51
|
Lim R, Barker G, Lappas M. IRF5 is increased in labouring myometrium and regulates pro-labour mediators. Reproduction 2018; 156:207-218. [PMID: 30006439 DOI: 10.1530/rep-18-0140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/12/2018] [Indexed: 01/16/2023]
Abstract
Preterm birth continues to be the leading cause of neonatal mortality and morbidities that can extend into adult life. Few treatment options stem from our incomplete understanding of the mechanisms of human labour and delivery. Activation of the inflammatory response in gestational tissues by inflammation and/or infection leads to the production of pro-inflammatory and pro-labour mediators, thus preterm birth. Interferon regulatory factor 5 (IRF5) has recently emerged as an important pro-inflammatory transcription factor involved in acute and chronic inflammation. The aims of this study were to determine the expression of IRF5 in human myometrium from labouring and non-labouring women, and whether IRF5 is involved in the genesis of pro-inflammatory and pro-labour mediators induced by pro-inflammatory cytokines or toll-like receptor (TLR) ligands. IRF5 mRNA and protein expression was significantly higher in human myometrium after spontaneous term labour, compared to non-labouring tissues. IRF5 mRNA expression was also significantly higher in primary myometrial cells treated with the pro-inflammatory cytokines IL1B or TNF. In primary myometrial cells, IRF5 knockdown by siRNA (siIRF5) was associated with significantly decreased expression and or secretion of pro-inflammatory cytokines (IL1A, IL6), chemokines (CXCL8, CCL2), adhesion molecules (ICAM1, VCAM1) and contraction-associated proteins PTGS2, PGF2α and PTGFR when in the presence of IL1B, TNF, fsl-1 (TLR2/6 ligand) or flagellin (TLR5 ligand). siIRF5-transfected cells also displayed decreased NF-κB RELA transcriptional activity in the presence of these preterm birth mediators. Our study suggests a novel role for IRF5 in the regulation of the inflammatory response in human myometrium.
Collapse
Affiliation(s)
- Ratana Lim
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Gillian Barker
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Victoria, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
52
|
Schneider A, Weier M, Herderschee J, Perreau M, Calandra T, Roger T, Giannoni E. IRF5 Is a Key Regulator of Macrophage Response to Lipopolysaccharide in Newborns. Front Immunol 2018; 9:1597. [PMID: 30050534 PMCID: PMC6050365 DOI: 10.3389/fimmu.2018.01597] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022] Open
Abstract
Infections are a leading cause of mortality and morbidity in newborns. The high susceptibility of newborns to infection has been associated with a limited capacity to mount protective immune responses. Monocytes and macrophages are involved in the initiation, amplification, and termination of immune responses. Depending on cues received from their environment, monocytes differentiate into M1 or M2 macrophages with proinflammatory or anti-inflammatory and tissue repair properties, respectively. The purpose of this study was to characterize differences in monocyte to macrophage differentiation and polarization between newborns and adults. Monocytes from umbilical cord blood of healthy term newborns and from peripheral blood of adult healthy subjects were exposed to GM-CSF or M-CSF to induce M1 or M2 macrophages. Newborn monocytes differentiated into M1 and M2 macrophages with similar morphology and expression of differentiation/polarization markers as adult monocytes, with the exception of CD163 that was expressed at sevenfold higher levels in newborn compared to adult M1 macrophages. Upon TLR4 stimulation, newborn M1 macrophages produced threefold to sixfold lower levels of TNF than adult macrophages, while production of IL-1-β, IL-6, IL-8, IL-10, and IL-23 was at similar levels as in adults. Nuclear levels of IRF5, a transcription factor involved in M1 polarization, were markedly reduced in newborns, whereas the NF-κB and MAP kinase pathways were not altered. In line with a functional role for IRF5, adenoviral-mediated IRF5 overexpression in newborn M1 macrophages restored lipopolysaccharide-induced TNF production. Altogether, these data highlight a distinct immune response of newborn macrophages and identify IRF5 as a key regulator of macrophage TNF response in newborns.
Collapse
Affiliation(s)
- Anina Schneider
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Manuela Weier
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Jacobus Herderschee
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Eric Giannoni
- Clinic of Neonatology, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
53
|
Stein T, Wollschlegel A, Te H, Weiss J, Joshi K, Kinzel B, Billich A, Guntermann C, Lehmann JCU. Interferon regulatory factor 5 and nuclear factor kappa‐B exhibit cooperating but also divergent roles in the regulation of pro‐inflammatory cytokines important for the development of
TH
1 and
TH
17 responses. FEBS J 2018; 285:3097-3113. [DOI: 10.1111/febs.14600] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 05/16/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Thomas Stein
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Audrey Wollschlegel
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Helene Te
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Jessica Weiss
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Kushal Joshi
- Department of Chemical Biology & Therapeutics Novartis Institutes for Biomedical Research Novartis Pharma AG Cambridge MA USA
| | - Bernd Kinzel
- Department of Chemical Biology & Therapeutics Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Andreas Billich
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Christine Guntermann
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| | - Joachim C. U. Lehmann
- Department of Autoimmunity, Transplantation & Inflammation Novartis Institutes for Biomedical Research Novartis Pharma AG Basel Switzerland
| |
Collapse
|
54
|
Liu Y, Xie R, Li H, Hu Z, Wang S, Yin Z. Expression, polymorphism of IRF5 gene and association with serum cytokine levels in pig. ITALIAN JOURNAL OF ANIMAL SCIENCE 2018. [DOI: 10.1080/1828051x.2017.1420431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Yang Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Rui Xie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hejun Li
- Shanghai Animal Disease Control Center, Shanghai, China
| | - Zhengzheng Hu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Shiwei Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
55
|
Wu J, Wang Q, Dai W, Wang W, Yue M, Wang J. Massive GGAAs in genomic repetitive sequences serve as a nuclear reservoir of NF-κB. J Genet Genomics 2018; 45:193-203. [PMID: PMID : 29748061 DOI: 10.1016/j.jgg.2018.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/01/2022]
Abstract
Nuclear factor κB (NF-κB) is a DNA-binding transcription factor. Characterizing its genomic binding sites is crucial for understanding its gene regulatory function and mechanism in cells. This study characterized the binding sites of NF-κB RelA/p65 in the tumor neurosis factor-α (TNFα) stimulated HeLa cells by a precise chromatin immunoprecipitation-sequencing (ChIP-seq). The results revealed that NF-κB binds nontraditional motifs (nt-motifs) containing conserved GGAA quadruplet. Moreover, nt-motifs mainly distribute in the peaks nearby centromeres that contain a larger number of repetitive elements such as satellite, simple repeats and short interspersed nuclear elements (SINEs). This intracellular binding pattern was then confirmed by the in vitro detection, indicating that NF-κB dimers can bind the nontraditional κB (nt-κB) sites with low affinity. However, this binding hardly activates transcription. This study thus deduced that NF-κB binding nt-motifs may realize functions other than gene regulation as NF-κB binding traditional motifs (t-motifs). To testify the deduction, many ChIP-seq data of other cell lines were then analyzed. The results indicate that NF-κB binding nt-motifs is also widely present in other cells. The ChIP-seq data analysis also revealed that nt-motifs more widely distribute in the peaks with low-fold enrichment. Importantly, it was also found that NF-κB binding nt-motifs is mainly present in the resting cells, whereas NF-κB binding t-motifs is mainly present in the stimulated cells. Astonishingly, no known function was enriched by the gene annotation of nt-motif peaks. Based on these results, this study proposed that the nt-κB sites that extensively distribute in larger numbers of repeat elements function as a nuclear reservoir of NF-κB. The nuclear NF-κB proteins stored at nt-κB sites in the resting cells may be recruited to the t-κB sites for regulating its target genes upon stimulation.
Collapse
Affiliation(s)
- Jian Wu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Qiao Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Ming Yue
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China.
| |
Collapse
|
56
|
De S, Zhang B, Shih T, Singh S, Winkler A, Donnelly R, Barnes BJ. B Cell-Intrinsic Role for IRF5 in TLR9/BCR-Induced Human B Cell Activation, Proliferation, and Plasmablast Differentiation. Front Immunol 2018; 8:1938. [PMID: 29367853 PMCID: PMC5768180 DOI: 10.3389/fimmu.2017.01938] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Upon recognition of antigen, B cells undergo rapid proliferation followed by differentiation to specialized antibody secreting cells (ASCs). During this transition, B cells are reliant upon a multilayer transcription factor network to achieve a dramatic remodeling of the B cell transcriptional landscape. Increased levels of ASCs are often seen in autoimmune diseases and it is believed that altered expression of regulatory transcription factors play a role in this imbalance. The transcription factor interferon regulatory factor 5 (IRF5) is one such candidate as polymorphisms in IRF5 associate with risk of numerous autoimmune diseases and correlate with elevated IRF5 expression. IRF5 genetic risk has been widely replicated in systemic lupus erythematosus (SLE), and loss of Irf5 ameliorates disease in murine lupus models, in part, through the lack of pathogenic autoantibody secretion. It remains unclear, however, whether IRF5 is contributing to autoantibody production through a B cell-intrinsic function. To date, IRF5 function in healthy human B cells has not been characterized. Using human primary naive B cells, we define a critical intrinsic role for IRF5 in B cell activation, proliferation, and plasmablast differentiation. Targeted IRF5 knockdown resulted in significant immunoglobulin (Ig) D retention, reduced proliferation, plasmablast differentiation, and IgG secretion. The observed decreases were due to impaired B cell activation and clonal expansion. Distinct from murine studies, we identify and confirm new IRF5 target genes, IRF4, ERK1, and MYC, and pathways that mediate IRF5 B cell-intrinsic function. Together, these results identify IRF5 as an early regulator of human B cell activation and provide the first dataset in human primary B cells to map IRF5 dysfunction in SLE.
Collapse
Affiliation(s)
- Saurav De
- Rutgers Graduate School of Biomedical Sciences, Newark, NJ, United States.,Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Baohong Zhang
- Clinical Genetics and Bioinformatics, Pfizer Inc., Cambridge, MA, United States
| | - Tiffany Shih
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Sukhwinder Singh
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Aaron Winkler
- Department of Inflammation and Immunology, Pfizer Inc., Cambridge, MA, United States
| | - Robert Donnelly
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY, United States.,Rutgers Biomedical and Health Sciences, New Jersey Medical School-Cancer Center, Newark, NJ, United States
| |
Collapse
|
57
|
Amici SA, Dong J, Guerau-de-Arellano M. Molecular Mechanisms Modulating the Phenotype of Macrophages and Microglia. Front Immunol 2017; 8:1520. [PMID: 29176977 PMCID: PMC5686097 DOI: 10.3389/fimmu.2017.01520] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/26/2017] [Indexed: 12/24/2022] Open
Abstract
Macrophages and microglia play crucial roles during central nervous system development, homeostasis and acute events such as infection or injury. The diverse functions of tissue macrophages and microglia are mirrored by equally diverse phenotypes. A model of inflammatory/M1 versus a resolution phase/M2 macrophages has been widely used. However, the complexity of macrophage function can only be achieved by the existence of varied, plastic and tridimensional macrophage phenotypes. Understanding how tissue macrophages integrate environmental signals via molecular programs to define pathogen/injury inflammatory responses provides an opportunity to better understand the multilayered nature of macrophages, as well as target and modulate cellular programs to control excessive inflammation. This is particularly important in MS and other neuroinflammatory diseases, where chronic inflammatory macrophage and microglial responses may contribute to pathology. Here, we perform a comprehensive review of our current understanding of how molecular pathways modulate tissue macrophage phenotype, covering both classic pathways and the emerging role of microRNAs, receptor-tyrosine kinases and metabolism in macrophage phenotype. In addition, we discuss pathway parallels in microglia, novel markers helpful in the identification of peripheral macrophages versus microglia and markers linked to their phenotype.
Collapse
Affiliation(s)
- Stephanie A Amici
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Joycelyn Dong
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,McCormick School of Engineering, Division of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
58
|
Denisenko E, Guler R, Mhlanga MM, Suzuki H, Brombacher F, Schmeier S. Genome-wide profiling of transcribed enhancers during macrophage activation. Epigenetics Chromatin 2017; 10:50. [PMID: 29061167 PMCID: PMC5654053 DOI: 10.1186/s13072-017-0158-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/13/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Macrophages are sentinel cells essential for tissue homeostasis and host defence. Owing to their plasticity, macrophages acquire a range of functional phenotypes in response to microenvironmental stimuli, of which M(IFN-γ) and M(IL-4/IL-13) are well known for their opposing pro- and anti-inflammatory roles. Enhancers have emerged as regulatory DNA elements crucial for transcriptional activation of gene expression. RESULTS Using cap analysis of gene expression and epigenetic data, we identify on large-scale transcribed enhancers in bone marrow-derived mouse macrophages, their time kinetics, and target protein-coding genes. We observe an increase in target gene expression, concomitant with increasing numbers of associated enhancers, and find that genes associated with many enhancers show a shift towards stronger enrichment for macrophage-specific biological processes. We infer enhancers that drive transcriptional responses of genes upon M(IFN-γ) and M(IL-4/IL-13) macrophage activation and demonstrate stimuli specificity of regulatory associations. Finally, we show that enhancer regions are enriched for binding sites of inflammation-related transcription factors, suggesting a link between stimuli response and enhancer transcriptional control. CONCLUSIONS Our study provides new insights into genome-wide enhancer-mediated transcriptional control of macrophage genes, including those implicated in macrophage activation, and offers a detailed genome-wide catalogue of transcribed enhancers in bone marrow-derived mouse macrophages.
Collapse
Affiliation(s)
- Elena Denisenko
- Institute of Natural and Mathematical Sciences, Massey University, Albany, Auckland, 0632 New Zealand
| | - Reto Guler
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925 South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, 7925 South Africa
| | - Musa M. Mhlanga
- Gene Expression and Biophysics Group, CSIR Synthetic Biology ERA, Pretoria, 0001 South Africa
- Division of Chemical Systems and Synthetic Biology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, 7925 South Africa
- Gene Expression and Biophysics Unit, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Harukazu Suzuki
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045 Japan
| | - Frank Brombacher
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925 South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, 7925 South Africa
| | - Sebastian Schmeier
- Institute of Natural and Mathematical Sciences, Massey University, Albany, Auckland, 0632 New Zealand
| |
Collapse
|
59
|
Treuter E, Fan R, Huang Z, Jakobsson T, Venteclef N. Transcriptional repression in macrophages-basic mechanisms and alterations in metabolic inflammatory diseases. FEBS Lett 2017; 591:2959-2977. [DOI: 10.1002/1873-3468.12850] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Eckardt Treuter
- Department of Biosciences and Nutrition; Center for Innovative Medicine (CIMED); Karolinska Institutet; Huddinge Sweden
| | - Rongrong Fan
- Department of Biosciences and Nutrition; Center for Innovative Medicine (CIMED); Karolinska Institutet; Huddinge Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition; Center for Innovative Medicine (CIMED); Karolinska Institutet; Huddinge Sweden
| | - Tomas Jakobsson
- Department of Laboratory Medicine; Karolinska Institutet; Huddinge Sweden
| | - Nicolas Venteclef
- UMR_S 1138 Cordeliers Research; Institut National de la Santé et de la Recherche Médicale (INSERM); Sorbonne Universités; Université Pierre et Marie-Curie; Paris France
| |
Collapse
|
60
|
Cushing L, Winkler A, Jelinsky SA, Lee K, Korver W, Hawtin R, Rao VR, Fleming M, Lin LL. IRAK4 kinase activity controls Toll-like receptor-induced inflammation through the transcription factor IRF5 in primary human monocytes. J Biol Chem 2017; 292:18689-18698. [PMID: 28924041 DOI: 10.1074/jbc.m117.796912] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/22/2017] [Indexed: 12/20/2022] Open
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in innate immune signaling by Toll-like receptors (TLRs), and loss of IRAK4 activity in mice and humans increases susceptibility to bacterial infections and causes defects in TLR and IL1 ligand sensing. However, the mechanism by which IRAK4 activity regulates the production of downstream inflammatory cytokines is unclear. Using transcriptomic and biochemical analyses of human monocytes treated with a highly potent and selective inhibitor of IRAK4, we show that IRAK4 kinase activity controls the activation of interferon regulatory factor 5 (IRF5), a transcription factor implicated in the pathogenesis of multiple autoimmune diseases. Following TLR7/8 stimulation by its agonist R848, chemical inhibition of IRAK4 abolished IRF5 translocation to the nucleus and thus prevented IRF5 binding to and activation of the promoters of inflammatory cytokines in human monocytes. We also found that IKKβ, an upstream IRF5 activator, is phosphorylated in response to the agonist-induced TLR signaling. Of note, IRAK4 inhibition blocked IKKβ phosphorylation but did not block the nuclear translocation of NFκB, which was surprising, given the canonical role of IKKβ in phosphorylating IκB to allow NFκB activation. Moreover, pharmacological inhibition of either IKKβ or the serine/threonine protein kinase TAK1 in monocytes blocked TLR-induced cytokine production and IRF5 translocation to the nucleus, but not nuclear translocation of NFκB. Taken together, our data suggest a mechanism by which IRAK4 activity regulates TAK1 and IKKβ activation, leading to the nuclear translocation of IRF5 and induction of inflammatory cytokines in human monocytes.
Collapse
Affiliation(s)
- Leah Cushing
- From the Departments of Inflammation and Immunology and
| | - Aaron Winkler
- From the Departments of Inflammation and Immunology and
| | | | - Katherine Lee
- Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139 and
| | - Wouter Korver
- Nodality Inc., South San Francisco, California 94080
| | | | - Vikram R Rao
- From the Departments of Inflammation and Immunology and
| | | | - Lih-Ling Lin
- From the Departments of Inflammation and Immunology and
| |
Collapse
|
61
|
Potenza N, Panella M, Castiello F, Mosca N, Amendola E, Russo A. Molecular mechanisms governing microRNA-125a expression in human hepatocellular carcinoma cells. Sci Rep 2017; 7:10712. [PMID: 28878257 PMCID: PMC5587745 DOI: 10.1038/s41598-017-11418-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/24/2017] [Indexed: 12/13/2022] Open
Abstract
MicroRNA-125a-5p (miR-125a) is a vertebrate homolog of lin-4, the first discovered microRNA, and plays a fundamental role in embryo development by downregulating Lin-28 protein. MiR-125a is also expressed in differentiated cells where it generally acts as an antiproliferative factor by targeting membrane receptors or intracellular transductors of mitogenic signals. MiR-125a expression is downregulated in several tumors, including hepatocellular carcinoma (HCC) where it targets sirtuin-7, matrix metalloproteinase-11, VEGF-A, Zbtb7a, and c-Raf. In this study, we have isolated the transcription promoter of human miR-125a and characterized its activity in HCC cells. It is a TATA-less Pol II promoter provided with an initiator element and a downstream promoter element, located 3939 bp upstream the genomic sequence of the miRNA. The activity of the promoter is increased by the transcription factor NF-kB, a master regulator of inflammatory response, and miR-125a itself was found to strengthen this activation through inhibition of TNFAIP3, a negative regulator of NF-kB. This finding contributes to explain the increased levels of miR-125a observed in the liver of patients with chronic hepatitis B.
Collapse
Affiliation(s)
- Nicoletta Potenza
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Marta Panella
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Filomena Castiello
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Nicola Mosca
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Elena Amendola
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131, Napoli, Italy
| | - Aniello Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy.
| |
Collapse
|
62
|
Cai H, Yao Z, Li W. IRF-5 accelerates leukocyte adhesion to endothelial cells in ischemia-reperfusion injury through regulating the transcription of VCAM-1. Biochem Biophys Res Commun 2017; 492:192-198. [PMID: 28818665 DOI: 10.1016/j.bbrc.2017.08.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/12/2017] [Indexed: 10/19/2022]
Abstract
Ischemia-reperfusion injury (IRI) has been implicated in many pathological conditions, including cardiovascular diseases. Adhesion of leukocytes to the surface of endothelial cells has been considered as one of the principle steps in the pathological cascade of inflammatory tissue damage during IRI. The role of the transcriptional factor interferon regulatory factor-5 (IRF-5) in endothelial physiology remains unknown. Here, we report that IRF-5 is expressed in human umbilical vein endothelial cells (HUVECs) and is rapidly upregulated in response to IRI, mediated by the JAK2/STAT3 pathway. Importantly, IRF-5 is involved in IRI-induced attachment of THP-1 leukocytes to HUVECs. Mechanistically, it was found that IRF-5 targeted the expression of vascular cell adhesion molecule 1 (VCAM-1) at the transcriptional level by binding to its promoter. In conclusion, we identify IRF-5 as a new regulator and thus a therapeutic target in IRI-driven cardiovascular pathologies.
Collapse
Affiliation(s)
- Hongbin Cai
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China
| | - Zhuhua Yao
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China.
| | - Wenting Li
- Department of Cardiology, Tianjin People's Hospital, Tianjin, 300120, China
| |
Collapse
|
63
|
Seneviratne AN, Edsfeldt A, Cole JE, Kassiteridi C, Swart M, Park I, Green P, Khoyratty T, Saliba D, Goddard ME, Sansom SN, Goncalves I, Krams R, Udalova IA, Monaco C. Interferon Regulatory Factor 5 Controls Necrotic Core Formation in Atherosclerotic Lesions by Impairing Efferocytosis. Circulation 2017; 136:1140-1154. [PMID: 28698173 PMCID: PMC5598917 DOI: 10.1161/circulationaha.117.027844] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/13/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Myeloid cells are central to atherosclerotic lesion development and vulnerable plaque formation. Impaired ability of arterial phagocytes to uptake apoptotic cells (efferocytosis) promotes lesion growth and establishment of a necrotic core. The transcription factor interferon regulatory factor (IRF)-5 is an important modulator of myeloid function and programming. We sought to investigate whether IRF5 affects the formation and phenotype of atherosclerotic lesions. METHODS We investigated the role of IRF5 in atherosclerosis in 2 complementary models. First, atherosclerotic lesion development in hyperlipidemic apolipoprotein E-deficient (ApoE-/-) mice and ApoE-/- mice with a genetic deletion of IRF5 (ApoE-/-Irf5-/-) was compared and then lesion development was assessed in a model of shear stress-modulated vulnerable plaque formation. RESULTS Both lesion and necrotic core size were significantly reduced in ApoE-/-Irf5-/- mice compared with IRF5-competent ApoE-/- mice. Necrotic core size was also reduced in the model of shear stress-modulated vulnerable plaque formation. A significant loss of CD11c+ macrophages was evident in ApoE-/-Irf5-/- mice in the aorta, draining lymph nodes, and bone marrow cell cultures, indicating that IRF5 maintains CD11c+ macrophages in atherosclerosis. Moreover, we revealed that the CD11c gene is a direct target of IRF5 in macrophages. In the absence of IRF5, CD11c- macrophages displayed a significant increase in expression of the efferocytosis-regulating integrin-β3 and its ligand milk fat globule-epidermal growth factor 8 protein and enhanced efferocytosis in vitro and in situ. CONCLUSIONS IRF5 is detrimental in atherosclerosis by promoting the maintenance of proinflammatory CD11c+ macrophages within lesions and controlling the expansion of the necrotic core by impairing efferocytosis.
Collapse
Affiliation(s)
- Anusha N Seneviratne
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Andreas Edsfeldt
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Jennifer E Cole
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Christina Kassiteridi
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Maarten Swart
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Inhye Park
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Patricia Green
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Tariq Khoyratty
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - David Saliba
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Michael E Goddard
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Stephen N Sansom
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Isabel Goncalves
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Rob Krams
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Irina A Udalova
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.)
| | - Claudia Monaco
- From Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, United Kingdom (A.N.S., A.E., J.E.C., C.K., M.S., I.P., P.G., T.K., D.S., M.E.G., S.N.S., I.A.U., C.M.); Department of Bioengineering, Imperial College London, United Kingdom (A.N.S., R.K.); Experimental Cardiovascular Research Unit, Clinical Research Centre, Clinical Sciences Malmö, Lund University, Sweden (A.E., I.G.); Department of Cardiology, Skåne University Hospital, Lund/Malmö, Sweden (A.E., I.G.); and School of Engineering and Materials Science, Queen Mary University of London, United Kingdom (R.K.).
| |
Collapse
|
64
|
Abstract
Tumor necrosis factor (TNF) is one of the most important cytokines produced by macrophages. TNF is a very important component of host defense, released very rapidly after all types of injuries and stimuli. The kinetics of TNF release are short, and so it is perhaps not surprising that prolonged TNF production is associated with pathology. This was first elucidated in rheumatoid arthritis but extends to other chronic inflammatory diseases such as Crohn's disease and psoriasis. In this chapter, the discovery of anti-TNF therapy is reviewed, with its benefit but also its limitations. The potential of anti-TNF therapy in other diseases, e.g., cardiovascular and fibrosis, is discussed, as is the opportunity to define ways of blocking TNF synthesis.
Collapse
|
65
|
Bai Q, Liu L, Xia Y, Wang J, Xi W, Qu Y, Xiong Y, Long Q, Xu J, Guo J. IRF5 is associated with adverse postoperative prognosis of patients with non-metastatic clear cell renal cell carcinoma. Oncotarget 2017; 8:44186-44194. [PMID: 28562332 PMCID: PMC5546472 DOI: 10.18632/oncotarget.17777] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/25/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND IRF5 is one member of IRFs family, and is critical for host immunity and cell response. In the present study, we sought to search the clinical and prognostic value of IFR5 in patients with non-metastatic ccRCC. RESULTS IRF5 proved to be an adverse independent prognostic factor for overall survival (p < 0.001) and recurrence free survival (p = 0.002). The newly built nomograms could give better prediction for overall survival and recurrence free survival in ccRCC patients. MATERIALS AND METHODS We included 264 individuals who were diagnosed with non-metastatic clear cell renal cell carcinoma in the present study. Immunohistochemistry staining was performed on tissue microarrays to evaluate the IRF5 expression. χ2 test, Fisher's exact test, t test, Kaplan-Meier method and Cox proportional hazard model were applied to evaluate the prognostic value of IRF5. Two nomograms were constructed to predict clinical outcomes for ccRCC patients after surgery. CONCLUSIONS IRF5 was an adverse independent prognostic factor for both overall survival and recurrence free survival in patients with non-metastatic ccRCC.
Collapse
Affiliation(s)
- Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiajun Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Xi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yang Qu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Xiong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qilai Long
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
66
|
NOX2-dependent ATM kinase activation dictates pro-inflammatory macrophage phenotype and improves effectiveness to radiation therapy. Cell Death Differ 2017; 24:1632-1644. [PMID: 28574504 DOI: 10.1038/cdd.2017.91] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 04/19/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022] Open
Abstract
Although tumor-associated macrophages have been extensively studied in the control of response to radiotherapy, the molecular mechanisms involved in the ionizing radiation-mediated activation of macrophages remain elusive. Here we show that ionizing radiation induces the expression of interferon regulatory factor 5 (IRF5) promoting thus macrophage activation toward a pro-inflammatory phenotype. We reveal that the activation of the ataxia telangiectasia mutated (ATM) kinase is required for ionizing radiation-elicited macrophage activation, but also for macrophage reprogramming after treatments with γ-interferon, lipopolysaccharide or chemotherapeutic agent (such as cisplatin), underscoring the fact that the kinase ATM plays a central role during macrophage phenotypic switching toward a pro-inflammatory phenotype through the regulation of mRNA level and post-translational modifications of IRF5. We further demonstrate that NADPH oxidase 2 (NOX2)-dependent ROS production is upstream to ATM activation and is essential during this process. We also report that the inhibition of any component of this signaling pathway (NOX2, ROS and ATM) impairs pro-inflammatory activation of macrophages and predicts a poor tumor response to preoperative radiotherapy in locally advanced rectal cancer. Altogether, our results identify a novel signaling pathway involved in macrophage activation that may enhance the effectiveness of radiotherapy through the reprogramming of tumor-infiltrating macrophages.
Collapse
|
67
|
Genes directly regulated by NF-κB in human hepatocellular carcinoma HepG2. Int J Biochem Cell Biol 2017; 89:157-170. [PMID: 28579529 DOI: 10.1016/j.biocel.2017.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/25/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
It has been well-known that over activation of NF-κB has close relationship with hepatitis and hepatocellular carcinoma (HCC). However, the complete and exact underlying molecular pathways and mechanisms still remain not fully understood. By manipulating NF-κB activity with its recognized activator TNFα and using ChIP-seq and RNA-seq techniques, this study identified 699 NF-κB direct target genes (DTGs) in a widely used HCC cell line, HepG2, including 399 activated and 300 repressed genes. In these NF-κB DTGs, 216 genes (126 activated and 90 repressed genes) are among the current HCC gene signature. In comparison with NF-κB target genes identified in LPS-induced THP-1 and TNFα-induced HeLa cells, only limited numbers (24-46) of genes were shared by the two cell lines, indicating the HCC specificity of identified genes. Functional annotation revealed that NF-κB DTGs in HepG2 cell are mainly related with many typical NF-κB-related biological processes including immune system process, response to stress, response to stimulus, defense response, and cell death, and signaling pathways of MAPK, TNF, TGF-beta, Chemokine, NF-kappa B, and Toll-like receptor. Some NF-κB DTGs are also involved in Hepatitis C and B pathways. It was found that 82 NF-κB DTGs code secretory proteins, which include CCL2 and DKK1 that have already been used as HCC markers. Finally, the NF-κB DTGs were further confirmed by detecting the NF-κB binding and expression of 14 genes with ChIP-PCR and RT-PCR. This study thus provides a useful NF-κB DTG list for future studies of NF-κB-related molecular mechanisms and theranostic biomarkers of HCC.
Collapse
|
68
|
Oriss TB, Raundhal M, Morse C, Huff RE, Das S, Hannum R, Gauthier MC, Scholl KL, Chakraborty K, Nouraie SM, Wenzel SE, Ray P, Ray A. IRF5 distinguishes severe asthma in humans and drives Th1 phenotype and airway hyperreactivity in mice. JCI Insight 2017; 2:91019. [PMID: 28515358 PMCID: PMC5436536 DOI: 10.1172/jci.insight.91019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 04/18/2017] [Indexed: 01/07/2023] Open
Abstract
Severe asthma (SA) is a significant problem both clinically and economically, given its poor response to corticosteroids (CS). We recently reported a complex type 1-dominated (IFN-γ-dominated) immune response in more than 50% of severe asthmatics despite high-dose CS treatment. Also, IFN-γ was found to be critical for increased airway hyperreactivity (AHR) in our model of SA. The transcription factor IRF5 expressed in M1 macrophages can induce a Th1/Th17 response in cocultured human T cells. Here we show markedly higher expression of IRF5 in bronchoalveolar lavage (BAL) cells of severe asthmatics as compared with that in cells from milder asthmatics or healthy controls. Using our SA mouse model, we demonstrate that lack of IRF5 in lymph node migratory DCs severely limits their ability to stimulate the generation of IFN-γ- and IL-17-producing CD4+ T cells and IRF5-/- mice subjected to the SA model displayed significantly lower IFN-γ and IL-17 responses, albeit showing a reciprocal increase in Th2 response. However, the absence of IRF5 rendered the mice responsive to CS with suppression of the heightened Th2 response. These data support the notion that IRF5 inhibition in combination with CS may be a viable approach to manage disease in a subset of severe asthmatics.
Collapse
Affiliation(s)
- Timothy B. Oriss
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mahesh Raundhal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Department of Immunology, and
| | - Christina Morse
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Rachael E. Huff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Sudipta Das
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Rachel Hannum
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Marc C. Gauthier
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kathryn L. Scholl
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | - Seyed M. Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Sally E. Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Department of Immunology, and
- University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Department of Immunology, and
- University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Department of Immunology, and
- University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
69
|
S100-alarmin-induced innate immune programming protects newborn infants from sepsis. Nat Immunol 2017; 18:622-632. [PMID: 28459433 DOI: 10.1038/ni.3745] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/11/2017] [Indexed: 12/22/2022]
Abstract
The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.
Collapse
|
70
|
Alzaid F, Lagadec F, Albuquerque M, Ballaire R, Orliaguet L, Hainault I, Blugeon C, Lemoine S, Lehuen A, Saliba DG, Udalova IA, Paradis V, Foufelle F, Venteclef N. IRF5 governs liver macrophage activation that promotes hepatic fibrosis in mice and humans. JCI Insight 2016; 1:e88689. [PMID: 27942586 DOI: 10.1172/jci.insight.88689] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hepatic fibrosis arises from inflammation in the liver initiated by resident macrophage activation and massive leukocyte accumulation. Hepatic macrophages hold a central position in maintaining homeostasis in the liver and in the pathogenesis of acute and chronic liver injury linked to fibrogenesis. Interferon regulatory factor 5 (IRF5) has recently emerged as an important proinflammatory transcription factor involved in macrophage activation under acute and chronic inflammation. Here, we revealed that IRF5 is significantly induced in liver macrophages from human subjects developing liver fibrosis from nonalcoholic fatty liver disease or hepatitis C virus infection. Furthermore, IRF5 expression positively correlated with clinical markers of liver damage, such as plasma transaminase and bilirubin levels. Interestingly, mice lacking IRF5 in myeloid cells (MKO) were protected from hepatic fibrosis induced by metabolic or toxic stresses. Transcriptional reprogramming of macrophages lacking IRF5 was characterized by immunosuppressive and antiapoptotic properties. Consequently, IRF5 MKO mice respond to hepatocellular stress by promoting hepatocyte survival, leading to complete protection from hepatic fibrogenesis. Our findings reveal a regulatory network, governed by IRF5, that mediates hepatocyte death and liver fibrosis in mice and humans. Therefore, modulating IRF5 function may be an attractive approach to experimental therapeutics in fibroinflammatory liver disease.
Collapse
Affiliation(s)
- Fawaz Alzaid
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| | - Floriane Lagadec
- INSERM UMRS 1149 and Department of Pathology Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Miguel Albuquerque
- INSERM UMRS 1149 and Department of Pathology Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Raphaëlle Ballaire
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| | - Lucie Orliaguet
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| | - Isabelle Hainault
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| | - Corinne Blugeon
- École normale supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), INSERM, Institut de Biologie de l'École Normale Supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Sophie Lemoine
- École normale supérieure, PSL Research University, Centre National de la Recherche Scientifique (CNRS), INSERM, Institut de Biologie de l'École Normale Supérieure (IBENS), Plateforme Génomique, Paris, France
| | - Agnès Lehuen
- INSERM UMRS 1016, Institut Cochin, Paris, France; CNRS UMR_S 8104, Paris, France; and Laboratoire d'Excellence INFLAMEX, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - David G Saliba
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Valérie Paradis
- INSERM UMRS 1149 and Department of Pathology Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| | - Nicolas Venteclef
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 1138, Sorbonne Universités, UPMC Université Paris 06; Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot; and Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
71
|
Abstract
Macrophage polarization refers to how macrophages have been activated at a given point in space and time. Polarization is not fixed, as macrophages are sufficiently plastic to integrate multiple signals, such as those from microbes, damaged tissues, and the normal tissue environment. Three broad pathways control polarization: epigenetic and cell survival pathways that prolong or shorten macrophage development and viability, the tissue microenvironment, and extrinsic factors, such as microbial products and cytokines released in inflammation. A plethora of advances have provided a framework for rationally purifying, describing, and manipulating macrophage polarization. Here, I assess the current state of knowledge about macrophage polarization and enumerate the major questions about how activated macrophages regulate the physiology of normal and damaged tissues.
Collapse
Affiliation(s)
- Peter J Murray
- Departments of Infectious Diseases and Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105;
| |
Collapse
|
72
|
Kolovos P, Georgomanolis T, Koeferle A, Larkin JD, Brant L, Nikolicć M, Gusmao EG, Zirkel A, Knoch TA, van Ijcken WF, Cook PR, Costa IG, Grosveld FG, Papantonis A. Binding of nuclear factor κB to noncanonical consensus sites reveals its multimodal role during the early inflammatory response. Genome Res 2016; 26:1478-1489. [PMID: 27633323 PMCID: PMC5088591 DOI: 10.1101/gr.210005.116] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/14/2016] [Indexed: 01/25/2023]
Abstract
Mammalian cells have developed intricate mechanisms to interpret, integrate, and respond to extracellular stimuli. For example, tumor necrosis factor (TNF) rapidly activates proinflammatory genes, but our understanding of how this occurs against the ongoing transcriptional program of the cell is far from complete. Here, we monitor the early phase of this cascade at high spatiotemporal resolution in TNF-stimulated human endothelial cells. NF-κB, the transcription factor complex driving the response, interferes with the regulatory machinery by binding active enhancers already in interaction with gene promoters. Notably, >50% of these enhancers do not encode canonical NF-κB binding motifs. Using a combination of genomics tools, we find that binding site selection plays a key role in NF-κΒ–mediated transcriptional activation and repression. We demonstrate the latter by describing the synergy between NF-κΒ and the corepressor JDP2. Finally, detailed analysis of a 2.8-Mbp locus using sub-kbp-resolution targeted chromatin conformation capture and genome editing uncovers how NF-κΒ that has just entered the nucleus exploits pre-existing chromatin looping to exert its multimodal role. This work highlights the involvement of topology in cis-regulatory element function during acute transcriptional responses, where primary DNA sequence and its higher-order structure constitute a regulatory context leading to either gene activation or repression.
Collapse
Affiliation(s)
- Petros Kolovos
- Department of Cell Biology, Erasmus Medical Centre, 3015 CN Rotterdam, The Netherlands
| | | | - Anna Koeferle
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Joshua D Larkin
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Lilija Brant
- Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Miloš Nikolicć
- Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Eduardo G Gusmao
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, 52062 Aachen, Germany
| | - Anne Zirkel
- Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Tobias A Knoch
- Department of Cell Biology, Erasmus Medical Centre, 3015 CN Rotterdam, The Netherlands
| | | | - Peter R Cook
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE Oxford, United Kingdom
| | - Ivan G Costa
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, 52062 Aachen, Germany
| | - Frank G Grosveld
- Department of Cell Biology, Erasmus Medical Centre, 3015 CN Rotterdam, The Netherlands
| | - Argyris Papantonis
- Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
73
|
Hedl M, Yan J, Abraham C. IRF5 and IRF5 Disease-Risk Variants Increase Glycolysis and Human M1 Macrophage Polarization by Regulating Proximal Signaling and Akt2 Activation. Cell Rep 2016; 16:2442-55. [PMID: 27545875 DOI: 10.1016/j.celrep.2016.07.060] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 04/21/2016] [Accepted: 07/22/2016] [Indexed: 12/19/2022] Open
Abstract
Interferon regulatory factor 5 (IRF5) regulates inflammatory M1 macrophage polarization, and disease-associated IRF5 genetic variants regulate pattern-recognition-receptor (PRR)-induced cytokines. PRR-stimulated macrophages and M1 macrophages exhibit enhanced glycolysis, a central mediator of inflammation. We find that IRF5 is needed for PRR-enhanced glycolysis in human macrophages and in mice in vivo. Upon stimulation of the PRR nucleotide binding oligomerization domain containing 2 (NOD2) in human macrophages, IRF5 binds RIP2, IRAK1, and TRAF6. IRF5, in turn, is required for optimal Akt2 activation, which increases expression of glycolytic pathway genes and HIF1A as well as pro-inflammatory cytokines and M1 polarization. Furthermore, pro-inflammatory cytokines and glycolytic pathways co-regulate each other. Rs2004640/rs2280714 TT/TT IRF5 disease-risk-carrier cells demonstrate increased IRF5 expression and increased PRR-induced Akt2 activation, glycolysis, pro-inflammatory cytokines, and M1 polarization relative to GG/CC carrier macrophages. Our findings identify that IRF5 disease-associated polymorphisms regulate diverse immunological and metabolic outcomes and provide further insight into mechanisms contributing to the increasingly recognized important role for glycolysis in inflammation.
Collapse
Affiliation(s)
- Matija Hedl
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Jie Yan
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Clara Abraham
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
74
|
Lyn Kinase Suppresses the Transcriptional Activity of IRF5 in the TLR-MyD88 Pathway to Restrain the Development of Autoimmunity. Immunity 2016; 45:319-32. [PMID: 27521268 DOI: 10.1016/j.immuni.2016.07.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 05/05/2016] [Accepted: 05/24/2016] [Indexed: 01/16/2023]
Abstract
Interferon regulatory factor-5 (IRF5), a transcription factor critical for the induction of innate immune responses, contributes to the pathogenesis of the autoimmune disease systemic lupus erythematosus (SLE) in humans and mice. Lyn, a Src family kinase, is also implicated in human SLE, and Lyn-deficient mice develop an SLE-like disease. Here, we found that Lyn physically interacted with IRF5 to inhibit ubiquitination and phosphorylation of IRF5 in the TLR-MyD88 pathway, thereby suppressing the transcriptional activity of IRF5 in a manner independent of Lyn's kinase activity. Conversely, Lyn did not inhibit NF-κB signaling, another major branch downstream of MyD88. Monoallelic deletion of Irf5 alleviated the hyperproduction of cytokines in TLR-stimulated Lyn(-/-) dendritic cells and the development of SLE-like symptoms in Lyn(-/-) mice. Our results reveal a role for Lyn as a specific suppressor of the TLR-MyD88-IRF5 pathway and illustrate the importance of fine-tuning IRF5 activity for the maintenance of immune homeostasis.
Collapse
|
75
|
|
76
|
Kutilek VD, Andrews CL, Richards MP, Xu Z, Sun T, Chen Y, Hashke A, Smotrov N, Fernandez R, Nickbarg EB, Chamberlin C, Sauvagnat B, Curran PJ, Boinay R, Saradjian P, Allen SJ, Byrne N, Elsen NL, Ford RE, Hall DL, Kornienko M, Rickert KW, Sharma S, Shipman JM, Lumb KJ, Coleman K, Dandliker PJ, Kariv I, Beutel B. Integration of Affinity Selection-Mass Spectrometry and Functional Cell-Based Assays to Rapidly Triage Druggable Target Space within the NF-κB Pathway. ACTA ACUST UNITED AC 2016; 21:608-19. [PMID: 26969322 DOI: 10.1177/1087057116637353] [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: 11/03/2015] [Accepted: 02/15/2016] [Indexed: 11/15/2022]
Abstract
The primary objective of early drug discovery is to associate druggable target space with a desired phenotype. The inability to efficiently associate these often leads to failure early in the drug discovery process. In this proof-of-concept study, the most tractable starting points for drug discovery within the NF-κB pathway model system were identified by integrating affinity selection-mass spectrometry (AS-MS) with functional cellular assays. The AS-MS platform Automated Ligand Identification System (ALIS) was used to rapidly screen 15 NF-κB proteins in parallel against large-compound libraries. ALIS identified 382 target-selective compounds binding to 14 of the 15 proteins. Without any chemical optimization, 22 of the 382 target-selective compounds exhibited a cellular phenotype consistent with the respective target associated in ALIS. Further studies on structurally related compounds distinguished two chemical series that exhibited a preliminary structure-activity relationship and confirmed target-driven cellular activity to NF-κB1/p105 and TRAF5, respectively. These two series represent new drug discovery opportunities for chemical optimization. The results described herein demonstrate the power of combining ALIS with cell functional assays in a high-throughput, target-based approach to determine the most tractable drug discovery opportunities within a pathway.
Collapse
Affiliation(s)
- Victoria D Kutilek
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Christine L Andrews
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Matthew P Richards
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Zangwei Xu
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Tianxiao Sun
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Yiping Chen
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Andrew Hashke
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Nadya Smotrov
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Rafael Fernandez
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Elliott B Nickbarg
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Chad Chamberlin
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Berengere Sauvagnat
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Patrick J Curran
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Ryan Boinay
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Peter Saradjian
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Samantha J Allen
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Noel Byrne
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Nathaniel L Elsen
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA Current address: AbbVie, North Chicago, IL USA
| | - Rachael E Ford
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Dawn L Hall
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Maria Kornienko
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Keith W Rickert
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA Current address: Medimmune, Gaithersburg, MD, USA
| | - Sujata Sharma
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Jennifer M Shipman
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Kevin J Lumb
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Kevin Coleman
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA Current address: Arvinas, New Haven, CT, USA
| | - Peter J Dandliker
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Ilona Kariv
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| | - Bruce Beutel
- Department of Pharmacology, Screening and Protein Sciences, Merck & Co, Kenilworth, NJ, USA
| |
Collapse
|
77
|
Abstract
Interferon regulatory factor 5 (IRF5) has been demonstrated as a key transcription factor of the immune system, playing important roles in modulating inflammatory immune responses in numerous cell types including dendritic cells, macrophages, and B cells. As well as driving the expression of type I interferon in antiviral responses, IRF5 is also crucial for driving macrophages toward a proinflammatory phenotype by regulating cytokine and chemokine expression and modulating B-cell maturity and antibody production. This review highlights the functional importance of IRF5 in a disease setting, by discussing polymorphic mutations at the human Irf5 locus that lead to susceptibility to systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. In concordance with this, we also discuss lessons in IRF5 functionality learned from murine in vivo models of autoimmune disease and inflammation and hypothesize that modulation of IRF5 activity and expression could provide potential therapeutic benefits in the clinic.
Collapse
Affiliation(s)
- Hayley L Eames
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| | - Alastair L Corbin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
78
|
Abstract
Whereas the importance of macrophages in chronic inflammatory diseases is well recognized, there is an increasing awareness that neutrophils may also play an important role. In addition to the well-documented heterogeneity of macrophage phenotypes and functions, neutrophils also show remarkable phenotypic diversity among tissues. Understanding the molecular pathways that control this heterogeneity should provide abundant scope for the generation of more specific and effective therapeutics. We have shown that the transcription factor IFN regulatory factor 5 (IRF5) polarizes macrophages toward an inflammatory phenotype. IRF5 is also expressed in other myeloid cells, including neutrophils, where it was linked to neutrophil function. In this study we explored the role of IRF5 in models of acute inflammation, including antigen-induced inflammatory arthritis and lung injury, both involving an extensive influx of neutrophils. Mice lacking IRF5 accumulate far fewer neutrophils at the site of inflammation due to the reduced levels of chemokines important for neutrophil recruitment, such as the chemokine (C-X-C motif) ligand 1. Furthermore we found that neutrophils express little IRF5 in the joints and that their migratory properties are not affected by the IRF5 deficiency. These studies extend prior ones suggesting that inhibiting IRF5 might be useful for chronic macrophage-induced inflammation and suggest that IRF5 blockade would ameliorate more acute forms of inflammation, including lung injury.
Collapse
|
79
|
Ostuni R, Kratochvill F, Murray PJ, Natoli G. Macrophages and cancer: from mechanisms to therapeutic implications. Trends Immunol 2015; 36:229-39. [PMID: 25770924 DOI: 10.1016/j.it.2015.02.004] [Citation(s) in RCA: 514] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 01/06/2023]
Abstract
Infiltration by immune cells is a hallmark of most forms of malignancy. In this context, tumor-associated macrophages (TAMs) represent key regulators of the complex interplay between the immune system and cancer. We discuss evidence indicating that in many settings TAMs fuel, rather than limit, tumor progression, and negatively impact on responses to therapy. We discuss how the unique functional properties of TAMs are shaped by tumor-derived signals, placing TAM development in the context of the broader understanding of the cellular and molecular mechanisms controlling macrophage origin, differentiation, and maintenance in tissues. Finally, we provide examples of how a molecular understanding of the relationships between TAMs and the tumor microenvironment may lead to improved cancer therapies.
Collapse
Affiliation(s)
- Renato Ostuni
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan, Italy.
| | - Franz Kratochvill
- Department of Infectious Diseases and Immunology, St. Jude Children's Research Hospital Memphis, TN 38105, USA
| | - Peter J Murray
- Department of Infectious Diseases and Immunology, St. Jude Children's Research Hospital Memphis, TN 38105, USA
| | - Gioacchino Natoli
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan, Italy.
| |
Collapse
|
80
|
Jin J, Xiao Y, Hu H, Zou Q, Li Y, Gao Y, Ge W, Cheng X, Sun SC. Proinflammatory TLR signalling is regulated by a TRAF2-dependent proteolysis mechanism in macrophages. Nat Commun 2015; 6:5930. [PMID: 25565375 PMCID: PMC4286812 DOI: 10.1038/ncomms6930] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/21/2014] [Indexed: 12/31/2022] Open
Abstract
Signal transduction from toll-like receptors (TLRs) is important for innate immunity against infections, but deregulated TLR signalling contributes to inflammatory disorders. Here we show that myeloid cell-specific ablation of TRAF2 greatly promotes TLR-stimulated proinflammatory cytokine expression in macrophages and exacerbates colitis in an animal model of inflammatory bowel disease. TRAF2 deficiency does not enhance upstream signalling events, but it causes accumulation of two transcription factors, c-Rel and IRF5, known to mediate proinflammatory cytokine induction. Interestingly, TRAF2 controls the fate of c-Rel and IRF5 via a proteasome-dependent mechanism that also requires TRAF3 and the E3 ubiquitin ligase cIAP. We further show that TRAF2 also regulates inflammatory cytokine production in tumour-associated macrophages and facilitates tumour growth. These findings demonstrate an unexpected anti-inflammatory function of TRAF2 and suggest a proteasome-dependent mechanism that limits the proinflammatory TLR signalling.
Collapse
Affiliation(s)
- Jin Jin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Yichuan Xiao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Hongbo Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Qiang Zou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Yanchuan Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Yanpan Gao
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Dongdan Santiao 5#, Beijing 100005, China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Dongdan Santiao 5#, Beijing 100005, China
| | - Xuhong Cheng
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| | - Shao-Cong Sun
- 1] Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA [2] Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA [3] The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas 77030, USA
| |
Collapse
|
81
|
|