injection of LPS and cecal contents, respectively. Oleic acid (0.6 g/kg) was injected four times by intragastric administration every week, and this lasted for 1 wk before the LPS or cecal content challenge. We found that myeloid-specific deletion of FABP5 mitigated LPS-induced acute liver injury with reduced mortality of mice, histological liver damage, alanine aminotransferase, and proinflammatory factor levels. Metabolic analysis showed that FABP5 deletion increased the intracellular unsaturated fatty acids, especially oleic acid, in LPS-induced macrophages. The addition of oleic acid also decreased LPS-stimulated macrophage inflammation in vitro and reduced acute liver injury in LPS-induced or cecal content-induced sepsis mice. RNA-sequencing and molecular mechanism studies showed that FABP5 deletion or oleic acid supplementation increased the AMP/ATP ratio and AMP-activated protein kinase (AMPK) activation and inhibited the NF-κB pathway during the inflammatory response to LPS stimulation of macrophages. Inhibiting AMPK activation or expression by chemical or genetic approaches significantly rescued the decreased NF-κB signaling pathway and inflammatory response in LPS-treated FABP5-knockout macrophages. Our present study indicated that inhibiting FABP5 or supplementation of oleic acid might be used for the treatment of sepsis-caused acute liver injury.
Collapse
Affiliation(s)
- Yangxiao Hou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dong Wei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Elhusseny A Bossila
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Biotechnology Department, Faculty of Agriculture Al-Azhar University, Cairo, Egypt
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sihong Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Bao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huawen Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; and
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regeneration, Beijing, China
| |
Collapse
|
9
|
Huang Z, Chen Y, Yang C, Ma B, Guo S, Zhang J, Chen N, Umar T, Yin B, Deng G. Enhanced expression of miR-26a ameliorates lipopolysaccharide-induced endometritis by targeting MAP3K8 to inactivate MAPK signaling pathway. J Reprod Immunol 2022; 154:103751. [PMID: 36252394 DOI: 10.1016/j.jri.2022.103751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
Endometritis is a severe postpartum inflammatory disease that puts cows' reproductive health at risk and causes the dairy industry to suffer significant financial losses. The present study aimed to investigate the regulatory role of miR‑26a in LPS‑induced bovine endometrial epithelial cells (bEECs) and the implication for endometritis. Here, we found inflammatory cell infiltration and destruction of endometrial structure in cow uterus, and dramatic increase in myeloperoxidase (MPO) activity and upregulation of pro-inflammatory cytokines (IL-1β, TNF-α, and IL-6) in endometritis. Meanwhile, miR-26a was down-regulated, but MAP3K8 was increased in the uterine tissue of endometritis. Similarly, the expression of miR-26a was significantly decreased in LPS-stimulated bEECs, while MAP3K8 was risen. In addition, we further verified that MAP3K8 was a target of miR-26a by dual-luciferase reporter assay. Under LPS stress, over-expressing miR-26a markedly decreased MAP3K8 expression levels, along with the reduced expression of inflammatory factors, such as IL-1β, TNF-α and IL-6, whereas this effect was countered by the inhibition of miR-26a. Furthermore, we demonstrated that miR-26a overexpression prevented the MAPK pathway from being activated by targeting MAP3K8. Then we carried out experiments in LPS-stimulated mice uterus to expound that MAP3K8 was essential in endometritis development, which further confirmed the reliability of the above results. In conclusion, overexpression of miR-26a effectively inhibited the expression of MAP3K8 in LPS-induced bEECs and thereby partially suppressed the activation of MAPK signaling pathway. miR-26a and MAP3K8 may be a promising biomarker and therapeutic target for dairy cow endometritis.
Collapse
Affiliation(s)
- Zhi Huang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Cheng Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Bin Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Jinxin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Nuoer Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Talha Umar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Baoyi Yin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
10
|
Li W, Jin K, Luo J, Xu W, Wu Y, Zhou J, Wang Y, Xu R, Jiao L, Wang T, Yang G. NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front Cardiovasc Med 2022; 9:988266. [PMID: 36204587 PMCID: PMC9530249 DOI: 10.3389/fcvm.2022.988266] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a common cardiovascular disease with complex pathogenesis, in which multiple pathways and their interweaving regulatory mechanism remain unclear. The primary transcription factor NF-κB plays a critical role in AS via modulating the expression of a series of inflammatory mediators under various stimuli such as cytokines, microbial antigens, and intracellular stresses. Endoplasmic reticulum (ER) stress, caused by the disrupted synthesis and secretion of protein, links inflammation, metabolic signals, and other cellular processes via the unfolded protein response (UPR). Both NF-κB and ER stress share the intersection regarding their molecular regulation and function and are regarded as critical individual contributors to AS. In this review, we summarize the multiple interactions between NF-κB and ER stress activation, including the UPR, NLRP3 inflammasome, and reactive oxygen species (ROS) generation, which have been ignored in the pathogenesis of AS. Given the multiple links between NF-κB and ER stress, we speculate that the integrated network contributes to the understanding of molecular mechanisms of AS. This review aims to provide an insight into these interactions and their underlying roles in the progression of AS, highlighting potential pharmacological targets against the atherosclerotic inflammatory process.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liqun Jiao,
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Tao Wang,
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Tao Wang,
| |
Collapse
|
11
|
Blair L, Pattison MJ, Chakravarty P, Papoutsopoulou S, Bakiri L, Wagner EF, Smale S, Ley SC. TPL-2 Inhibits IFN-β Expression via an ERK1/2-TCF-FOS Axis in TLR4-Stimulated Macrophages. THE JOURNAL OF IMMUNOLOGY 2022; 208:941-954. [PMID: 35082159 PMCID: PMC9012084 DOI: 10.4049/jimmunol.2100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022]
Abstract
TPL-2 activation of ERK1/2 regulates gene expression in TLR-stimulated macrophages. TPL-2 regulates transcription via ERK1/2 phosphorylation of ternary complex factors. TPL-2 inhibits Ifnb1 transcription via ternary complex factor–induced Fos mRNA expression.
TPL-2 kinase plays an important role in innate immunity, activating ERK1/2 MAPKs in myeloid cells following TLR stimulation. We investigated how TPL-2 controls transcription in TLR4-stimulated mouse macrophages. TPL-2 activation of ERK1/2 regulated expression of genes encoding transcription factors, cytokines, chemokines, and signaling regulators. Bioinformatics analysis of gene clusters most rapidly induced by TPL-2 suggested that their transcription was mediated by the ternary complex factor (TCF) and FOS transcription factor families. Consistently, TPL-2 induced ERK1/2 phosphorylation of the ELK1 TCF and the expression of TCF target genes. Furthermore, transcriptomic analysis of TCF-deficient macrophages demonstrated that TCFs mediate approximately half of the transcriptional output of TPL-2 signaling, partially via induced expression of secondary transcription factors. TPL-2 signaling and TCFs were required for maximal TLR4-induced FOS expression. Comparative analysis of the transcriptome of TLR4-stimulated Fos−/− macrophages indicated that TPL-2 regulated a significant fraction of genes by controlling FOS expression levels. A key function of this ERK1/2-TCF-FOS pathway was to mediate TPL-2 suppression of type I IFN signaling, which is essential for host resistance against intracellular bacterial infection.
Collapse
Affiliation(s)
- Louise Blair
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael J Pattison
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Probir Chakravarty
- Bioinformatics and Biostatistics Technology Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Latifa Bakiri
- Laboratory of Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Laboratory of Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Laboratory of Genes and Disease, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Stephen Smale
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA; and
| | - Steven C Ley
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom;
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Fang C, Wang L, Qiao J, Chang L, He Q, Zhang X, Liu M. Differential regulation of lipopolysaccharide-induced IL-1β and TNF-α production in macrophages by palmitate via modulating TLR4 downstream signaling. Int Immunopharmacol 2021; 103:108456. [PMID: 34923420 DOI: 10.1016/j.intimp.2021.108456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 11/25/2022]
Abstract
Diabetic patients are susceptible to infectious diseases. Bacterial invasion activates immune cells such as macrophages through interaction between LPS and TLR4, and induces the expression of inflammatory mediators, including IL-1β and TNF-α, which play key roles in the elimination of infections. Unregulated overproduction or underproduction of these cytokines has been reported as a major factor in the development of septic shock, immune deficiency, and autoimmunity. Recent studies found that metabolic abnormalities of diabetes, such as hyperglycemia and dyslipidemia, played a major role in modulating the immune response. In this study, we studied the effects of palmitic acid (PA) pretreatment on LPS-induced IL-1β and TNF-α production and LPS-TLR4 signaling in macrophages. Compared with control, PA pretreatment significantly increased LPS-induced TNF-α production and secretion in macrophages. In contrast, LPS-induced IL-1β production and secretion was significantly suppressed by PA pretreatment. PA pretreatment did not affect the expression levels of TLR4 or Myd88, or the endocytosis of TLR4 in macrophages. However, PA pretreatment significantly suppressed the phosphorylation level and nuclear translocation of NF-κB, and the phosphorylation level of ERK1/2, whereas increased the phosphorylation levels of p38 and JNK. The activation of IKK which was upstream of NF-κB and ERK1/2 was attenuated, while the activation of TAK1 which was upstream of JNK and p38 was augmented by PA pretreatment. Inhibitors of NF-κB, MEK1/2, and p38 significantly decreased IL-1β expression, while JNK and p38 pathway inhibitors significantly inhibited TNF-α expression. The differential regulation of LPS-induced TNF-α and IL-1β production by PA was associated with cellular metabolism of PA, because inhibiting metabolism of PA with etomoxir or pretreatment with Br-PA which cannot be metabolized reversed these effects. We also showed that PA treatment increased acetylated IKK level which might contribute to the suppressed activation of IKK. The present study showed that LPS-induced production of TNF-α and IL-1β was regulated by different TLR4 downstream pathways in macrophages. PA differentially affected LPS-induced production of TNF-α and IL-1β in macrophages through differentially modulating these pathways. Further experiments will be needed to determine how these phenomena lead to the impaired immune response in patients with diabetes.
Collapse
Affiliation(s)
- Chunyun Fang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Lixia Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingting Qiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Chang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xiaona Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
13
|
Su C, Rousseau S, Emad A. Identification of transcriptional regulatory network associated with response of host epithelial cells to SARS-CoV-2. Sci Rep 2021; 11:23928. [PMID: 34907210 PMCID: PMC8671548 DOI: 10.1038/s41598-021-03309-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Identification of transcriptional regulatory mechanisms and signaling networks involved in the response of host cells to infection by SARS-CoV-2 is a powerful approach that provides a systems biology view of gene expression programs involved in COVID-19 and may enable the identification of novel therapeutic targets and strategies to mitigate the impact of this disease. In this study, our goal was to identify a transcriptional regulatory network that is associated with gene expression changes between samples infected by SARS-CoV-2 and those that are infected by other respiratory viruses to narrow the results on those enriched or specific to SARS-CoV-2. We combined a series of recently developed computational tools to identify transcriptional regulatory mechanisms involved in the response of epithelial cells to infection by SARS-CoV-2, and particularly regulatory mechanisms that are specific to this virus when compared to other viruses. In addition, using network-guided analyses, we identified kinases associated with this network. The results identified pathways associated with regulation of inflammation (MAPK14) and immunity (BTK, MBX) that may contribute to exacerbate organ damage linked with complications of COVID-19. The regulatory network identified herein reflects a combination of known hits and novel candidate pathways supporting the novel computational pipeline presented herein to quickly narrow down promising avenues of investigation when facing an emerging and novel disease such as COVID-19.
Collapse
Affiliation(s)
- Chen Su
- Department of Electrical and Computer Engineering, McGill University, 755, McConnell Engineering Building, 3480 University Street, Montreal, QC, H3A 0E9, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories at the Research Institute of McGill University Heath Centre (RI-MUHC), McGill University, E M3.2244, 1001 Décarie, Montreal, QC, H4A 3J1, Canada.
- Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, 755, McConnell Engineering Building, 3480 University Street, Montreal, QC, H3A 0E9, Canada.
- The Meakins-Christie Laboratories at the Research Institute of McGill University Heath Centre (RI-MUHC), McGill University, E M3.2244, 1001 Décarie, Montreal, QC, H4A 3J1, Canada.
- Mila, Quebec AI Institute, Montreal, QC, Canada.
| |
Collapse
|
14
|
Latha K, Jamison KF, Watford WT. Tpl2 Ablation Leads to Hypercytokinemia and Excessive Cellular Infiltration to the Lungs During Late Stages of Influenza Infection. Front Immunol 2021; 12:738490. [PMID: 34691044 PMCID: PMC8529111 DOI: 10.3389/fimmu.2021.738490] [Citation(s) in RCA: 4] [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: 07/08/2021] [Accepted: 09/07/2021] [Indexed: 01/22/2023] Open
Abstract
Tumor progression locus 2 (Tpl2) is a serine-threonine kinase known to promote inflammation in response to various pathogen-associated molecular patterns (PAMPs), inflammatory cytokines and G-protein-coupled receptors and consequently aids in host resistance to pathogens. We have recently shown that Tpl2-/- mice succumb to infection with a low-pathogenicity strain of influenza (x31, H3N2) by an unknown mechanism. In this study, we sought to characterize the cytokine and immune cell profile of influenza-infected Tpl2-/- mice to gain insight into its host protective effects. Although Tpl2-/- mice display modestly impaired viral control, no virus was observed in the lungs of Tpl2-/- mice on the day of peak morbidity and mortality suggesting that morbidity is not due to virus cytopathic effects but rather to an overactive antiviral immune response. Indeed, increased levels of interferon-β (IFN-β), the IFN-inducible monocyte chemoattractant protein-1 (MCP-1, CCL2), Macrophage inflammatory protein 1 alpha (MIP-1α; CCL3), MIP-1β (CCL4), RANTES (CCL5), IP-10 (CXCL10) and Interferon-γ (IFN-γ) was observed in the lungs of influenza-infected Tpl2-/- mice at 7 days post infection (dpi). Elevated cytokine and chemokines were accompanied by increased infiltration of the lungs with inflammatory monocytes and neutrophils. Additionally, we noted that increased IFN-β correlated with increased CCL2, CXCL1 and nitric oxide synthase (NOS2) expression in the lungs, which has been associated with severe influenza infections. Bone marrow chimeras with Tpl2 ablation localized to radioresistant cells confirmed that Tpl2 functions, at least in part, within radioresistant cells to limit pro-inflammatory response to viral infection. Collectively, this study suggests that Tpl2 tempers inflammation during influenza infection by constraining the production of interferons and chemokines which are known to promote the recruitment of detrimental inflammatory monocytes and neutrophils.
Collapse
Affiliation(s)
- Krishna Latha
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| | - Katelyn F. Jamison
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | - Wendy T. Watford
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
15
|
Breyer F, Härtlova A, Thurston T, Flynn HR, Chakravarty P, Janzen J, Peltier J, Heunis T, Snijders AP, Trost M, Ley SC. TPL-2 kinase induces phagosome acidification to promote macrophage killing of bacteria. EMBO J 2021; 40:e106188. [PMID: 33881780 PMCID: PMC8126920 DOI: 10.15252/embj.2020106188] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 02/05/2023] Open
Abstract
Tumour progression locus 2 (TPL‐2) kinase mediates Toll‐like receptor (TLR) activation of ERK1/2 and p38α MAP kinases in myeloid cells to modulate expression of key cytokines in innate immunity. This study identified a novel MAP kinase‐independent regulatory function for TPL‐2 in phagosome maturation, an essential process for killing of phagocytosed microbes. TPL‐2 catalytic activity was demonstrated to induce phagosome acidification and proteolysis in primary mouse and human macrophages following uptake of latex beads. Quantitative proteomics revealed that blocking TPL‐2 catalytic activity significantly altered the protein composition of phagosomes, particularly reducing the abundance of V‐ATPase proton pump subunits. Furthermore, TPL‐2 stimulated the phosphorylation of DMXL1, a regulator of V‐ATPases, to induce V‐ATPase assembly and phagosome acidification. Consistent with these results, TPL‐2 catalytic activity was required for phagosome acidification and the efficient killing of Staphylococcus aureus and Citrobacter rodentium following phagocytic uptake by macrophages. TPL‐2 therefore controls innate immune responses of macrophages to bacteria via V‐ATPase induction of phagosome maturation.
Collapse
Affiliation(s)
| | - Anetta Härtlova
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Teresa Thurston
- Department of Infectious Diseases, MRC Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | | | | | | | - Julien Peltier
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Tiaan Heunis
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, UK
| | - Steven C Ley
- The Francis Crick Institute, London, UK.,Department of Immunology & Inflammation, Centre for Molecular Immunology & Inflammation, Imperial College London, London, UK
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Liao X, Li Y. Nuclear Factor Kappa B in Autism Spectrum Disorder: A Systematic Review. Pharmacol Res 2020; 159:104918. [PMID: 32461184 DOI: 10.1016/j.phrs.2020.104918] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/02/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The nuclear factor kappa B (NF-κB) is composed of a series of transcription factors, which are involved in the expression of a plethora of target genes, many of these genes contributing to the regulation of inflammatory responses. Consistent with its central role in inflammatory responses, existing studies of the neurobiological basis for ASD propose the involvement of NF-κB in the etiology of this disorder. OBJECTIVES The present review aimed to systematically characterize extant literatures regarding the role of NF-κB in the etiology of ASD through data derived from both human studies and animal models. METHODS A systematic electronic search was conducted for records indexed within Pubmed, EMBASE, or Web of Science to identify potentially eligible studies. Study inclusion and data extraction was agreed by two independent authors after reviewing the abstract and full text. RESULTS Among the 371 articles identified in the initial screening, 18 articles met the eligibility criteria for this review, including 14 human case-control studies compared the expression or activation of NF-κB between ASD cases and controls as well as 4 animal studies used mouse model of ASD to examine the level of NF-κB and further evaluate its changes after different drug treatments. These included 18 studies, although relatively small in quantity, appear to support the role of NF-κB in the etiology of ASD. CONCLUSIONS Evidence generated from both human studies and animal models supported the involvement of NF-κB in the neurobiological basis of ASD, despite some concern about whether it functions as a primary contributor causes ASD onset or rather an ancillary factor regulates ASD pathogenesis. The increased understanding of NF-κB in the neurobiological basis of ASD could aid the emergence of clinically relevant diagnostic biomarkers and novel therapeutic strategies acting on the underlying disease pathogenesis. These results suggested that potential methodological differences between studies need to be accounted for and keep open the discussion over the existence of aberrantly NF-κB signaling in ASD subjects.
Collapse
Affiliation(s)
- Xiaoli Liao
- Xiangya Nursing School, Central South University, Changsha, Hunan, China.
| | - Yamin Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
19
|
The IκB-protein BCL-3 controls Toll-like receptor-induced MAPK activity by promoting TPL-2 degradation in the nucleus. Proc Natl Acad Sci U S A 2019; 116:25828-25838. [PMID: 31772019 PMCID: PMC6926074 DOI: 10.1073/pnas.1900408116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The NF-ĸB and mitogen-activated protein kinase (MAPK) pathways coordinate the cellular response to most immune stimuli. Toll-like (TLR) and TNF receptor activation of the MAPK pathway requires activation of the TPL-2 kinase. Active TPL-2 is an unstable, short-lived protein, which limits MAPK activity and controls inflammatory responses. Here we report the surprising discovery that active TPL-2 shuttles between the cytoplasm and the nucleus, where it is degraded by the proteasome. BCL-3, a nuclear regulator of NF-ĸB, promotes the nuclear localization and degradation of TPL-2 in order to limit MAPK activity and determines the amount of TLR ligand required to initiate an inflammatory response. Thus, the nucleus is a key site for the integrated regulation of NF-ĸB– and MAPK-driven inflammatory responses. Proinflammatory responses induced by Toll-like receptors (TLRs) are dependent on the activation of the NF-ĸB and mitogen-activated protein kinase (MAPK) pathways, which coordinate the transcription and synthesis of proinflammatory cytokines. We demonstrate that BCL-3, a nuclear IĸB protein that regulates NF-ĸB, also controls TLR-induced MAPK activity by regulating the stability of the TPL-2 kinase. TPL-2 is essential for MAPK activation by TLR ligands, and the rapid proteasomal degradation of active TPL-2 is a critical mechanism limiting TLR-induced MAPK activity. We reveal that TPL-2 is a nucleocytoplasmic shuttling protein and identify the nucleus as the primary site for TPL-2 degradation. BCL-3 interacts with TPL-2 and promotes its degradation by promoting its nuclear localization. As a consequence, Bcl3−/− macrophages have increased TPL-2 stability following TLR stimulation, leading to increased MAPK activity and MAPK-dependent responses. Moreover, BCL-3–mediated regulation of TPL-2 stability sets the MAPK activation threshold and determines the amount of TLR ligand required to initiate the production of inflammatory cytokines. Thus, the nucleus is a key site in the regulation of TLR-induced MAPK activity. BCL-3 links control of the MAPK and NF-ĸB pathways in the nucleus, and BCL-3–mediated TPL-2 regulation impacts on the cellular decision to initiate proinflammatory cytokine production in response to TLR activation.
Collapse
|
20
|
Yang L, Ding JL. MEK1/2 Inhibitors Unlock the Constrained Interferon Response in Macrophages Through IRF1 Signaling. Front Immunol 2019; 10:2020. [PMID: 31507609 PMCID: PMC6718554 DOI: 10.3389/fimmu.2019.02020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Macrophages are immune sentinels essential for pathogen recognition and immune defense. Nucleic acid-sensing toll-like receptors like TLR7 activate tailored proinflammatory and interferon responses in macrophages. Here we found that TLR7 activation constrained itself and other TLRs from inducing interferon response genes in macrophages through MAPK kinase 1/2 (MEK1/2)-dependent IRF1 inhibition. Downstream of the MEK1/2-ERK pathway, TLR7-activated macrophages induced interleukin-10 (IL-10), a signal transducer and activator of transcription 3 (STAT3) signaling axis, which constrained the expression of interferon response genes, immunomodulatory cytokines, and chemokines. Nevertheless, MEK1/2 inhibitors unlocked an IRF1-interferon signature response in an NF-κB-dependent manner. Deficiency in interferon regulatory factor 1 (Irf1) completely abrogated the interferon response and prevented the reprogramming of macrophages into an immunostimulatory phenotype. As a proof of concept, combination treatment with a TLR7 agonist and MEK1/2 inhibitor synergistically extended the survival of wild-type but not Irf1-deficient melanoma-bearing mice. In a retrospective study, higher expression of Irf1 and interferon response genes correlated with more favorable prognosis in patients with cutaneous melanoma. Our findings demonstrated how MEK1/2 inhibitor unlocks IRF1-mediated interferon signature response in macrophages, and the therapeutic potentials of combination therapy with MEK1/2 inhibitor and TLR7 agonist.
Collapse
Affiliation(s)
- Lei Yang
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jeak Ling Ding
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
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
|
22
|
Advancement in TPL2-regulated innate immune response. Immunobiology 2019; 224:383-387. [PMID: 30853309 DOI: 10.1016/j.imbio.2019.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/12/2019] [Accepted: 02/21/2019] [Indexed: 11/21/2022]
Abstract
Tumor progression locus 2 (TPL2) is a serine/threonine kinase that belongs to the MAP3K family. The activated TPL2 regulates the innate immune-relevant signaling pathways, such as ERK, JNK, and NF-κB, and the differentiation of immune cells, for example, CD4+ T and NK cells. Therefore, TPL2 plays a critical role in regulating the innate immune response. The present review summarizes the recent advancements in the TPL2-regulated innate immune response.
Collapse
|
23
|
Minshawi F, White MRH, Muller W, Humphreys N, Jackson D, Campbell BJ, Adamson A, Papoutsopoulou S. Human TNF-Luc reporter mouse: A new model to quantify inflammatory responses. Sci Rep 2019; 9:193. [PMID: 30655563 PMCID: PMC6336827 DOI: 10.1038/s41598-018-36969-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022] Open
Abstract
Tumour necrosis factor (TNF) is a key cytokine during inflammatory responses and its dysregulation is detrimental in many inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease. Here, we used a bacterial artificial chromosome (BAC) construct that expresses luciferase under the control of the human TNF locus to generate a novel transgenic mouse, the hTNF.LucBAC strain. In vitro stimulation of hTNF.LucBAC cells of different origin revealed a cell specific response to stimuli demonstrating the integrated construct's ability as a proxy for inflammatory gene response. Lipopolysaccharide was the most potent luciferase inducer in macrophages, while TNF was a strong activator in intestinal organoids. Lipopolysaccharide-induced luciferase activity in macrophages was downregulated by inhibitors of NF-κB pathway, as well as by Interleukin-10, a known anti-inflammatory cytokine. Moreover, the transgene-dependent luciferase activity showed a positive correlation to the endogenous murine soluble TNF secreted to the culture medium. In conclusion, the hTNF.LucBAC strain is a valuable tool for studying and screening molecules that target TNF synthesis and will allow further functional studies of the regulatory elements of the TNF locus.
Collapse
Affiliation(s)
- Faisal Minshawi
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom.,Department of Laboratory Medicine, Faculty of Applied Medical Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mike R H White
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Werner Muller
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Neil Humphreys
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Dean Jackson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Barry J Campbell
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Antony Adamson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom.
| | - Stamatia Papoutsopoulou
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom.
| |
Collapse
|
24
|
Shi JH, Sun SC. Tumor Necrosis Factor Receptor-Associated Factor Regulation of Nuclear Factor κB and Mitogen-Activated Protein Kinase Pathways. Front Immunol 2018; 9:1849. [PMID: 30140268 PMCID: PMC6094638 DOI: 10.3389/fimmu.2018.01849] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/26/2018] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor receptor (TNFR)-associated factors (TRAFs) are a family of structurally related proteins that transduces signals from members of TNFR superfamily and various other immune receptors. Major downstream signaling events mediated by the TRAF molecules include activation of the transcription factor nuclear factor κB (NF-κB) and the mitogen-activated protein kinases (MAPKs). In addition, some TRAF family members, particularly TRAF2 and TRAF3, serve as negative regulators of specific signaling pathways, such as the noncanonical NF-κB and proinflammatory toll-like receptor pathways. Thus, TRAFs possess important and complex signaling functions in the immune system and play an important role in regulating immune and inflammatory responses. This review will focus on the role of TRAF proteins in the regulation of NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Jian-Hong Shi
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, China
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
25
|
Bagnall J, Boddington C, England H, Brignall R, Downton P, Alsoufi Z, Boyd J, Rowe W, Bennett A, Walker C, Adamson A, Patel NMX, O’Cualain R, Schmidt L, Spiller DG, Jackson DA, Müller W, Muldoon M, White MRH, Paszek P. Quantitative analysis of competitive cytokine signaling predicts tissue thresholds for the propagation of macrophage activation. Sci Signal 2018; 11:11/540/eaaf3998. [DOI: 10.1126/scisignal.aaf3998] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Li W, Pan X, Cheng W, Cheng Y, Yin Y, Chen J, Xu G, Xie L. Serum biochemistry, histology and transcriptomic profile analysis reflect liver inflammation and damage following dietary histamine supplementation in yellow catfish (Pelteobagrus fulvidraco). FISH & SHELLFISH IMMUNOLOGY 2018; 77:83-90. [PMID: 29571769 DOI: 10.1016/j.fsi.2018.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/13/2018] [Accepted: 03/17/2018] [Indexed: 06/08/2023]
Abstract
Previous studies suggested that diets containing high levels of histamine influenced digestive system of aquatic animals. In addition, the exogenous histamine was first detoxified by diamine oxidase in the intestine, while the rest of histamine was further detoxified in the liver. Thus, based on the evidence from the previous studies, we hypothesized that high levels of histamine may lead to damage on liver of the aquatic animals. Here, in current attempt, we sought to investigate the toxic effect of histamine on yellow catfish (Pelteobagrus fulvidraco) liver physiology and pathogenesis. In the present study, yellow catfish were fed for 56 days on diets supplemented with 1000 mg kg-1 histamine (His) or a basal diet as the control group (Con). A significant change on the morphology of the intestine and liver was observed, followed with an induction of serum activity of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Furthermore, the transcriptomic analysis was performed to gain an overview of the gene expression profile in liver between control and histamine supplemented groups. Through the bioinformatics analysis, 431 differentially expressed genes were identified. Among these genes, Gene Ontology enrichment analysis (GO) suggests that immune-related genes are significantly dysregulated. In addition, TNF signaling pathway is enriched in Kyoto Encyclopedia of Genes and Genomes analysis (KEGG), and is also the dominant pathway in immune system, suggesting that the inflammatory response and apoptosis of hepatocytes are induced by exogenous histamine.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Xiaohan Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China; Department of Plastic Surgery, Guangzhou Overseas Chinese Hospital, The First Affiliated Hospital of Jinan University, 510632, Guangdong Province, PR China
| | - Weixuan Cheng
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Yanbo Cheng
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Yulong Yin
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Jintao Chen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangzhou 510070, Guangdong Province, PR China.
| |
Collapse
|
27
|
Chorzalska A, Ahsan N, Rao RSP, Roder K, Yu X, Morgan J, Tepper A, Hines S, Zhang P, Treaba DO, Zhao TC, Olszewski AJ, Reagan JL, Liang O, Gruppuso PA, Dubielecka PM. Overexpression of Tpl2 is linked to imatinib resistance and activation of MEK-ERK and NF-κB pathways in a model of chronic myeloid leukemia. Mol Oncol 2018; 12:630-647. [PMID: 29485707 PMCID: PMC5928369 DOI: 10.1002/1878-0261.12186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/15/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022] Open
Abstract
The introduction of tyrosine kinase inhibitors (TKI) has transformed chronic myeloid leukemia (CML) into a chronic disease with long-term survival exceeding 85%. However, resistance of CML stem cells to TKI may contribute to the 50% relapse rate observed after TKI discontinuation in molecular remission. We previously described a model of resistance to imatinib mesylate (IM), in which K562 cells cultured in high concentrations of imatinib mesylate showed reduced Bcr-Abl1 protein and activity levels while maintaining proliferative potential. Using quantitative phosphoproteomic analysis of these IM-resistant cells, we have now identified significant upregulation of tumor progression locus (Tpl2), also known as cancer Osaka thyroid (COT1) kinase or Map3k8. Overexpression of Tpl2 in IM-resistant cells was accompanied by elevated activities of Src family kinases (SFKs) and NF-κB, MEK-ERK signaling. CD34+ cells isolated from the bone marrow of patients with CML and exposed to IMin vitro showed increased MAP3K8 transcript levels. Dasatinib (SFK inhibitor), U0126 (MEK inhibitor), and PS-1145 (IκB kinase (IKK) inhibitor) used in combination resulted in elimination of 65% of IM-resistant cells and reduction in the colony-forming capacity of CML CD34+ cells in methylcellulose assays by 80%. In addition, CML CD34+ cells cultured with the combination of inhibitors showed reduced MAP3K8 transcript levels. Overall, our data indicate that elevated Tpl2 protein and transcript levels are associated with resistance to IM and that combined inhibition of SFK, MEK, and NF-κB signaling attenuates the survival of IM-resistant CML cells and CML CD34+ cells. Therefore, combination of SFK, MEK, and NF-κB inhibitors may offer a new therapeutic approach to overcome TKI resistance in CML patients.
Collapse
Affiliation(s)
- Anna Chorzalska
- Signal Transduction Lab, Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Nagib Ahsan
- Division of Biology and Medicine, COBRE CCRD Proteomics Core Facility, Rhode Island Hospital, Brown University, Providence, RI, USA
| | - R Shyama Prasad Rao
- Division of Biostatistics and Bioinformatics, Yenepoya Research Center, Yenepoya University, Mangalore, India
| | - Karim Roder
- Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Xiaoqing Yu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - John Morgan
- Flow Cytometry and Cell Sorting Core Facility, Roger Williams Medical Center, Providence, RI, USA
| | - Alexander Tepper
- Signal Transduction Lab, Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Steven Hines
- Signal Transduction Lab, Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Peng Zhang
- Cardiovascular Research Center, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Diana O Treaba
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Ting C Zhao
- Cardiovascular Lab, Department of Surgery, Roger Williams Medical Center, Boston University School of Medicine, Providence, RI, USA
| | - Adam J Olszewski
- Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - John L Reagan
- Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Olin Liang
- Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Philip A Gruppuso
- Department of Pediatrics, Rhode Island Hospital, Brown University, Providence, RI, USA
| | - Patrycja M Dubielecka
- Signal Transduction Lab, Division of Hematology/Oncology, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
28
|
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
|
29
|
Zhou X, Chang Y, Zhan Y, Wang X, Lin K. Integrative mRNA-miRNA interaction analysis associate with immune response of sea cucumber Apostichopus japonicus based on transcriptome database. FISH & SHELLFISH IMMUNOLOGY 2018; 72:69-76. [PMID: 29054825 DOI: 10.1016/j.fsi.2017.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 06/07/2023]
Abstract
MicroRNAs (miRNAs) constitute a family of endogenous non-coding small RNAs that have been demonstrated to be the key effectors in mediating host-pathogen interactions. Additionally, high-throughput sequencing provides unexampled opportunities to identify the pathogenic mechanism underlying miRNAs. In the present study, the target genes of immune-related miRNAs (miR-31, miR-2008, miR-92a, miR-210 and miR-7) and specific miRNAs (miR-2004) in Echinodermata were predicted in silico and validated. Gene ontology (GO) analysis of the target genes of these six miRNAs were conducted to further understand the regulatory function in the host immunity of Apostichopus japonicus (A. japonicus). Among the putative target genes of the six miRNAs, various immune-related targets were annotated, such as Nephl, SEC14Ll, p105, GL2, LYS, FNIAL, mTOR, LITAF, SLC44, TLR3, Apaf-1, and CNTN4. This work will provide valuable genetic resources to understand the interaction of multiple mRNA-miRNAs and the regulation mechanism in the anti-bacterial process in the sea cucumber.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China
| | - Yaqing Chang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China.
| | - Yaoyao Zhan
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China
| | - Xiuli Wang
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China
| | - Kai Lin
- Key Laboratory of Mariculture & Stock Enhancement in North China's Sea, Ministry of Agriculture, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
30
|
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
|
31
|
Modulation of mitogen‑activated protein kinase attenuates sepsis‑induced acute lung injury in acute respiratory distress syndrome rats. Mol Med Rep 2017; 16:9652-9658. [PMID: 29039541 DOI: 10.3892/mmr.2017.7811] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 08/10/2017] [Indexed: 11/05/2022] Open
Abstract
Sepsis is the most important predisposing cause inducing acute respiratory distress syndrome (ARDS); however, the mechanism of sepsis leading to the development of ARDS remains to be elucidated. Suppression of the mitogen‑activated protein kinase (MAPK) signal by blocking the phosphorylation of Jun N‑terminal kinase (JNK) and p38 in lung tissues could alleviate acute lung injury induced by sepsis. MAPK signaling may have a crucial role in development of the sepsis‑induced acute lung injury. The specific inhibitors of JNK and p38 MAPK, SP600125 and SB203580, were administrated by intragastric injection 4 h before induction of ARDS after cecal ligation and puncture (CLP). Rats were sacrificed at 1, 6 or 24 h after CLP challenge. The histological evaluation, lung water content, and biochemical analysis were performed. The results revealed that the JNK and p38 MAPK inhibitor improved lung permeability, attenuated system inflammation, further alleviated the lung injury induced by sepsis. In conclusion, JNK and p38 MAPK signaling are essential for the development of ARDS following sepsis. Further studies are needed to illuminate the detailed mechanisms of JNK and p38 MAPK signaling in sepsis‑induced ARDS.
Collapse
|
32
|
Mitchell JP, Carmody RJ. NF-κB and the Transcriptional Control of Inflammation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:41-84. [PMID: 29305014 DOI: 10.1016/bs.ircmb.2017.07.007] [Citation(s) in RCA: 342] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The NF-κB transcription factor was discovered 30 years ago and has since emerged as the master regulator of inflammation and immune homeostasis. It achieves this status by means of the large number of important pro- and antiinflammatory factors under its transcriptional control. NF-κB has a central role in inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and autoimmunity, as well as diseases comprising a significant inflammatory component such as cancer and atherosclerosis. Here, we provide an overview of the studies that form the basis of our understanding of the role of NF-κB subunits and their regulators in controlling inflammation. We also describe the emerging importance of posttranslational modifications of NF-κB in the regulation of inflammation, and highlight the future challenges faced by researchers who aim to target NF-κB transcriptional activity for therapeutic benefit in treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jennifer P Mitchell
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ruaidhrí J Carmody
- Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
33
|
The non-canonical NF-κB pathway in immunity and inflammation. NATURE REVIEWS. IMMUNOLOGY 2017. [PMID: 28580957 DOI: 10.1038/nri.2017.52)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
|
34
|
TPL2 meets p38MAPK: emergence of a novel positive feedback loop in inflammation. Biochem J 2017; 473:2995-9. [PMID: 27679858 DOI: 10.1042/bcj20160672c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/28/2016] [Indexed: 01/06/2023]
Abstract
The activation of p38(MAPK) by Toll-like receptor signalling is essential for the inflammatory response of innate immunity due to its role in post-transcriptional regulation of TNFα and cytokine biosynthesis. p38(MAPK) activation proceeds by the upstream MAP2Ks, MAPK kinase (MKK)3/6 as well as MKK4, which in turn are substrates for MAP3Ks, such as TGFβ-activated protein kinase-1 (TAK1). In contrast, TPL2 has been described as an exclusive MAP3K of MKK1/2-triggering activation of the classical ERKs, ERK1/2. In the recent issue of the Biochemical Journal, Pattison et al report their screening for TPL2 substrates in LPS-stimulated macrophages and the identification of MKK3/6. Using catalytic-dead TPL2 (Map3k8(D270A/D270A)) knockin macrophages, they demonstrated that activation of MKK3/6 by TPL2 significantly contributes to LPS-dependent TNFα biosynthesis and is also essential for TNF-receptor 1 signalling. Hence, a new signalling pathway from TAK1 via IκB kinase, p105 NFκB and TPL2 to MKK3/6 and p38(MAPK) is established in macrophages. Taking into account that some isoforms of p38(MAPK) are necessary for maintaining functional steady-state levels of TPL2, a positive feedback loop in inflammation emerges.
Collapse
|
35
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, MD Anderson Cancer Center UT Heath Graduate School of Biomedical Sciences, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| |
Collapse
|
36
|
Senger K, Pham VC, Varfolomeev E, Hackney JA, Corzo CA, Collier J, Lau VWC, Huang Z, Hamidzhadeh K, Caplazi P, Peng I, Setiadi AF, Francis R, Paler-Martinez A, Kwon YC, Ramirez-Carrozzi V, Sun Y, Grigg PW, Roose-Girma M, Jeet S, Barck KH, Pham A, Ota N, Ha C, Stinson J, Guillory J, Tam L, Modrusan Z, Emson C, McKenzie BS, Townsend MJ, Carano RAD, Warming S, Vucic D, DeVoss J, Lee WP, Lill JR, Zarrin AA. The kinase TPL2 activates ERK and p38 signaling to promote neutrophilic inflammation. Sci Signal 2017; 10:10/475/eaah4273. [PMID: 28420753 DOI: 10.1126/scisignal.aah4273] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor progression locus 2 (TPL2; also known as MAP3K8) is a mitogen-activated protein kinase (MAPK) kinase kinase (MAP3K) that phosphorylates the MAPK kinases MEK1 and MEK2 (MEK1/2), which, in turn, activate the MAPKs extracellular signal-regulated kinase 1 (ERK1) and ERK2 (ERK1/2) in macrophages stimulated through the interleukin-1 receptor (IL-1R), Toll-like receptors (TLRs), or the tumor necrosis factor receptor (TNFR). We describe a conserved and critical role for TPL2 in mediating the effector functions of neutrophils through the activation of the p38 MAPK signaling pathway. Gene expression profiling and functional studies of neutrophils and monocytes revealed a MEK1/2-independent branch point downstream of TPL2 in neutrophils. Biochemical analyses identified the MAPK kinases MEK3 and MEK6 and the MAPKs p38α and p38δ as downstream effectors of TPL2 in these cells. Genetic ablation of the catalytic activity of TPL2 or therapeutic intervention with a TPL2-specific inhibitor reduced the production of inflammatory mediators by neutrophils in response to stimulation with the TLR4 agonist lipopolysaccharide (LPS) in vitro, as well as in rodent models of inflammatory disease. Together, these data suggest that TPL2 is a drug target that activates not only MEK1/2-dependent but also MEK3/6-dependent signaling to promote inflammatory responses.
Collapse
Affiliation(s)
- Kate Senger
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Victoria C Pham
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Eugene Varfolomeev
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A Hackney
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Cesar A Corzo
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jenna Collier
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Vivian W C Lau
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhiyu Huang
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kajal Hamidzhadeh
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patrick Caplazi
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ivan Peng
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - A Francesca Setiadi
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ross Francis
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Youngsu C Kwon
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Yonglian Sun
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patricia W Grigg
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Merone Roose-Girma
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Surinder Jeet
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kai H Barck
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Anna Pham
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Naruhisa Ota
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Connie Ha
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jeremy Stinson
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph Guillory
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lucinda Tam
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Claire Emson
- 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
| | - Michael J Townsend
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Richard A D Carano
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Søren Warming
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Domagoj Vucic
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason DeVoss
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wyne P Lee
- Genentech Research, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jennie R Lill
- 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
|
37
|
Rosenberger CM, Podyminogin RL, Diercks AH, Treuting PM, Peschon JJ, Rodriguez D, Gundapuneni M, Weiss MJ, Aderem A. miR-144 attenuates the host response to influenza virus by targeting the TRAF6-IRF7 signaling axis. PLoS Pathog 2017; 13:e1006305. [PMID: 28380049 PMCID: PMC5393898 DOI: 10.1371/journal.ppat.1006305] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/17/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023] Open
Abstract
Antiviral responses must rapidly defend against infection while minimizing inflammatory damage, but the mechanisms that regulate the magnitude of response within an infected cell are not well understood. miRNAs are small non-coding RNAs that suppress protein levels by binding target sequences on their cognate mRNA. Here, we identify miR-144 as a negative regulator of the host antiviral response. Ectopic expression of miR-144 resulted in increased replication of three RNA viruses in primary mouse lung epithelial cells: influenza virus, EMCV, and VSV. We identified the transcriptional network regulated by miR-144 and demonstrate that miR-144 post-transcriptionally suppresses TRAF6 levels. In vivo ablation of miR-144 reduced influenza virus replication in the lung and disease severity. These data suggest that miR-144 reduces the antiviral response by attenuating the TRAF6-IRF7 pathway to alter the cellular antiviral transcriptional landscape. Antiviral responses must be regulated to rapidly defend against infection while minimizing inflammatory damage. However, the mechanisms for establishing the magnitude of response within an infected cell are incompletely understood. miRNAs are small non-coding RNAs that negatively regulate protein levels by binding complementary sequences on their target mRNA. In this study, we show that microRNA-144 impairs the ability of host cells to control the replication of three viruses: influenza virus, EMCV, and VSV. We identify a mechanism underlying the effect of this microRNA on antiviral responses. microRNA-144 suppresses TRAF6 levels and impairs the gene expression program regulated by the transcription factor IRF7. The resulting dysregulated expression of antiviral genes correlates with enhanced viral replication. Our findings in isolated lung epithelial cells were consistent with the effects observed in influenza virus-infected mice lacking miR-144. Together, these data support a role for miRNAs in tuning transcriptional programs during early responses to viral infection.
Collapse
Affiliation(s)
- Carrie M. Rosenberger
- Center for Infectious Disease Research, Seattle, WA United States of America
- * E-mail: (CMR); (AA)
| | | | - Alan H. Diercks
- Center for Infectious Disease Research, Seattle, WA United States of America
| | - Piper M. Treuting
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Jacques J. Peschon
- Center for Infectious Disease Research, Seattle, WA United States of America
| | - David Rodriguez
- Center for Infectious Disease Research, Seattle, WA United States of America
| | | | - Mitchell J. Weiss
- Hematology, St. Jude Children's Research Hospital, Memphis, TN United States of America
| | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA United States of America
- * E-mail: (CMR); (AA)
| |
Collapse
|
38
|
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
|
39
|
Adamson A, Boddington C, Downton P, Rowe W, Bagnall J, Lam C, Maya-Mendoza A, Schmidt L, Harper CV, Spiller DG, Rand DA, Jackson DA, White MRH, Paszek P. Signal transduction controls heterogeneous NF-κB dynamics and target gene expression through cytokine-specific refractory states. Nat Commun 2016; 7:12057. [PMID: 27381163 PMCID: PMC4935804 DOI: 10.1038/ncomms12057] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 05/25/2016] [Indexed: 02/03/2023] Open
Abstract
Cells respond dynamically to pulsatile cytokine stimulation. Here we report that single, or well-spaced pulses of TNFα (>100 min apart) give a high probability of NF-κB activation. However, fewer cells respond to shorter pulse intervals (<100 min) suggesting a heterogeneous refractory state. This refractory state is established in the signal transduction network downstream of TNFR and upstream of IKK, and depends on the level of the NF-κB system negative feedback protein A20. If a second pulse within the refractory phase is IL-1β instead of TNFα, all of the cells respond. This suggests a mechanism by which two cytokines can synergistically activate an inflammatory response. Gene expression analyses show strong correlation between the cellular dynamic response and NF-κB-dependent target gene activation. These data suggest that refractory states in the NF-κB system constitute an inherent design motif of the inflammatory response and we suggest that this may avoid harmful homogenous cellular activation.
Collapse
Affiliation(s)
- Antony Adamson
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Christopher Boddington
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Polly Downton
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - William Rowe
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - James Bagnall
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Connie Lam
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Apolinar Maya-Mendoza
- The Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Denmark
| | - Lorraine Schmidt
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Claire V. Harper
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - David G. Spiller
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - David A. Rand
- Warwick Systems Biology and Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK
| | - Dean A. Jackson
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Michael R. H. White
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Pawel Paszek
- Systems Microscopy Centre, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
40
|
Cartwright T, Perkins ND, L Wilson C. NFKB1: a suppressor of inflammation, ageing and cancer. FEBS J 2016; 283:1812-22. [PMID: 26663363 DOI: 10.1111/febs.13627] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/23/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022]
Abstract
The pleiotropic consequences of nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) pathway activation result from the combinatorial effects of the five subunits that form the homo- and heterodimeric NF-κB complexes. Although biochemical and gene knockout studies have demonstrated overlapping and distinct functions for these proteins, much is still not known about the mechanisms determining context-dependent functions, the formation of different dimer complexes and transcriptional control in response to diverse stimuli. Here we discuss recent results that reveal that the nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1) (p105/p50) subunit is an important regulator of NF-κB activity in vivo. These effects are not restricted to being a dimer partner for other NF-κB subunits. Rather p50 homodimers have a critical role as suppressors of the NF-κB response, while the p105 precursor has a variety of NF-κB-independent functions. The importance of Nfkb1 function can be seen in mouse models, where Nfkb1(-/-) mice display increased inflammation and susceptibility to certain forms of DNA damage, leading to cancer, and a rapid ageing phenotype. In humans, low expression of Kip1 ubiquitination-promoting complex 1 (KPC1), a ubiquitin ligase required for p105 to p50 processing, was shown to correlate with a reduction in p50 and glioblastoma incidence. Therefore, while the majority of research in this field has focused on the upstream signalling pathways leading to NF-κB activation or the function of other NF-κB subunits, such as RelA (p65), these data demonstrate a critical role for NFKB1, potentially revealing new strategies for targeting this pathway in inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Tyrell Cartwright
- Fibrosis Laboratory, Institute of Cellular Medicine, Newcastle University, UK
| | - Neil D Perkins
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, UK
| | - Caroline L Wilson
- Fibrosis Laboratory, Institute of Cellular Medicine, Newcastle University, UK
| |
Collapse
|
41
|
Bacterial Stimulation of Toll-Like Receptor 4 Drives Macrophages To Hemophagocytose. Infect Immun 2015; 84:47-55. [PMID: 26459510 DOI: 10.1128/iai.01149-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/07/2015] [Indexed: 01/25/2023] Open
Abstract
During acute infection with bacteria, viruses or parasites, a fraction of macrophages engulf large numbers of red and white blood cells, a process called hemophagocytosis. Hemophagocytes persist into the chronic stage of infection and have an anti-inflammatory phenotype. Salmonella enterica serovar Typhimurium infection of immunocompetent mice results in acute followed by chronic infection, with the accumulation of hemophagocytes. The mechanism(s) that triggers a macrophage to become hemophagocytic is unknown, but it has been reported that the proinflammatory cytokine gamma interferon (IFN-γ) is responsible. We show that primary macrophages become hemophagocytic in the absence or presence of IFN-γ upon infection with Gram-negative bacterial pathogens or prolonged exposure to heat-killed Salmonella enterica, the Gram-positive bacterium Bacillus subtilis, or Mycobacterium marinum. Moreover, conserved microbe-associated molecular patterns are sufficient to stimulate macrophages to hemophagocytose. Purified bacterial lipopolysaccharide (LPS) induced hemophagocytosis in resting and IFN-γ-pretreated macrophages, whereas lipoteichoic acid and synthetic unmethylated deoxycytidine-deoxyguanosine dinucleotides, which mimic bacterial DNA, induced hemophagocytosis only in IFN-γ-pretreated macrophages. Chemical inhibition or genetic deletion of Toll-like receptor 4, a pattern recognition receptor responsive to LPS, prevented both Salmonella- and LPS-stimulated hemophagocytosis. Inhibition of NF-κB also prevented hemophagocytosis. These results indicate that recognition of microbial products by Toll-like receptors stimulates hemophagocytosis, a novel outcome of prolonged Toll-like receptor signaling, suggesting hemophagocytosis is a highly conserved innate immune response.
Collapse
|
42
|
Gutmann S, Hinniger A, Fendrich G, Drückes P, Antz S, Mattes H, Möbitz H, Ofner S, Schmiedeberg N, Stojanovic A, Rieffel S, Strauss A, Troxler T, Glatthar R, Sparrer H. The Crystal Structure of Cancer Osaka Thyroid Kinase Reveals an Unexpected Kinase Domain Fold. J Biol Chem 2015; 290:15210-8. [PMID: 25918157 DOI: 10.1074/jbc.m115.648097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Indexed: 12/31/2022] Open
Abstract
Macrophages are important cellular effectors in innate immune responses and play a major role in autoimmune diseases such as rheumatoid arthritis. Cancer Osaka thyroid (COT) kinase, also known as mitogen-activated protein kinase kinase kinase 8 (MAP3K8) and tumor progression locus 2 (Tpl-2), is a serine-threonine (ST) kinase and is a key regulator in the production of pro-inflammatory cytokines in macrophages. Due to its pivotal role in immune biology, COT kinase has been identified as an attractive target for pharmaceutical research that is directed at the discovery of orally available, selective, and potent inhibitors for the treatment of autoimmune disorders and cancer. The production of monomeric, recombinant COT kinase has proven to be very difficult, and issues with solubility and stability of the enzyme have hampered the discovery and optimization of potent and selective inhibitors. We developed a protocol for the production of recombinant human COT kinase that yields pure and highly active enzyme in sufficient yields for biochemical and structural studies. The quality of the enzyme allowed us to establish a robust in vitro phosphorylation assay for the efficient biochemical characterization of COT kinase inhibitors and to determine the x-ray co-crystal structures of the COT kinase domain in complex with two ATP-binding site inhibitors. The structures presented in this study reveal two distinct ligand binding modes and a unique kinase domain architecture that has not been observed previously. The structurally versatile active site significantly impacts the design of potent, low molecular weight COT kinase inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Helmut Sparrer
- Autoimmunity Transplantation Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland
| |
Collapse
|
43
|
Kuriakose T, Rada B, Watford WT. Tumor progression locus 2-dependent oxidative burst drives phosphorylation of extracellular signal-regulated kinase during TLR3 and 9 signaling. J Biol Chem 2014; 289:36089-100. [PMID: 25378393 DOI: 10.1074/jbc.m114.587121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Signal transduction via NFκB and MAP kinase cascades is a universal response initiated upon pathogen recognition by Toll-like receptors (TLRs). How activation of these divergent signaling pathways is integrated to dictate distinct immune responses to diverse pathogens is still incompletely understood. Herein, contrary to current perception, we demonstrate that a signaling pathway defined by the inhibitor of κB kinase β (IKKβ), MAP3 kinase tumor progression locus 2 (Tpl2/MAP3K8), and MAP kinase ERK is differentially activated by TLRs. TLRs 2, 4, and 7 directly activate this inflammatory axis, inducing immediate ERK phosphorylation and early TNFα secretion. In addition to TLR adaptor proteins, IKKβ-Tpl2-ERK activation by TLR4 is regulated by the TLR4 co-receptor CD14 and the tyrosine kinase Syk. Signals from TLRs 3 and 9 do not initiate early activation of IKKβ-Tpl2-ERK pathway but instead induce delayed, NADPH-oxidase-dependent ERK phosphorylation and TNFα secretion via autocrine reactive oxygen species signaling. Unexpectedly, Tpl2 is an essential regulator of ROS production during TLR signaling. Overall, our study reveals distinct mechanisms activating a common inflammatory signaling cascade and delineates differences in MyD88-dependent signaling between endosomal TLRs 7 and 9. These findings further confirm the importance of Tpl2 in innate host defense mechanisms and also enhance our understanding of how the immune system tailors pathogen-specific gene expression patterns.
Collapse
Affiliation(s)
- Teneema Kuriakose
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| | - Balázs Rada
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| | - Wendy T Watford
- From the Department of Infectious Diseases, The University of Georgia, College of Veterinary Medicine, Athens, Georgia 30602
| |
Collapse
|
44
|
An unexpected twist to the activation of IKKβ: TAK1 primes IKKβ for activation by autophosphorylation. Biochem J 2014; 461:531-7. [PMID: 24911653 PMCID: PMC4206954 DOI: 10.1042/bj20140444] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
IKKβ {IκB [inhibitor of NF-κB (nuclear factor κB)] kinase β} is required to activate the transcription factor NF-κB, but how IKKβ itself is activated in vivo is still unclear. It was found to require phosphorylation by one or more ‘upstream’ protein kinases in some reports, but by autophosphorylation in others. In the present study, we resolve this contro-versy by demonstrating that the activation of IKKβ induced by IL-1 (interleukin-1) or TNF (tumour necrosis factor) in embryonic fibroblasts, or by ligands that activate Toll-like receptors in macrophages, requires two distinct phosphorylation events: first, the TAK1 [TGFβ (transforming growth factor β)-activated kinase-1]-catalysed phosphorylation of Ser177 and, secondly, the IKKβ-catalysed autophosphorylation of Ser181. The phosphorylation of Ser177 by TAK1 is a priming event required for the subsequent autophosphorylation of Ser181, which enables IKKβ to phosphorylate exogenous substrates. We also provide genetic evidence which indicates that the IL-1-stimulated, LUBAC (linear ubiquitin chain assembly complex)-catalysed formation of linear ubiquitin chains and their interaction with the NEMO (NF-κB essential modulator) component of the canonical IKK complex permits the TAK1-catalysed priming phosphorylation of IKKβ at Ser177 and IKKα at Ser176. These findings may be of general significance for the activation of other protein kinases. We have discovered how a key enzyme that controls the immune system is switched on during infection by bacteria and viruses. Known by the acronym IKKβ, it triggers the production of many proteins that are needed to combat these pathogens.
Collapse
|
45
|
Jacque E, Schweighoffer E, Visekruna A, Papoutsopoulou S, Janzen J, Zillwood R, Tarlinton DM, Tybulewicz VLJ, Ley SC. IKK-induced NF-κB1 p105 proteolysis is critical for B cell antibody responses to T cell-dependent antigen. ACTA ACUST UNITED AC 2014; 211:2085-101. [PMID: 25225457 PMCID: PMC4172221 DOI: 10.1084/jem.20132019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Jacque et al. investigate the functions of NF-κB1 p105 and its associated NF-κB–binding partners in B cells, using a mutant mouse strain that carries a form of the NF-κB1 precursor that is resistant to IKK-induced proteolysis. They identify a critical B cell–intrinsic role for this IKK signaling pathway in the antigen-induced survival and differentiation of follicular mature B cells. The importance of IκB kinase (IKK)–induced proteolysis of NF-κB1 p105 in B cells was investigated using Nfkb1SSAA/SSAA mice, in which this NF-κB signaling pathway is blocked. Nfkb1SSAA mutation had no effect on the development and homeostasis of follicular mature (FM) B cells. However, analysis of mixed bone marrow chimeras revealed that Nfkb1SSAA/SSAA FM B cells were completely unable to mediate T cell–dependent antibody responses. Nfkb1SSAA mutation decreased B cell antigen receptor (BCR) activation of NF-κB in FM B cells, which selectively blocked BCR stimulation of cell survival and antigen-induced differentiation into plasmablasts and germinal center B cells due to reduced expression of Bcl-2 family proteins and IRF4, respectively. In contrast, the antigen-presenting function of FM B cells and their BCR-induced migration to the follicle T cell zone border, as well as their growth and proliferation after BCR stimulation, were not affected. All of the inhibitory effects of Nfkb1SSAA mutation on B cell functions were rescued by normalizing NF-κB activation genetically. Our study identifies critical B cell-intrinsic functions for IKK-induced NF-κB1 p105 proteolysis in the antigen-induced survival and differentiation of FM B cells, which are essential for T-dependent antibody responses.
Collapse
Affiliation(s)
- Emilie Jacque
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Edina Schweighoffer
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Alexander Visekruna
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Stamatia Papoutsopoulou
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Julia Janzen
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Rachel Zillwood
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - David M Tarlinton
- The Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria 3052, Australia
| | - Victor L J Tybulewicz
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Steven C Ley
- Division of Immune Cell Biology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| |
Collapse
|
46
|
Yang G, Wright CJ, Hinson MD, Fernando AP, Sengupta S, Biswas C, La P, Dennery PA. Oxidative stress and inflammation modulate Rev-erbα signaling in the neonatal lung and affect circadian rhythmicity. Antioxid Redox Signal 2014; 21:17-32. [PMID: 24252172 PMCID: PMC4048579 DOI: 10.1089/ars.2013.5539] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS The response to oxidative stress and inflammation varies with diurnal rhythms. Nevertheless, it is not known whether circadian genes are regulated by these stimuli. We evaluated whether Rev-erbα, a key circadian gene, was regulated by oxidative stress and/or inflammation in vitro and in a mouse model. RESULTS A unique sequence consisting of overlapping AP-1 and nuclear factor kappa B (NFκB) consensus sequences was identified on the mouse Rev-erbα promoter. This sequence mediates Rev-erbα promoter activity and transcription in response to oxidative stress and inflammation. This region serves as an NrF2 platform both to receive oxidative stress signals and to activate Rev-erbα, as well as an NFκB-binding site to repress Rev-erbα with inflammatory stimuli. The amplitude of the rhythmicity of Rev-erbα was altered by pre-exposure to hyperoxia or disruption of NFκB in a cell culture model of circadian simulation. Oxidative stress overcame the inhibitory effect of NFκB binding on Rev-erbα transcription. This was confirmed in neonatal mice exposed to hyperoxia, where hyperoxia-induced lung Rev-erbα transcription was further increased with NFκB disruption. Interestingly, this effect was not observed in similarly exposed adult mice. INNOVATION These data provide novel mechanistic insights into how key circadian genes are regulated by oxidative stress and inflammation in the neonatal lung. CONCLUSION Rev-erbα transcription and circadian oscillation are susceptible to oxidative stress and inflammation in the neonate. Due to Rev-erbα's role in cellular metabolism, this could contribute to lung cellular function and injury from inflammation and oxidative stress.
Collapse
Affiliation(s)
- Guang Yang
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Clyde J. Wright
- Department of Pediatrics, University of Colorado, Aurora, Colorado
| | - Maurice D. Hinson
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Amal P. Fernando
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shaon Sengupta
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Chhanda Biswas
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ping La
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Phyllis A. Dennery
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
IκB kinase-induced interaction of TPL-2 kinase with 14-3-3 is essential for Toll-like receptor activation of ERK-1 and -2 MAP kinases. Proc Natl Acad Sci U S A 2014; 111:E2394-403. [PMID: 24912162 DOI: 10.1073/pnas.1320440111] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The MEK-1/2 kinase TPL-2 is critical for Toll-like receptor activation of the ERK-1/2 MAP kinase pathway during inflammatory responses, but it can transform cells following C-terminal truncation. IκB kinase (IKK) complex phosphorylation of the TPL-2 C terminus regulates full-length TPL-2 activation of ERK-1/2 by a mechanism that has remained obscure. Here, we show that TPL-2 Ser-400 phosphorylation by IKK and TPL-2 Ser-443 autophosphorylation cooperated to trigger TPL-2 association with 14-3-3. Recruitment of 14-3-3 to the phosphorylated C terminus stimulated TPL-2 MEK-1 kinase activity, which was essential for TPL-2 activation of ERK-1/2. The binding of 14-3-3 to TPL-2 was also indispensible for lipopolysaccharide-induced production of tumor necrosis factor by macrophages, which is regulated by TPL-2 independently of ERK-1/2 activation. Our data identify a key step in the activation of TPL-2 signaling and provide a mechanistic insight into how C-terminal deletion triggers the oncogenic potential of TPL-2 by rendering its kinase activity independent of 14-3-3 binding.
Collapse
|
48
|
Abstract
Toll-like receptors (TLRs) and the receptors for interleukin (IL)-1, IL-18 and IL-33 are required for defence against microbial pathogens but, if hyper-activated or not switched off efficiently, can cause tissue damage and inflammatory and autoimmune diseases. Understanding how the checks and balances in the system are integrated to fight infection without the network operating out of control will be crucial for the development of improved drugs to treat these diseases in the future. In this Cell Science at a Glance article and the accompanying poster, I provide a brief overview of how one of these intricate networks is controlled by the interplay of protein phosphorylation and protein ubiquitylation events, and the mechanisms in myeloid cells that restrict and terminate its activation to prevent inflammatory and autoimmune diseases. Finally, I suggest a few protein kinases that have been neglected as drug targets, but whose therapeutic potential should be explored in the light of recent advances in our understanding of their roles in the innate immune system.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
49
|
Basavarajappa HD, Lee B, Fei X, Lim D, Callaghan B, Mund JA, Case J, Rajashekhar G, Seo SY, Corson TW. Synthesis and mechanistic studies of a novel homoisoflavanone inhibitor of endothelial cell growth. PLoS One 2014; 9:e95694. [PMID: 24752613 PMCID: PMC3994091 DOI: 10.1371/journal.pone.0095694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/30/2014] [Indexed: 12/13/2022] Open
Abstract
Preventing pathological ocular angiogenesis is key to treating retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration. At present there is no small molecule drug on the market to target this process and hence there is a pressing need for developing novel small molecules that can replace or complement the present surgical and biologic therapies for these neovascular eye diseases. Previously, an antiangiogenic homoisoflavanone was isolated from the bulb of a medicinal orchid, Cremastra appendiculata. In this study, we present the synthesis of a novel homoisoflavanone isomer of this compound. Our compound, SH-11052, has antiproliferative activity against human umbilical vein endothelial cells, and also against more ocular disease-relevant human retinal microvascular endothelial cells (HRECs). Tube formation and cell cycle progression of HRECs were inhibited by SH-11052, but the compound did not induce apoptosis at effective concentrations. SH-11052 also decreased TNF-α induced p38 MAPK phosphorylation in these cells. Intriguingly, SH-11052 blocked TNF-α induced IκB-α degradation, and therefore decreased NF-κB nuclear translocation. It decreased the expression of NF-κB target genes and the pro-angiogenic or pro-inflammatory markers VCAM-1, CCL2, IL8, and PTGS2. In addition SH-11052 inhibited VEGF induced activation of Akt but not VEGF receptor autophosphorylation. Based on these results we propose that SH-11052 inhibits inflammation induced angiogenesis by blocking both TNF-α and VEGF mediated pathways, two major pathways involved in pathological angiogenesis. Synthesis of this novel homoisoflavanone opens the door to structure-activity relationship studies of this class of compound and further evaluation of its mechanism and potential to complement existing antiangiogenic drugs.
Collapse
Affiliation(s)
- Halesha D. Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Xiang Fei
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Daesung Lim
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Breedge Callaghan
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Julie A. Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Jamie Case
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Gangaraju Rajashekhar
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
- * E-mail: (S-YS); (TWC)
| | - Timothy W. Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (S-YS); (TWC)
| |
Collapse
|
50
|
Sriskantharajah S, Gückel E, Tsakiri N, Kierdorf K, Brender C, Ben-Addi A, Veldhoen M, Tsichlis PN, Stockinger B, O’Garra A, Prinz M, Kollias G, Ley SC. Regulation of experimental autoimmune encephalomyelitis by TPL-2 kinase. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:3518-3529. [PMID: 24639351 PMCID: PMC3979668 DOI: 10.4049/jimmunol.1300172] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor progression locus 2 (TPL-2) expression is required for efficient polarization of naive T cells to Th1 effector cells in vitro, as well as for Th1-mediated immune responses. In the present study, we investigated the potential role of TPL-2 in Th17 cells. TPL-2 was found to be dispensable for Th17 cell differentiation in vitro, and for the initial priming of Th17 cells in experimental autoimmune encephalomyelitis (EAE), a Th17 cell-mediated disease model for multiple sclerosis. Nevertheless, TPL-2-deficient mice were protected from EAE, which correlated with reduced immune cell infiltration, demyelination, and axonal damage in the CNS. Adoptive transfer experiments demonstrated that there was no T cell-intrinsic function for TPL-2 in EAE, and that TPL-2 signaling was not required in radiation-sensitive hematopoietic cells. Rather, TPL-2 signaling in radiation-resistant stromal cells promoted the effector phase of the disease. Importantly, using a newly generated mouse strain expressing a kinase-inactive form of TPL-2, we demonstrated that stimulation of EAE was dependent on the catalytic activity of TPL-2 and not its adaptor function to stabilize the associated ubiquitin-binding protein ABIN-2. Our data therefore raise the possibility that small molecule inhibitors of TPL-2 may be beneficial in multiple sclerosis therapy.
Collapse
Affiliation(s)
| | - Eva Gückel
- Divisions of Immune Cell Biology, National Institute for Medical Research, London, NW7 1AA, UK
| | - Niki Tsakiri
- Institute for Immunology, Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Attica, Greece
| | - Katrin Kierdorf
- Department of Neuropathology & BIOSS Center for Biological Signaling, University of Freiburg, D-79106 Freiburg, Germany
| | - Christine Brender
- Divisions of Immune Cell Biology, National Institute for Medical Research, London, NW7 1AA, UK
| | - Abduelhakem Ben-Addi
- Divisions of Immune Cell Biology, National Institute for Medical Research, London, NW7 1AA, UK
| | - Marc Veldhoen
- Molecular Immunology, National Institute for Medical Research, London, NW7 1AA, UK
| | - Philip N. Tsichlis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Brigitta Stockinger
- Molecular Immunology, National Institute for Medical Research, London, NW7 1AA, UK
| | - Anne O’Garra
- Immunoregulation, National Institute for Medical Research, London, NW7 1AA, UK
| | - Marco Prinz
- Department of Neuropathology & BIOSS Center for Biological Signaling, University of Freiburg, D-79106 Freiburg, Germany
| | - George Kollias
- Institute for Immunology, Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Attica, Greece
| | - Steven C. Ley
- Divisions of Immune Cell Biology, National Institute for Medical Research, London, NW7 1AA, UK
| |
Collapse
|