1
|
Wu Z, Zhang T, Ma X, Guo S, Zhou Q, Zahoor A, Deng G. Recent advances in anti-inflammatory active components and action mechanisms of natural medicines. Inflammopharmacology 2023; 31:2901-2937. [PMID: 37947913 DOI: 10.1007/s10787-023-01369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/16/2023] [Indexed: 11/12/2023]
Abstract
Inflammation is a series of reactions caused by the body's resistance to external biological stimuli. Inflammation affects the occurrence and development of many diseases. Anti-inflammatory drugs have been used widely to treat inflammatory diseases, but long-term use can cause toxic side-effects and affect human functions. As immunomodulators with long-term conditioning effects and no drug residues, natural products are being investigated increasingly for the treatment of inflammatory diseases. In this review, we focus on the inflammatory process and cellular mechanisms in the development of diseases such as inflammatory bowel disease, atherosclerosis, and coronavirus disease-2019. Also, we focus on three signaling pathways (Nuclear factor-kappa B, p38 mitogen-activated protein kinase, Janus kinase/signal transducer and activator of transcription-3) to explain the anti-inflammatory effect of natural products. In addition, we also classified common natural products based on secondary metabolites and explained the association between current bidirectional prediction progress of natural product targets and inflammatory diseases.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaofei Ma
- College of Veterinary Medicine, Gansu Agriculture University, Lanzhou, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qingqing Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Arshad Zahoor
- College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Chiang TI, Hung YY, Wu MK, Huang YL, Kang HY. TNIP2 mediates GRβ-promoted inflammation and is associated with severity of major depressive disorder. Brain Behav Immun 2021; 95:454-461. [PMID: 33932528 DOI: 10.1016/j.bbi.2021.04.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/17/2022] Open
Abstract
In depression, continual activation of the hypothalamic-pituitaryadrenal (HPA) axis with excess cortisol release leads to impair sensitivity of the glucocorticoid receptor (GR) and increase activity of the pro-inflammatory immune responses. Aberrant expression of GR has been associated with inflammation in patients with major depressive disorder (MDD). Our previous studies showed that the aberrant expression of TNFAIP3 gene, which encodes the NF-κB regulatory protein A20, TNFAIP3-associated proteins and Toll-like receptors (TLRs) are involved in inflammation-associated depression. However, the link between desensitization of GR actions and negative regulation of the TLRs-mediated inflammatory pathway in MDD is yet to be established. Here, we examined the association of depression severity, measured via the 17-item Hamilton Depression Rating Scale (HAMD-17), with the mRNA expression profiling of GRα, GRβ, TNFAIP3-interacting proteins (TNIP), including TNIP1, TNIP2, and TNIP3, and TNFAIP3-like proteins, such as cezanne1, cezanne2, trabid, and valosin-containing protein p97/p47 complex-interacting protein p135 (VCIP135), in monocytes from 69 patients with MDD and 42 healthy controls. Herein we found the mRNA expressions of GRβ and TNIP2 were significantly higher in monocytes from patients with MDD. Notably, TNIP2 level was positively correlated with the GRβ expression and severity of depression, as determined via Pearson's correlation analysis. Mechanistically, we demonstrated that overexpression of GRβ promotes the mRNA levels of TNIP2 and tumor necrosis factor alpha (TNF-α) in human monocytes. The promoting effect of GRβ on TNF-α expression was partially attenuated upon depletion of TNIP2, suggesting that TNIP2 was required for GRβ-mediated enhancement of TNF-α levels. Together, these results suggest that activation of GRβ/TNIP2/TNF-α axis may induce inflammation in MDD patients and targeting this newly identified pathway may help in the development of better therapeutic approaches to reduce the development of MDD.
Collapse
Affiliation(s)
- Ting-I Chiang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Yung Hung
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Kung Wu
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ya-Ling Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, College of Medicine, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
5
|
Zannikou M, Barbayianni I, Fanidis D, Grigorakaki T, Vlachopoulou E, Konstantopoulos D, Fousteri M, Nikitopoulou I, Kotanidou A, Kaffe E, Aidinis V. MAP3K8 Regulates Cox-2-Mediated Prostaglandin E 2 Production in the Lung and Suppresses Pulmonary Inflammation and Fibrosis. THE JOURNAL OF IMMUNOLOGY 2020; 206:607-620. [PMID: 33443087 DOI: 10.4049/jimmunol.2000862] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by exuberant deposition of extracellular matrix components, leading to the deterioration of lung architecture and respiratory functions. Profibrotic mechanisms are controlled by multiple regulatory molecules, including MAPKs, in turn regulated by multiple phosphorylation cascades. MAP3K8 is an MAPK kinase kinase suggested to pleiotropically regulate multiple pathogenic pathways in the context of inflammation and cancer; however, a possible role in the pathogenesis of IPF has not been investigated. In this report, MAP3K8 mRNA levels were found decreased in the lungs of IPF patients and of mice upon bleomycin-induced pulmonary fibrosis. Ubiquitous genetic deletion of Map3k8 in mice exacerbated the modeled disease, whereas bone marrow transfer experiments indicated that although MAP3K8 regulatory functions are active in both hematopoietic and nonhematopoietic cells, Map3k8 in hematopoietic cells has a more dominant role. Macrophage-specific deletion of Map3k8 was further found to be sufficient for disease exacerbation thus confirming a major role for macrophages in pulmonary fibrotic responses and suggesting a main role for Map3k8 in the homeostasis of their effector functions in the lung. Map3k8 deficiency was further shown to be associated with decreased Cox-2 expression, followed by a decrease in PGE2 production in the lung; accordingly, exogenous administration of PGE2 reduced inflammation and reversed the exacerbated fibrotic profile of Map3k8 -/- mice. Therefore, MAP3K8 has a central role in the regulation of inflammatory responses and Cox-2-mediated PGE2 production in the lung, and the attenuation of its expression is integral to pulmonary fibrosis development.
Collapse
Affiliation(s)
- Markella Zannikou
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Ilianna Barbayianni
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Dionysios Fanidis
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Theodora Grigorakaki
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Evlalia Vlachopoulou
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Dimitris Konstantopoulos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Ioanna Nikitopoulou
- GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; and
| | - Anastasia Kotanidou
- GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; and.,First Department of Critical Care and Pulmonary Services, Medical School, National and Kapodistrian University of Athens, Evangelismos Hospital, Athens 106 76, Greece
| | - Eleanna Kaffe
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece
| | - Vassilis Aidinis
- Institute of Bio-Innovation, Biomedical Sciences Research Center Alexander Fleming, 16672 Athens, Greece;
| |
Collapse
|
6
|
Njunge LW, Estania AP, Guo Y, Liu W, Yang L. Tumor progression locus 2 (TPL2) in tumor-promoting Inflammation, Tumorigenesis and Tumor Immunity. Am J Cancer Res 2020; 10:8343-8364. [PMID: 32724474 PMCID: PMC7381748 DOI: 10.7150/thno.45848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Over the years, tumor progression locus 2 (TPL2) has been identified as an essential modulator of immune responses that conveys inflammatory signals to downstream effectors, subsequently modulating the generation and function of inflammatory cells. TPL2 is also differentially expressed and activated in several cancers, where it is associated with increased inflammation, malignant transformation, angiogenesis, metastasis, poor prognosis and therapy resistance. However, the relationship between TPL2-driven inflammation, tumorigenesis and tumor immunity has not been addressed. Here, we reconcile the function of TPL2-driven inflammation to oncogenic functions such as inflammation, proliferation, apoptosis resistance, angiogenesis, metastasis, immunosuppression and immune evasion. We also address the controversies reported on TPL2 function in tumor-promoting inflammation and tumorigenesis, and highlight the potential role of the TPL2 adaptor function in regulating the mechanisms leading to pro-tumorigenic inflammation and tumor progression. We discuss the therapeutic implications and limitations of targeting TPL2 for cancer treatment. The ideas presented here provide some new insight into cancer pathophysiology that might contribute to the development of more integrative and specific anti-inflammatory and anti-cancer therapeutics.
Collapse
|
7
|
van der Vorst EPC, Weber C. Novel Features of Monocytes and Macrophages in Cardiovascular Biology and Disease. Arterioscler Thromb Vasc Biol 2019; 39:e30-e37. [PMID: 30673349 DOI: 10.1161/atvbaha.118.312002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Emiel P C van der Vorst
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany (E.P.C.v.d.V., C.W.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany (E.P.C.v.d.V., C.W.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (C.W.).,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.W.)
| |
Collapse
|
8
|
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
|
9
|
Gong J, Fang C, Zhang P, Wang PX, Qiu Y, Shen LJ, Zhang L, Zhu XY, Tian S, Li F, Wang Z, Huang Z, Wang A, Zhang XD, She ZG. Tumor Progression Locus 2 in Hepatocytes Potentiates Both Liver and Systemic Metabolic Disorders in Mice. Hepatology 2019; 69:524-544. [PMID: 29381809 DOI: 10.1002/hep.29820] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/25/2018] [Indexed: 12/27/2022]
Abstract
Tumor progression locus 2 (TPL2), a serine/threonine kinase, has been regarded as a potentially interesting target for the treatment of various diseases with an inflammatory component. However, the function of TPL2 in regulating hepatocyte metabolism and liver inflammation during the progression of nonalcoholic fatty liver disease (NAFLD) is poorly understood. Here, we report that TPL2 protein expression was significantly increased in fatty liver from diverse species, including humans, monkeys, and mice. Further investigations revealed that compared to wild-type (WT) littermates, hepatocyte-specific TPL2 knockout (HKO) mice exhibited improved lipid and glucose imbalance, reserved insulin sensitivity, and alleviated inflammation in response to high-fat diet (HFD) feeding. Overexpression of TPL2 in hepatocytes led to the opposite phenotype. Regarding the mechanism, we found that mitogen-activated protein kinase kinase 7 (MKK7) was the specific substrate of TPL2 for c-Jun N-terminal kinase (JNK) activation. TPL2-MKK7-JNK signaling in hepatocytes represents a promising drugable target for treating NAFLD and associated metabolic disorders. Conclusion: In hepatocytes, TPL2 acts as a key mediator that promotes both liver and systemic metabolic disturbances by specifically increasing MKK7-JNK activation.
Collapse
Affiliation(s)
- Jun Gong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Chun Fang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Peng Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yixing Qiu
- Lab of Animal Models and Functional Genomics (LAMFG), College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- TCM and Ethnomedicine Innovation & Development Laboratory, Sino-Pakistan TCM Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Li-Jun Shen
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Li Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Song Tian
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Feng Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Zhihua Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zan Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Aibing Wang
- Lab of Animal Models and Functional Genomics (LAMFG), College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xiao-Dong Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Caruana BT, Byrne FL, Knights AJ, Quinlan KGR, Hoehn KL. Characterization of Glucose Transporter 6 in Lipopolysaccharide-Induced Bone Marrow-Derived Macrophage Function. THE JOURNAL OF IMMUNOLOGY 2019; 202:1826-1832. [PMID: 30700586 DOI: 10.4049/jimmunol.1801063] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/31/2018] [Indexed: 11/19/2022]
Abstract
The polarization processes for M1 versus M2 macrophages are quite distinct in the context of changes in cellular metabolism. M1 macrophages are highly glycolytic, whereas M2 macrophages require a more oxidative nutrient metabolism. An important part of M1 polarization involves upregulation of the glucose transporter (GLUT) GLUT1 to facilitate increased glucose uptake and glycolytic metabolism; however, the role of other glucose transporters in this process is largely unknown. In surveying the Functional Annotation of the Mammalian Genome and Gene Expression Omnibus Profiles databases, we discovered that the glucose transporter GLUT6 is highly upregulated in LPS-activated macrophages. In our previous work, we have not detected mouse GLUT6 protein expression in any noncancerous tissue; therefore, in this study, we investigated the expression and significance of GLUT6 in bone marrow-derived macrophages from wild-type and GLUT6 knockout C57BL/6 mice. We show that LPS-induced M1 polarization markedly upregulated GLUT6 protein, whereas naive macrophages and IL-4-induced M2 macrophages do not express GLUT6 protein. However, despite strong upregulation of GLUT6 in M1 macrophages, the absence of GLUT6 did not alter M1 polarization in the context of glucose uptake, glycolytic metabolism, or cytokine production. Collectively, these data show that GLUT6 is dispensable for LPS-induced M1 polarization and function. These findings are important because GLUT6 is an anticancer drug target, and this study suggests that inhibition of GLUT6 may not impart detrimental side effects on macrophage function to interfere with their antitumor properties.
Collapse
Affiliation(s)
- Beth T Caruana
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia; and
| | - Frances L Byrne
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia; and
| | - Alexander J Knights
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia; and
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia; and
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia; and .,Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
11
|
Reduction of Real-Time Imaging of M1 Macrophage Chemotaxis toward Damaged Muscle Cells is PI3K-Dependent. Antioxidants (Basel) 2018; 7:antiox7100138. [PMID: 30297636 PMCID: PMC6210562 DOI: 10.3390/antiox7100138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/25/2018] [Accepted: 10/04/2018] [Indexed: 11/16/2022] Open
Abstract
Macrophages migrate and invade into damaged muscle rapidly and are important for muscle repair and subsequent regeneration. The exact cellular and biological events that cause macrophage migration toward injured muscle are not completely understood. In this study, the effect of macrophage differentiation on the chemotactic capability to invade local damaged muscle was investigated using an in vitro model of muscle injury. We used C2C12 cell myoblasts and J774 cell macrophages, and the "killed-C2C12" cells were combined with live C2C12 cells as a partially damaged muscle model. The cultured J774 cells, with or without lipopolysaccharide (LPS), were treated with Ly294002 (Ly), which is an inhibitor of phosphoinositide 3-kinase (PI3K). In order to evaluate the polarization effect of LPS stimulation on J774 cells, expression of cell surface Toll-like receptor 4 (TLR4), CD11c and CCR2, and expression of F-actin intensity, were analyzed by flow cytometry. The real-time horizontal chemotaxis assay of J774 cells was tested using the TAXIScan device. The expressions of TLR4, CD11c, and F-actin intensity in LPS-treated cells were significantly higher than those in Ctrl cells. In LPS-treated cells, the chemotactic activity toward damaged muscle cells completely disappeared. Moreover, the reduced chemotaxis depended far more on directionality than velocity. However, Ly treatment reversed the reduced chemotactic activity of the LPS-treated cells. In addition, cell-adhesion and F-actin intensity, but not CCR2 expression, in LPS-treated cells, was significantly reduced by Ly treatment. Taken together, our results suggest that the PI3K/Akt activation state drives migration behavior towards damaged muscle cells.
Collapse
|
12
|
TPL2 Is a Key Regulator of Intestinal Inflammation in Clostridium difficile Infection. Infect Immun 2018; 86:IAI.00095-18. [PMID: 29844241 DOI: 10.1128/iai.00095-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor progression locus 2 (TPL2), a serine/threonine protein kinase, is a major inflammatory mediator in immune cells. The predominant inflammatory actions of TPL2 depend on the activation of mitogen-activated protein kinases (MAPK) and the upregulated production of the cytokines tumor necrosis factor alpha (TNF-α) and interleukin 1β (IL-1β) in macrophages and dendritic cells in response to lipopolysaccharide (LPS). Significant increases in TNF-α, IL-6, IL-β, and IL-8 levels in patients with Clostridium difficile infection (CDI) have been reported. Both TNF-α and IL-6 have been postulated to play key roles in the systemic inflammatory response in CDI, and IL-8 is essential for the development of local intestinal inflammatory responses in CDI. The objective of this study was to elucidate the role of TPL2 in the pathogenesis of CDI. We found that TPL2 was significantly activated in human and mouse intestinal tissues upon C. difficile toxin exposure or CDI. We further demonstrated that TPL2 knockout (TPL2-KO) mice were significantly more resistant to CDI than wild-type mice, with significantly reduced production of TNF-α, IL-6, IL-1β, KC (a mouse homologue of IL-8), and myeloperoxidase (MPO) in the ceca and colons of TPL2-KO mice. Finally, we found that TPL2 inhibition by a specific inhibitor or TPL2 gene ablation significantly reduced TcdB-induced production of TNF-α, IL-6, IL-β, and KC by inhibiting the activation of p38, extracellular signal-regulated kinase (ERK), and c-Jun NH2-terminal kinase (JNK). Taken together, our data suggest that TPL2 represents a potential therapeutic target for CDI treatment.
Collapse
|
13
|
Kadzielawa K, Mathew B, Stelman CR, Lei AZ, Torres L, Roth S. Gene expression in retinal ischemic post-conditioning. Graefes Arch Clin Exp Ophthalmol 2018; 256:935-949. [PMID: 29504043 DOI: 10.1007/s00417-018-3905-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/30/2017] [Accepted: 01/08/2018] [Indexed: 12/29/2022] Open
Abstract
PURPOSE The pathophysiology of retinal ischemia involves mechanisms including inflammation and apoptosis. Ischemic post-conditioning (Post-C), a brief non-lethal ischemia, induces a long-term ischemic tolerance, but the mechanisms of ischemic post-conditioning in the retina have only been described on a limited basis. Accordingly, we conducted this study to determine the molecular events in retinal ischemic post-conditioning and to identify targets for therapeutic strategies for retinal ischemia. METHODS To determine global molecular events in ischemic post-conditioning, a comprehensive study of the transcriptome of whole retina was performed. We utilized RNA sequencing (RNA-Seq), a recently developed, deep sequencing technique enabling quantitative gene expression, with low background noise, dynamic detection range, and discovery of novel genes. Rat retina was subjected to ischemia in vivo by elevation of intraocular pressure above systolic blood pressure. At 24 h after ischemia, Post-C or sham Post-C was performed by another, briefer period of ischemia, and 24 h later, retinas were collected and RNA processed. RESULTS There were 71 significantly affected pathways in post-conditioned/ischemic vs. normals and 43 in sham post conditioned/ischemic vs. normals. Of these, 28 were unique to Post-C and ischemia. Seven biological pathways relevant to ischemic injury, in Post-C as opposed to sham Post-C, were examined in detail. Apoptosis, p53, cell cycle, JAK-STAT, HIF-1, MAPK and PI3K-Akt pathways significantly differed in the number as well as degree of fold change in genes between conditions. CONCLUSION Post-C is a complex molecular signaling process with a multitude of altered molecular pathways. We identified potential gene candidates in Post-C. Studying the impact of altering expression of these factors may yield insight into new methods for treating or preventing damage from retinal ischemic disorders.
Collapse
Affiliation(s)
- Konrad Kadzielawa
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Biji Mathew
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Clara R Stelman
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Arden Zhengdeng Lei
- Center for Research Bioinformatics, University of Illinois at Chicago, Chicago, IL, USA
| | - Leianne Torres
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven Roth
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA. .,Department of Ophthalmology, University of Illinois at Chicago, Chicago, IL, USA. .,Department of Anesthesiology, MC 515, University of Illinois Medical Center, 1740 West Taylor Street, Chicago, IL, 60612, USA.
| |
Collapse
|
14
|
Tu Y, Zhang L, Tong L, Wang Y, Zhang S, Wang R, Li L, Wang Z. EFhd2/swiprosin-1 regulates LPS-induced macrophage recruitment via enhancing actin polymerization and cell migration. Int Immunopharmacol 2017; 55:263-271. [PMID: 29288926 DOI: 10.1016/j.intimp.2017.12.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/22/2017] [Accepted: 12/23/2017] [Indexed: 12/25/2022]
Abstract
Macrophage motility is vital in innate immunity, which contributes strategically to the defensive inflammation process. During bacterial infection, lipopolysaccharide (LPS) potently activates the migration of macrophages via the NF-κB/iNOS/c-Src signaling pathway. However, the downstream region of c-Src that participates in macrophage migration is unclear. EFhd2, a novel actin bundling protein, was evaluated for its role in LPS-stimulated macrophage migration in this study. We found that LPS stimulated the up-regulation, tyrosine phosphorylation and membrane translocation of EFhd2 in macrophages. The absence of EFhd2 inhibited the recruitment of macrophages in the lungs of LPS-induced septic mice. LPS-induced macrophage migration was neutralized by the deletion of EFhd2. EFhd2-mediated up-regulation of NFPs (including Rac1/Cdc42, N-WASP/WAVE2 and Arp2/3 complex) induced by LPS could be used to explain the role of EFhd2 in promoting actin polymerization. Furthermore, the purified EFhd2 could directly promote actin polymerization in vitro. Dasatinib, a c-Src specific inhibitor, inhibited the up-regulation of EFhd2 stimulated by LPS. Therefore, our study demonstrated that EFhd2 might be involved in LPS-stimulated macrophage migration, which provides a potential target for LPS-activated c-Src during macrophage mobilization.
Collapse
Affiliation(s)
- Ye Tu
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China; Department of Medical Department, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Lichao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Lingchang Tong
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yue Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Su Zhang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Rongmei Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ling Li
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Zhibin Wang
- Department of Pharmacology, College of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
15
|
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
|
16
|
Castellanos-Rubio A, Kratchmarov R, Sebastian M, Garcia-Etxebarria K, Garcia L, Irastorza I, Ghosh S. Cytoplasmic Form of Carlr lncRNA Facilitates Inflammatory Gene Expression upon NF-κB Activation. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28626066 DOI: 10.4049/jimmunol.1700023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical regulators of inflammation. To further understand the interaction between inflammatory signaling pathways and lncRNAs, we characterized the function of cardiac and apoptosis-related lncRNA (Carlr), an lncRNA expressed in both mouse and human cells of diverse tissues. Carlr expression is increased following NF-κB signaling in macrophages, with concomitant translocation to, and enrichment of, the transcript in the cytoplasm. Knockdown of Carlr results in impaired expression of NF-κB pathway genes and influences the interaction between macrophages and intestinal cells in an inflammatory environment. In human celiac disease patient samples, increased levels of the Carlr transcript were detected in the cytoplasm, alongside elevated expression of NF-κB pathway genes. These findings suggest that increased Carlr expression and/or cytoplasmic localization is required for efficient NF-κB signaling and is associated with the inflamed tissue state observed in human celiac disease.
Collapse
Affiliation(s)
- Ainara Castellanos-Rubio
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032; .,Immunogenetics Research Laboratory, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain.,BioCruces Health Research Institute, 48903 Barakaldo, Spain; and
| | - Radomir Kratchmarov
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Maialen Sebastian
- Immunogenetics Research Laboratory, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain.,BioCruces Health Research Institute, 48903 Barakaldo, Spain; and
| | - Koldo Garcia-Etxebarria
- Immunogenetics Research Laboratory, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain.,BioCruces Health Research Institute, 48903 Barakaldo, Spain; and
| | - Liher Garcia
- Immunogenetics Research Laboratory, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain.,BioCruces Health Research Institute, 48903 Barakaldo, Spain; and
| | - Iñaki Irastorza
- Pediatric Gastroenterology Unit, Cruces University Hospital, University of the Basque Country, UPV/EHU, 48903 Barakaldo 48903, Spain
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| |
Collapse
|
17
|
Acuff NV, Li X, Elmore J, Rada B, Watford WT. Tpl2 promotes neutrophil trafficking, oxidative burst, and bacterial killing. J Leukoc Biol 2017; 101:1325-1333. [PMID: 28356348 DOI: 10.1189/jlb.3a0316-146r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 02/03/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022] Open
Abstract
Tumor progression locus 2 (Tpl2) is a serine/threonine kinase that promotes inflammatory cytokine production by activating the MEK/ERK pathway. Tpl2 has been shown to be important for eliciting the inflammatory properties of macrophages; however, there is relatively little known about the contribution of Tpl2 to neutrophil effector functions. This is an important consideration, as neutrophils provide the first line of defense against infection in the innate immune system. We found that Tpl2 is expressed in both human and murine neutrophils, suggesting a potential function for Tpl2 in this lineage. Despite significantly higher proportions of bone marrow (BM) neutrophils in Tpl2-deficient (Tpl2-/- ) mice compared with wild-type (WT) mice, Tpl2-/- mice have significantly reduced proportions of circulating neutrophils. Tpl2-/- neutrophils show impaired recruitment to thioglycollate, which was primarily a result of neutrophil-extrinsic factors in the host. In response to infection, neutrophils secrete inflammatory cytokines and produce reactive oxygen species (ROS), which promote bacterial killing. Tpl2 ablation impaired neutrophil TNF secretion in response to LPS stimulation, superoxide generation in response to the chemotactic peptide fMLP, and killing of the extracellular bacterium, Citrobacter rodentium, despite normal bacterial phagocytosis. These results implicate Tpl2 in the regulation of multiple neutrophil antimicrobial pathways, including inflammatory cytokine secretion and oxidative burst. Furthermore, they indicate that Tpl2 functions early during infection to bolster neutrophil-mediated innate immunity against extracellular bacteria.
Collapse
Affiliation(s)
- Nicole V Acuff
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Xin Li
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Jessica Elmore
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Wendy T Watford
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
18
|
Fenn AM, Swirski FK. Monocytosis, Hypercholesterolemia, and the Kinase That Binds Them. Arterioscler Thromb Vasc Biol 2017; 37:173-175. [DOI: 10.1161/atvbaha.116.308768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ashley M. Fenn
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Filip K. Swirski
- From the Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
19
|
Familial Mediterranean fever mutations lift the obligatory requirement for microtubules in Pyrin inflammasome activation. Proc Natl Acad Sci U S A 2016; 113:14384-14389. [PMID: 27911804 DOI: 10.1073/pnas.1613156113] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease worldwide. It is caused by mutations in the inflammasome adaptor Pyrin, but how FMF mutations alter signaling in FMF patients is unknown. Herein, we establish Clostridium difficile and its enterotoxin A (TcdA) as Pyrin-activating agents and show that wild-type and FMF Pyrin are differentially controlled by microtubules. Diverse microtubule assembly inhibitors prevented Pyrin-mediated caspase-1 activation and secretion of IL-1β and IL-18 from mouse macrophages and human peripheral blood mononuclear cells (PBMCs). Remarkably, Pyrin inflammasome activation persisted upon microtubule disassembly in PBMCs of FMF patients but not in cells of patients afflicted with other autoinflammatory diseases. We further demonstrate that microtubules control Pyrin activation downstream of Pyrin dephosphorylation and that FMF mutations enable microtubule-independent assembly of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) micrometer-sized perinuclear structures (specks). The discovery that Pyrin mutations remove the obligatory requirement for microtubules in inflammasome activation provides a conceptual framework for understanding FMF and enables immunological screening of FMF mutations.
Collapse
|
20
|
Gabai VL, Yaglom JA, Wang Y, Meng L, Shao H, Kim G, Colvin T, Gestwicki J, Sherman MY. Anticancer Effects of Targeting Hsp70 in Tumor Stromal Cells. Cancer Res 2016; 76:5926-5932. [PMID: 27503927 DOI: 10.1158/0008-5472.can-16-0800] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/12/2016] [Indexed: 12/27/2022]
Abstract
The stress-induced chaperone protein Hsp70 enables the initiation and progression of many cancers, making it an appealing therapeutic target for development. Here, we show that cancer cells resistant to Hsp70 inhibitors in vitro remain sensitive to them in vivo, revealing the pathogenic significance of Hsp70 in tumor stromal cells rather than tumor cells as widely presumed. Using transgenic mouse models of cancer, we found that expression of Hsp70 in host stromal cells was essential to support tumor growth. Furthermore, genetic ablation or pharmacologic inhibition of Hsp70 suppressed tumor infiltration by macrophages needed to enable tumor growth. Overall, our results illustrate how Hsp70 inhibitors mediate the anticancer effects by targeting both tumor cells and tumor stromal cells, with implications for the broad use of these inhibitors as tools to ablate tumor-associated macrophages that enable malignant progression. Cancer Res; 76(20); 5926-32. ©2016 AACR.
Collapse
Affiliation(s)
- Vladimir L Gabai
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts. CureLab Oncology, Needham, Massachusetts
| | - Julia A Yaglom
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Yongmei Wang
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Le Meng
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts. Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, Massachusetts
| | - Hao Shao
- Department of Pharmaceutical Chemistry and Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, California
| | - Geunwon Kim
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Teresa Colvin
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Jason Gestwicki
- Department of Pharmaceutical Chemistry and Institute for Neurodegenerative Disease, University of California at San Francisco, San Francisco, California
| | - Michael Y Sherman
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
21
|
Kannan Y, Perez-Lloret J, Li Y, Entwistle LJ, Khoury H, Papoutsopoulou S, Mahmood R, Mansour NR, Ching-Cheng Huang S, Pearce EJ, Pedro S. de Carvalho L, Ley SC, Wilson MS. TPL-2 Regulates Macrophage Lipid Metabolism and M2 Differentiation to Control TH2-Mediated Immunopathology. PLoS Pathog 2016; 12:e1005783. [PMID: 27487182 PMCID: PMC4972396 DOI: 10.1371/journal.ppat.1005783] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/30/2016] [Indexed: 01/05/2023] Open
Abstract
Persistent TH2 cytokine responses following chronic helminth infections can often lead to the development of tissue pathology and fibrotic scarring. Despite a good understanding of the cellular mechanisms involved in fibrogenesis, there are very few therapeutic options available, highlighting a significant medical need and gap in our understanding of the molecular mechanisms of TH2-mediated immunopathology. In this study, we found that the Map3 kinase, TPL-2 (Map3k8; Cot) regulated TH2-mediated intestinal, hepatic and pulmonary immunopathology following Schistosoma mansoni infection or S. mansoni egg injection. Elevated inflammation, TH2 cell responses and exacerbated fibrosis in Map3k8–/–mice was observed in mice with myeloid cell-specific (LysM) deletion of Map3k8, but not CD4 cell-specific deletion of Map3k8, indicating that TPL-2 regulated myeloid cell function to limit TH2-mediated immunopathology. Transcriptional and metabolic assays of Map3k8–/–M2 macrophages identified that TPL-2 was required for lipolysis, M2 macrophage activation and the expression of a variety of genes involved in immuno-regulatory and pro-fibrotic pathways. Taken together this study identified that TPL-2 regulated TH2-mediated inflammation by supporting lipolysis and M2 macrophage activation, preventing TH2 cell expansion and downstream immunopathology and fibrosis. Chronic helminth infections can cause significant morbidity and organ damage in their definitive mammalian hosts. Managing this collateral damage can reduce morbidity and preserve vital tissues for normal organ function. One particular consequence of some chronic helminth infections is the deposition of fibrotic scar tissue, following immune responses directed towards helminth material. In this study we tested the role of a particular signalling kinase, TPL-2, and identified that it critically regulated the magnitude of fibrotic scarring following infection. Using several murine models with genetic deletions of TPL-2 in either all cells or specific deletion in subsets of immune cells (Map3k8–/–Map3k8fl/fl) we identified that expression of TPL-2 in myeloid cells was essential to prevent severe immune-mediated pathology. Using genome-wide analyses and metabolic assays, we discovered that TPL-2 was required for normal lipid metabolism and appropriate activation of myeloid cells / macrophages to limit fibrosis. These results revealed a previously unappreciated role for TPL-2 in preventing severe pathology following infection. Thus, activating this pathway may limit immune mediated pathology following chronic helminth infection. More broadly, this pathway is being targeted to treat inflammatory diseases and cancer [1, 2]. This study would suggest that caution should be taken to prevent untoward co-morbidities and fibrosis-related pathologies in patients when targeting TPL-2.
Collapse
Affiliation(s)
- Yashaswini Kannan
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jimena Perez-Lloret
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Yanda Li
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Lewis J. Entwistle
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Hania Khoury
- Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Radma Mahmood
- Experimental Histopathology, Mill Hill Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nuha R. Mansour
- Department of Infection and Immunity, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stanley Ching-Cheng Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Edward J. Pearce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Luiz Pedro S. de Carvalho
- Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Steven C. Ley
- Immune Cell Signaling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mark S. Wilson
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
Li X, Acuff NV, Peeks AR, Kirkland R, Wyatt KD, Nagy T, Watford WT. Tumor Progression Locus 2 (Tpl2) Activates the Mammalian Target of Rapamycin (mTOR) Pathway, Inhibits Forkhead Box P3 (FoxP3) Expression, and Limits Regulatory T Cell (Treg) Immunosuppressive Functions. J Biol Chem 2016; 291:16802-15. [PMID: 27261457 DOI: 10.1074/jbc.m116.718783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Indexed: 11/06/2022] Open
Abstract
The serine/threonine kinase tumor progression locus 2 (Tpl2, also known as Map3k8/Cot) is a potent inflammatory mediator that drives the production of TNFα, IL-1β, and IFNγ. We previously demonstrated that Tpl2 regulates T cell receptor (TCR) signaling and modulates T helper cell differentiation. However, very little is known about how Tpl2 modulates the development of regulatory T cells (Tregs). Tregs are a specialized subset of T cells that express FoxP3 and possess immunosuppressive properties to limit excess inflammation. Because of the documented role of Tpl2 in promoting inflammation, we hypothesized that Tpl2 antagonizes Treg development and immunosuppressive function. Here we demonstrate that Tpl2 constrains the development of inducible Tregs. Tpl2(-/-) naïve CD4(+) T cells preferentially develop into FoxP3(+) inducible Tregs in vitro as well as in vivo in a murine model of ovalbumin (OVA)-induced systemic tolerance. Treg biasing of Tpl2(-/-) T cells depended on TCR signal strength and corresponded with reduced activation of the mammalian target of rapamycin (mTOR) pathway. Importantly, Tpl2(-/-) Tregs have basally increased expression of FoxP3 and immunosuppressive molecules, IL-10 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). Furthermore, they were more immunosuppressive in vivo in a T cell transfer model of colitis, as evidenced by reduced effector T cell accumulation, systemic production of inflammatory cytokines, and colonic inflammation. These results demonstrate that Tpl2 promotes inflammation in part by constraining FoxP3 expression and Treg immunosuppressive functions. Overall, these findings suggest that Tpl2 inhibition could be used to preferentially drive Treg induction and thereby limit inflammation in a variety of autoimmune diseases.
Collapse
Affiliation(s)
- Xin Li
- From the Departments of Infectious Diseases and
| | | | | | | | | | - Tamas Nagy
- Pathology, University of Georgia, Athens, Georgia 30602-7387
| | | |
Collapse
|
23
|
Kuriakose T, Tripp RA, Watford WT. Tumor Progression Locus 2 Promotes Induction of IFNλ, Interferon Stimulated Genes and Antigen-Specific CD8+ T Cell Responses and Protects against Influenza Virus. PLoS Pathog 2015; 11:e1005038. [PMID: 26241898 PMCID: PMC4524623 DOI: 10.1371/journal.ppat.1005038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 06/22/2015] [Indexed: 11/18/2022] Open
Abstract
Mitogen-activated protein kinase (MAP) cascades are important in antiviral immunity through their regulation of interferon (IFN) production as well as virus replication. Although the serine-threonine MAP kinase tumor progression locus 2 (Tpl2/MAP3K8) has been implicated as a key regulator of Type I (IFNα/β) and Type II (IFNγ) IFNs, remarkably little is known about how Tpl2 might contribute to host defense against viruses. Herein, we investigated the role of Tpl2 in antiviral immune responses against influenza virus. We demonstrate that Tpl2 is an integral component of multiple virus sensing pathways, differentially regulating the induction of IFNα/β and IFNλ in a cell-type specific manner. Although Tpl2 is important in the regulation of both IFNα/β and IFNλ, only IFNλ required Tpl2 for its induction during influenza virus infection both in vitro and in vivo. Further studies revealed an unanticipated function for Tpl2 in transducing Type I IFN signals and promoting expression of interferon-stimulated genes (ISGs). Importantly, Tpl2 signaling in nonhematopoietic cells is necessary to limit early virus replication. In addition to early innate alterations, impaired expansion of virus-specific CD8+ T cells accompanied delayed viral clearance in Tpl2-/- mice at late time points. Consistent with its critical role in facilitating both innate and adaptive antiviral responses, Tpl2 is required for restricting morbidity and mortality associated with influenza virus infection. Collectively, these findings establish an essential role for Tpl2 in antiviral host defense mechanisms.
Collapse
Affiliation(s)
- Teneema Kuriakose
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Wendy T. Watford
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
24
|
del Reino P, Alsina-Beauchamp D, Escós A, Cerezo-Guisado MI, Risco A, Aparicio N, Zur R, Fernandez-Estévez M, Collantes E, Montans J, Cuenda A. Pro-Oncogenic Role of Alternative p38 Mitogen-Activated Protein Kinases p38γ and p38δ, Linking Inflammation and Cancer in Colitis-Associated Colon Cancer. Cancer Res 2014; 74:6150-60. [DOI: 10.1158/0008-5472.can-14-0870] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|