1
|
Wiggins DA, Maxwell JN, Nelson DE. Exploring the role of CITED transcriptional regulators in the control of macrophage polarization. Front Immunol 2024; 15:1365718. [PMID: 38646545 PMCID: PMC11032013 DOI: 10.3389/fimmu.2024.1365718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Macrophages are tissue resident innate phagocytic cells that take on contrasting phenotypes, or polarization states, in response to the changing combination of microbial and cytokine signals at sites of infection. During the opening stages of an infection, macrophages adopt the proinflammatory, highly antimicrobial M1 state, later shifting to an anti-inflammatory, pro-tissue repair M2 state as the infection resolves. The changes in gene expression underlying these transitions are primarily governed by nuclear factor kappaB (NF-κB), Janus kinase (JAK)/signal transducer and activation of transcription (STAT), and hypoxia-inducible factor 1 (HIF1) transcription factors, the activity of which must be carefully controlled to ensure an effective yet spatially and temporally restricted inflammatory response. While much of this control is provided by pathway-specific feedback loops, recent work has shown that the transcriptional co-regulators of the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxy-terminal domain (CITED) family serve as common controllers for these pathways. In this review, we describe how CITED proteins regulate polarization-associated gene expression changes by controlling the ability of transcription factors to form chromatin complexes with the histone acetyltransferase, CBP/p300. We will also cover how differences in the interactions between CITED1 and 2 with CBP/p300 drive their contrasting effects on pro-inflammatory gene expression.
Collapse
Affiliation(s)
| | | | - David E. Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
2
|
Kunte N, Westerfield M, McGraw E, Choi J, Akinsipe T, Whitaker SK, Brannen A, Panizzi P, Tomich JM, Avila LA. Evaluation of transfection efficacy, biodistribution, and toxicity of branched amphiphilic peptide capsules (BAPCs) associated with mRNA. Biomater Sci 2022; 10:6980-6991. [PMID: 36254388 DOI: 10.1039/d2bm01314b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanoparticles (NPs) have been shown to be a suitable mRNA delivery platform by conferring protection against ribonucleases and facilitating cellular uptake. Several NPs have succeeded in delivering mRNA intranasally, intratracheally, and intramuscularly in preclinical settings. However, intravenous mRNA delivery has been less explored. Only a few NPs have been tested for systemic delivery of mRNA, many of which are formulated with polyethylene glycol (PEG). The incorporation of PEG presents some tradeoffs that must be carefully considered when designing a systemic delivery model. For example, while the addition of PEG may prolong circulation time by preventing early clearance by the mononuclear phagocytic system (MPS), it has also been reported that treating patients with PEGylated drugs can result in hypersensitivity reactions due to anti-PEG antibodies. Thus, it is desirable to have alternative PEG-free delivery methods for mRNA to avoid these adverse effects while preserving the beneficial effects. Our research group developed BAPCs (branched amphiphilic peptide capsules), a peptide-based nanoparticle that resists disruption by chaotropes, proteases, and elevated temperature, thus displaying significant stability and shelf-life. In this study, we demonstrated that similarly to PEG, mRNA shields the BAPC cationic surface to avoid early clearance by the MPS. Multispectral optoacoustic tomography (MSOT) and fluorescence reflectance imaging were imaging techniques used to analyze biodistribution within major MPS organs. Analysis of pro-inflammatory cytokine expression showed that BAPC-mRNA complexes do not cause chronic inflammation. Additionally, BAPCs enhance intracellular delivery of mRNA with negligible cytotoxicity or oxidative stress. These results might pave the way for future therapeutic applications of BAPCs as a delivery platform for systemic mRNA delivery.
Collapse
Affiliation(s)
- Nitish Kunte
- Department of Biological Sciences, Auburn University, Auburn, AL- 36849, USA.
| | - Matthew Westerfield
- Department of Biological Sciences, Auburn University, Auburn, AL- 36849, USA.
| | - Erin McGraw
- Department of Biological Sciences, Auburn University, Auburn, AL- 36849, USA.
| | - Jiyeong Choi
- School of Integrative Plant Science, Cornell University, NY- 14853, USA
| | - Tosin Akinsipe
- Department of Biological Sciences, Auburn University, Auburn, AL- 36849, USA.
| | - Susan K Whitaker
- Department of Biochemistry and Molecular Biophysics, 141 Chalmers Hall, Kansas State University, Manhattan, Kansas- 66506, USA
| | | | - Peter Panizzi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - John M Tomich
- School of Integrative Plant Science, Cornell University, NY- 14853, USA
| | - L Adriana Avila
- Department of Biological Sciences, Auburn University, Auburn, AL- 36849, USA.
| |
Collapse
|
3
|
Interferon-gamma modulates articular chondrocyte and osteoblast metabolism through protein kinase R-independent and dependent mechanisms. Biochem Biophys Rep 2022; 32:101323. [PMID: 36105611 PMCID: PMC9464860 DOI: 10.1016/j.bbrep.2022.101323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Osteoarthritis (OA) affects multiple tissues of the synovial joint and is characterised by articular cartilage degeneration and bone remodelling. Interferon-γ (IFN-γ) is implicated in osteoarthritis pathology exerting its biological effects via various mechanisms including activation of protein kinase R (PKR), which has been implicated in inflammation and arthritis. This study investigated whether treatment of articular cartilage chondrocytes and osteoblasts with IFN-γ could induce a degradative phenotype that was mediated through the PKR signalling pathway. IFN-γ treatment of chondrocytes increased transcription of key inflammatory mediators (TNF-α, IL-6), matrix degrading enzymes (MMP-13), the transcription factor STAT1, and PKR. Activation of PKR was involved in the regulation of TNF-α, IL-6, and STAT1. In osteoblasts, IFN-γ increased human and mouse STAT1, and human IL-6 through a mechanism involving PKR. ALP, COL1A1 (human and mouse), RUNX2 (mouse), and PHOSPHO1 (mouse) were decreased by IFN-γ. The number of PKR positive cells were increased in post-traumatic OA (PTOA). This study has revealed that IFN-γ propagates inflammatory and degenerative events in articular chondrocytes and osteoblasts via PKR activation. Since IFN-γ and PKR signalling are both activated in early PTOA, these mechanisms are likely to contribute to joint degeneration after injury and might offer attractive targets for therapeutic intervention. •IFN-γ treatment of chondrocytes increased transcription of TNF-α, IL-6, and STAT1 via PKR activation. •In osteoblasts, IFN-γ increased STAT1 and IL-6 via PKR activation. •The number of PKR positive cells were increased in post-traumatic OA (PTOA). •IFN-γ propagates inflammatory and degenerative events in articular chondrocytes and osteoblasts via PKR activation. •IFN-γ and PKR signalling are both activated in early PTOA and are likely to contribute to joint degeneration after injury.
Collapse
|
4
|
Majerczyk D, Ayad E, Brewton K, Saing P, Hart P. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ. Biosci Rep 2022; 42:BSR20220713. [PMID: 36300375 PMCID: PMC9670245 DOI: 10.1042/bsr20220713] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother's immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
Collapse
Affiliation(s)
- Daniel Majerczyk
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
- Loyola Medicine, Berwyn, Illinois 60402, U.S.A
| | - Elizabeth G. Ayad
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Kari L. Brewton
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Pichrasmei Saing
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| | - Peter C. Hart
- College of Science, Health and Pharmacy, Roosevelt University, Illinois 60173, U.S.A
| |
Collapse
|
5
|
Sumayin Ngamdu K, Mallawaarachchi I, Dunipace EA, Chuang LH, Jafri SH, Shah NR, Jeong YN, Morrison AR, Bhatt DL. Association Between Periodontal Disease and Cardiovascular Disease (from the NHANES). Am J Cardiol 2022; 178:163-168. [PMID: 35773046 DOI: 10.1016/j.amjcard.2022.05.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/01/2022]
Abstract
As observational studies support the association between periodontal disease (PD) and cardiovascular diseases (CVDs), we examined this relationship using the National Health and Nutrition Examination Survey 2013 to 2014 data. This cross-sectional study involved 2,830 adult participants, aged ≥30 years who underwent a home interview, followed by a standardized assessment at a mobile examination center from 2013 to 2014. PD was defined using the new classification scheme issued by American Academy of Periodontology Workshop on the Classification of Periodontal and Peri-implant Diseases and Conditions in 2017, and CVD was defined as the composite of coronary artery disease and stroke. The gathered data were subjected to weighted statistical analysis to examine the relation between CVD prevalence and PD. The sample (mean age 51.5 ± 13.6 years) comprised 50.1% men and 69.5% White participants. Stage I (mild/subclinical), II (moderate), and III to IV (severe) PD was noted in 16.7% (95% confidence interval [CI] 12.7 to 21.7), 57.4% (95% CI 53.9 to 60.9), and 25.9% (95% CI 21.4 to 30.8) of the participants, respectively. Patients with stage III and IV PD were more likely to have CVD than those with stage I (adjusted odds ratio 3.59, 95% CI 1.12 to 11.54, p = 0.03). Similarly, participants who reported fair/poor gum health were more likely to have CVD than those who reported excellent/very good gum health (adjusted odds ratio 2.17, 95% CI 0.98 to 4.79, p = 0.055). In conclusion, the data from the National Health and Nutrition Examination Survey 2013 to 2014 demonstrated that PD severity is associated with CVD risk. However, the information gathered by trained professionals during periodontal examinations is a more reliable predictor of PD-CVD associations compared with self-reported measures of oral health.
Collapse
Affiliation(s)
- Kyari Sumayin Ngamdu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Providence VA Medical Center, Providence, Rhode Island; Division of Cardiovascular Medicine, Department of Internal Medicine, Alpert Medical School at Brown University, Providence, Rhode Island
| | - Indika Mallawaarachchi
- Department of Public Health Sciences, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Eric A Dunipace
- David Geffen School of Medicine, University of California, Los Angeles, California
| | - Lin-Hsin Chuang
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts
| | - S Hammad Jafri
- Division of Cardiovascular Medicine, Department of Internal Medicine, Providence VA Medical Center, Providence, Rhode Island; Division of Cardiovascular Medicine, Department of Internal Medicine, Alpert Medical School at Brown University, Providence, Rhode Island
| | - Nishant R Shah
- Division of Cardiovascular Medicine, Department of Internal Medicine, Providence VA Medical Center, Providence, Rhode Island; Division of Cardiovascular Medicine, Department of Internal Medicine, Alpert Medical School at Brown University, Providence, Rhode Island
| | - Youjin N Jeong
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts
| | - Alan R Morrison
- Division of Cardiovascular Medicine, Department of Internal Medicine, Providence VA Medical Center, Providence, Rhode Island; Division of Cardiovascular Medicine, Department of Internal Medicine, Alpert Medical School at Brown University, Providence, Rhode Island
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
6
|
Chang WT, Hong MY, Chen CL, Hwang CY, Tsai CC, Chuang CC. Mutant glucocorticoid receptor binding elements on the interleukin-6 promoter regulate dexamethasone effects. BMC Immunol 2021; 22:24. [PMID: 33771121 PMCID: PMC7995394 DOI: 10.1186/s12865-021-00413-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/14/2021] [Indexed: 12/15/2022] Open
Abstract
Background Glucocorticoids (GCs) have been extensively used as essential modulators in clinical infectious and inflammatory diseases. The GC receptor (GR) is a transcription factor belonging to the nuclear receptor family that regulates anti-inflammatory processes and releases pro-inflammatory cytokines, such as interleukin (IL)-6. Results Five putative GR binding sites and other transcriptional factor binding sites were identified on theIL-6 promoter, and dexamethasone (DEX) was noted to reduce the lipopolysaccharide (LPS)-induced IL-6 production. Among mutant transcriptional factor binding sites, nuclear factor-kappa B (NF-κB), activator protein (AP)-1, and specificity protein (Sp)1–2 sites reduced basal and LPS-induced IL-6 promoter activities through various responses. The second GR binding site (GR2) was noted to play a crucial role in both basal and inducible promoter activities in LPS-induced inflammation. Conclusions We concluded that selective GR2 modulator might exert agonistic and antagonistic effects and could activate crucial signaling pathways during the LPS-stimulated inflammatory process. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00413-z.
Collapse
Affiliation(s)
- Wen-Teng Chang
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, 701, Taiwan
| | - Ming-Yuan Hong
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Liang Chen
- Department of Physical Therapy, I-Shou University, Kaohsiung, Taiwan
| | - Chi-Yuan Hwang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Chieh Tsai
- Department of Nursing, Chung Hwa University of Medical Technology, Tainan, 701, Taiwan
| | - Chia-Chang Chuang
- Department of Emergency Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
7
|
Leão FB, Vaughn LS, Bhatt D, Liao W, Maloney D, Carvalho BC, Oliveira L, Ghosh S, Silva AM. Toll-like Receptor (TLR)-induced Rasgef1b expression in macrophages is regulated by NF-κB through its proximal promoter. Int J Biochem Cell Biol 2020; 127:105840. [PMID: 32866686 DOI: 10.1016/j.biocel.2020.105840] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/31/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022]
Abstract
Ras Guanine Exchange Factor (RasGEF) domain family member 1b is encoded by a Toll-like receptor (TLR)-inducible gene expressed in macrophages, but transcriptional mechanisms that govern its expression are still unknown. Here, we have functionally characterized the 5' flanking Rasgef1b sequence and analyzed its transcriptional activation. We have identified that the inflammation-responsive promoter is contained within a short sequence (-183 to +119) surrounding the transcriptional start site. The promoter sequence is evolutionarily conserved and harbors a cluster of five NF-κB binding sites. Luciferase reporter gene assay showed that the promoter is responsive to TLR activation and RelA or cRel, but not RelB, transcription factors. Besides, site-directed mutagenesis showed that the κB binding sites are required for maximal promoter activation induced by LPS. Analysis by Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) revealed that the promoter is located in an accessible chromatin region. More important, Chromatin Immunoprecipitation sequencing (ChIP-seq) showed that RelA is recruited to the promoter region upon LPS stimulation of bone marrow-derived macrophages. Finally, studies with Rela-deficient macrophages or pharmacological inhibition by Bay11-7082 showed that NF-κB is required for optimal Rasgef1b expression induced by TLR agonists. Our data provide evidence of the regulatory mechanism mediated by NF-κB that facilitates Rasgef1b expression after TLR activation in macrophages.
Collapse
Affiliation(s)
- Felipe B Leão
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lauren S Vaughn
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Dev Bhatt
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Will Liao
- New York Genome Center, New York, NY 10013, USA
| | | | - Brener C Carvalho
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Leonardo Oliveira
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Columbia University, College of Physicians and Surgeons, New York, NY 10031, USA
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
8
|
Rius-Pérez S, Pérez S, Martí-Andrés P, Monsalve M, Sastre J. Nuclear Factor Kappa B Signaling Complexes in Acute Inflammation. Antioxid Redox Signal 2020; 33:145-165. [PMID: 31856585 DOI: 10.1089/ars.2019.7975] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Nuclear factor kappa B (NF-κB) is a master regulator of the inflammatory response and represents a key regulatory node in the complex inflammatory signaling network. In addition, selective NF-κB transcriptional activity on specific target genes occurs through the control of redox-sensitive NF-κB interactions. Recent Advances: The selective NF-κB response is mediated by redox-modulated NF-κB complexes with ribosomal protein S3 (RPS3), Pirin (PIR). cAMP response element-binding (CREB)-binding protein (CBP)/p300, peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), activator protein-1 (AP-1), signal transducer and activator of transcription 3 (STAT3), early growth response protein 1 (EGR-1), and SP-1. NF-κB is cooperatively coactivated with AP-1, STAT3, EGR-1, and SP-1 during the inflammatory process, whereas NF-κB complexes with CBP/p300 and PGC-1α regulate the expression of antioxidant genes. PGC-1α may act as selective repressor of phospho-p65 toward interleukin-6 (IL-6) in acute inflammation. p65 and nuclear factor erythroid 2-related factor 2 (NRF2) compete for binding to coactivator CBP/p300 playing opposite roles in the regulation of inflammatory genes. S-nitrosylation or tyrosine nitration favors the recruitment of specific NF-κB subunits to κB sites. Critical Issues: NF-κB is a redox-sensitive transcription factor that forms specific signaling complexes to regulate selectively the expression of target genes in acute inflammation. Protein-protein interactions with coregulatory proteins, other transcription factors, and chromatin-remodeling proteins provide transcriptional specificity to NF-κB. Furthermore, different NF-κB subunits may form distinct redox-sensitive homo- and heterodimers with distinct affinities for κB sites. Future Directions: Further research is required to elucidate the whole NF-κB interactome to fully characterize the complex NF-κB signaling network in redox signaling, inflammation, and cancer.
Collapse
Affiliation(s)
- Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Pablo Martí-Andrés
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - María Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| |
Collapse
|
9
|
Zhang Y, Yu Y, Ou C, Ma J, Wang Q, Du S, Xu Z, Li R, Guo F. Alleviation of infectious-bursal-disease-virus-induced bursal injury by betaine is associated with DNA methylation in IL-6 and interferon regulatory factor 7 promoter. Poult Sci 2019; 98:4457-4464. [PMID: 31162616 DOI: 10.3382/ps/pez280] [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: 10/27/2018] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Infectious bursal disease virus (IBDV) often infects young chickens and causes severe immunosuppression and inflammatory injury. Betaine is an antiviral and anti-inflammatory ingredient that may exert functions through epigenetic regulation. However, the effects of betaine on an IBDV-induced bursal injury and their underlying mechanisms have not been investigated. In this study, betaine was supplemented to the drinking water of newly hatched commercial broilers for 3 wk. Afterward, the chickens were infected with the IBDV. After 5 D of infection, the bursal lesions were examined. The mRNA expression levels of IBDV VP2 gene, pro-inflammatory cytokines, and interferons were detected. Furthermore, the 5-methylcytosine level of the CpG island in the promoter region of IL-6 and interferon regulatory factor 7 (IRF7) were determined. The IBDV induced the depletion of lymphocytes and inflammation in the bursal follicles. IBDV infection considerably elevated the mRNA levels of VP2, IL-6, types I (IFNα and IFNβ) and II (IFNγ) interferons, and IRF7. The CpG island methylation in the promoter regions of IL-6 and IRF7 were substantially decreased after IBDV infection. Betaine administration attenuated the IBDV-induced bursal lesions. Meanwhile, the IBDV-induced mRNA expression levels of IL-6, IFNβ, and IRF7 were suppressed by betaine consumption. Furthermore, the hypomethylation effects of IBDV infection to the promoter regions of IL-6 and IRF7 genes were eliminated and relieved by betaine administration. Our results indicated that the IBDV-induced expression levels of IL-6 and IRF7 genes are associated with the suppression of methylation in the promoter region. Betaine administration through drinking water may alleviate the IBDV-induced bursal injury via epigenetic regulation.
Collapse
Affiliation(s)
- Yanhong Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Yan Yu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Changbo Ou
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Qiuxia Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Shouyang Du
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Zhiyong Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Renfeng Li
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| | - Feng Guo
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China.,Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang 453003, Henan, China
| |
Collapse
|
10
|
Hamel-Côté G, Lapointe F, Véronneau S, Mayhue M, Rola-Pleszczynski M, Stankova J. Regulation of platelet-activating factor-mediated interleukin-6 promoter activation by the 48 kDa but not the 45 kDa isoform of protein tyrosine phosphatase non-receptor type 2. Cell Biosci 2019; 9:51. [PMID: 31289638 PMCID: PMC6593612 DOI: 10.1186/s13578-019-0316-9] [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: 02/21/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background An underlying state of inflammation is thought to be an important cause of cardiovascular disease. Among cells involved in the early steps of atherosclerosis, monocyte-derived dendritic cells (Mo-DCs) respond to inflammatory stimuli, including platelet-activating factor (PAF), by the induction of various cytokines, such as interleukin 6 (IL-6). PAF is a potent phospholipid mediator involved in both the onset and progression of atherosclerosis. It mediates its effects by binding to its cognate G-protein coupled receptor, PAFR. Activation of PAFR-induced signaling pathways is tightly coordinated to ensure specific cell responses. Results Here, we report that PAF stimulated the phosphatase activity of both the 45 and 48 kDa isoforms of the protein tyrosine phosphatase non-receptor type 2 (PTPN2). However, we found that only the 48 kDa PTPN2 isoform has a role in PAFR-induced signal transduction, leading to activation of the IL-6 promoter. In luciferase reporter assays, expression of the 48 kDa, but not the 45 kDa, PTPN2 isoform increased human IL-6 (hIL-6) promoter activity by 40% after PAF stimulation of HEK-293 cells, stably transfected with PAFR (HEK-PAFR). Our results suggest that the differential localization of the PTPN2 isoforms and the differences in PAF-induced phosphatase activation may contribute to the divergent modulation of PAF-induced IL-6 promoter activation. The involvement of PTPN2 in PAF-induced IL-6 expression was confirmed in immature Mo-DCs (iMo-DCs), using siRNAs targeting the two isoforms of PTPN2, where siRNAs against the 48 kDa PTPN2 significantly inhibited PAF-stimulated IL-6 mRNA expression. Pharmacological inhibition of several signaling pathways suggested a role for PTPN2 in early signaling events. Results obtained by Western blot confirmed that PTPN2 increased the activation of the PI3K/Akt pathway via the modulation of protein kinase D (PKD) activity. WT PKD expression counteracted the effect of PTPN2 on PAF-induced IL-6 promoter transactivation and phosphorylation of Akt. Using siRNAs targeting the individual isoforms of PTPN2, we confirmed that these pathways were also active in iMo-DCs. Conclusion Taken together, our data suggest that PTPN2, in an isoform-specific manner, could be involved in the positive regulation of PI3K/Akt activation, via the modulation of PKD activity, allowing for the maximal induction of PAF-stimulated IL-6 mRNA expression.
Collapse
Affiliation(s)
- Geneviève Hamel-Côté
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Fanny Lapointe
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Steeve Véronneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Marian Mayhue
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Marek Rola-Pleszczynski
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Jana Stankova
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| |
Collapse
|
11
|
Chandler JD, Hu X, Ko EJ, Park S, Fernandes J, Lee YT, Orr ML, Hao L, Smith MR, Neujahr DC, Uppal K, Kang SM, Jones DP, Go YM. Low-dose cadmium potentiates lung inflammatory response to 2009 pandemic H1N1 influenza virus in mice. ENVIRONMENT INTERNATIONAL 2019; 127:720-729. [PMID: 30999129 PMCID: PMC6536378 DOI: 10.1016/j.envint.2019.03.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/08/2019] [Accepted: 03/24/2019] [Indexed: 05/04/2023]
Abstract
Cadmium (Cd) is a toxic, pro-inflammatory metal ubiquitous in the diet that accumulates in body organs due to inefficient elimination. Responses to influenza virus infection are variable, particularly severity of pneumonia. We used a murine model of chronic low-dose oral exposure to Cd to test if increased lung tissue Cd worsened inflammation in response to sub-lethal H1N1 infection. The results show that Cd-treated mice had increased lung tissue inflammatory cells, including neutrophils, monocytes, T lymphocytes and dendritic cells, following H1N1 infection. Lung genetic responses to infection (increasing TNF-α, interferon and complement, and decreasing myogenesis) were also exacerbated. To reveal the organization of a network structure, pinpointing molecules critical to Cd-altered lung function, global correlations were made for immune cell counts, leading edge gene transcripts and metabolites. This revealed that Cd increased correlation of myeloid immune cells with pro-inflammatory genes, particularly interferon-γ and metabolites. Together, the results show that Cd burden in mice increased inflammation in response to sub-lethal H1N1 challenge, which was coordinated by genetic and metabolic responses, and could provide new targets for intervention against lethal inflammatory pathology of clinical H1N1 infection.
Collapse
Affiliation(s)
- Joshua D Chandler
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Xin Hu
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Eun-Ju Ko
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303, United States of America
| | - Soojin Park
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303, United States of America
| | - Jolyn Fernandes
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Young-Tae Lee
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303, United States of America
| | - Michael L Orr
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Li Hao
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - M Ryan Smith
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - David C Neujahr
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Karan Uppal
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA 30303, United States of America
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America.
| | - Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322, United States of America.
| |
Collapse
|
12
|
Yang B, Ge Y, Zhou Y, Wang J, Xie X, Li S, Tang M, Xu L, Tian J. miR-124a inhibits the proliferation and inflammation in rheumatoid arthritis fibroblast-like synoviocytes via targeting PIK3/NF-κB pathway. Cell Biochem Funct 2019; 37:208-215. [PMID: 30941802 DOI: 10.1002/cbf.3386] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/04/2018] [Accepted: 01/27/2019] [Indexed: 01/06/2023]
Abstract
Abnormal hyperplasia of fibroblast-like synoviocytes (FLS) leads to the progression of rheumatoid arthritis (RA). This study aimed to investigate the role of miR-124a in the pathogenesis of RA. The viability and cell cycle of FLS in rheumatoid arthritis (RAFLS) were evaluated by Cell Counting Kit 8 and flow cytometry assay. The expression of PIK3CA, Akt, and NF-κB in RAFLS was examined by real-time PCR and Western blot analysis. The production of tumour necrosis factor (TNF)-α and interleukin (IL)-6 was detected by ELISA. The joint swelling and inflammation in collagen-induced arthritis (CIA) mice were examined by histological and immunohistochemical analysis. We found that miR-124a suppressed the viability and proliferation of RAFLS and increased the percentage of cells in the G1 phase. miR-124a suppressed PIK3CA 3'UTR luciferase reporter activity and decreased the expression of PIK3CA at mRNA and protein levels. Furthermore, miR-124a inhibited the expression of the key components of the PIK3/Akt/NF-κB signal pathway and inhibited the expression of pro-inflammatory factors TNF-α and IL-6. Local overexpression of miR-124a in the joints of CIA mice inhibited inflammation and promoted apoptosis in FLS by decreasing PIK3CA expression. In conclusion, miR-124a inhibits the proliferation and inflammation in RAFLS via targeting PIK3/NF-κB pathway. miR-124a is a promising therapeutic target for RA.
Collapse
Affiliation(s)
- BiLing Yang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Ge
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Zhou
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jia Wang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xi Xie
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shu Li
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mengshi Tang
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - LiChang Xu
- International Department of YALI High School, Changsha, Hunan, China
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Carlin AF, Plummer EM, Vizcarra EA, Sheets N, Joo Y, Tang W, Day J, Greenbaum J, Glass CK, Diamond MS, Shresta S. An IRF-3-, IRF-5-, and IRF-7-Independent Pathway of Dengue Viral Resistance Utilizes IRF-1 to Stimulate Type I and II Interferon Responses. Cell Rep 2018; 21:1600-1612. [PMID: 29117564 DOI: 10.1016/j.celrep.2017.10.054] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/25/2017] [Accepted: 10/13/2017] [Indexed: 01/19/2023] Open
Abstract
Interferon-regulatory factors (IRFs) are a family of transcription factors (TFs) that translate viral recognition into antiviral responses, including type I interferon (IFN) production. Dengue virus (DENV) and other clinically important flaviviruses are suppressed by type I IFN. While mice lacking the type I IFN receptor (Ifnar1-/-) succumb to DENV infection, we found that mice deficient in three transcription factors controlling type I IFN production (Irf3-/-Irf5-/-Irf7-/- triple knockout [TKO]) survive DENV challenge. DENV infection of TKO mice resulted in minimal type I IFN production but a robust type II IFN (IFN-γ) response. Using loss-of-function approaches for various molecules, we demonstrate that the IRF-3-, IRF-5-, IRF-7-independent pathway predominantly utilizes IFN-γ and, to a lesser degree, type I IFNs. This pathway signals via IRF-1 to stimulate interleukin-12 (IL-12) production and IFN-γ response. These results reveal a key antiviral role for IRF-1 by activating both type I and II IFN responses during DENV infection.
Collapse
Affiliation(s)
- Aaron F Carlin
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Emily M Plummer
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Edward A Vizcarra
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Nicholas Sheets
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Yunichel Joo
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - William Tang
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Jeremy Day
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Jay Greenbaum
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Michael S Diamond
- Departments of Medicine, Pathology and Immunology, and Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sujan Shresta
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA.
| |
Collapse
|
14
|
Péant B, Gilbert S, Le Page C, Poisson A, L'Ecuyer E, Boudhraa Z, Bienz MN, Delvoye N, Saad F, Mes-Masson AM. IκB-Kinase-epsilon (IKKε) over-expression promotes the growth of prostate cancer through the C/EBP-β dependent activation of IL-6 gene expression. Oncotarget 2017; 8:14487-14501. [PMID: 27577074 PMCID: PMC5362420 DOI: 10.18632/oncotarget.11629] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 08/20/2016] [Indexed: 01/13/2023] Open
Abstract
The inflammatory cytokine IL-6 has been shown to induce the nuclear translocation of androgen receptors in prostate cancer cells and to activate the androgen receptors in a ligand-independent manner, suggesting it may contribute to the development of a castrate-resistant phenotype. Elevated IL-6 serum levels have also been associated with metastasis-related morbidity in prostate cancer patients. We have previously established that over-expression of I-kappa-B-kinase-epsilon (IKKε also named IKKi or IκBKε) in hormone-sensitive prostate cancer cell lines induces IL-6 secretion. We have also reported that prostate cancer cell lines lacking androgen receptor expression exhibit high constitutive IKKε expression and IL-6 secretion. In the present study, we validated the impact of IKKε depletion on the in vitro proliferation of castrate-resistant prostate cancer cells, and characterized how IKKε depletion affects tumor growth and IL-6 tumor secretion in vivo through a mouse xenograft-based approach. We observed a significant growth delay in IKKε-silenced PC-3 cells injected in SCID mice fed with a doxycycline-supplemented diet in comparison with mice fed with a normal diet. We also found a decrease in IL-6 secretion levels that strongly correlated with tumor growth inhibition. Finally, using constructs with various IL-6-mutated promoters, we demonstrated that IKKε over-expression induces a NF-κB-independent stimulation of the IL-6 gene promoter through the activation and nuclear accumulation of the transcription factor C/EBP-β. Our study demonstrates the pro-proliferative role of the oncogene IKKε in castrate-resistant prostate cancer cell lines, involving the phosphorylation and nuclear translocation of C/EBP-β that initiates IL-6 gene expression.
Collapse
Affiliation(s)
- Benjamin Péant
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Sophie Gilbert
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Cécile Le Page
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Alexis Poisson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Emilie L'Ecuyer
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Zied Boudhraa
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Marc Nicolas Bienz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Nathalie Delvoye
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada
| | - Fred Saad
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada.,Department of Surgery, Hôpital Saint Luc (CHUM), Montreal, Canada.,Department of Surgery, Université de Montréal, Montreal, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Montreal, Canada.,Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
15
|
Chistiakov DA, Myasoedova VA, Revin VV, Orekhov AN, Bobryshev YV. The impact of interferon-regulatory factors to macrophage differentiation and polarization into M1 and M2. Immunobiology 2017; 223:101-111. [PMID: 29032836 DOI: 10.1016/j.imbio.2017.10.005] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/03/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
The mononuclear phagocytes control the body homeostasis through the involvement in resolving tissue injury and further wound healing. Indeed, local tissue microenvironmental changes can significantly influence the functional behavior of monocytes and macrophages. Such microenvironmental changes for example occur in an atherosclerotic plaque during all progression stages. In response to exogenous stimuli, macrophages show a great phenotypic plasticity and heterogeneity. Exposure of monocytes to inflammatory or anti-inflammatory conditions also induces predominant differentiation to proinflammatory (M1) or anti-inflammatory (M2) macrophage subsets and phenotype switch between macrophage subsets. The phenotype transition is accompanied with great changes in the macrophage transcriptome and regulatory networks. Interferon-regulatory factors (IRFs) play a key role in hematopoietic development of monocytes, their differentiation to macrophages, and regulating macrophage maturation, phenotypic polarization, phenotypic switch, and function. Of 9 IRFs, at least 3 (IRF-1, IRF-5, and IRF-8) are involved in the commitment of proinflammatory M1 whereas IRF-3 and IRF-4 control M2 polarization. The role of IRF-2 is context-dependent. The IRF impact on macrophage phenotype plasticity and heterogeneity is complex and involves activating and repressive function in triggering transcription of target genes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center of Psychiatry and Narcology, Moscow, Russia; Department of Molecular Genetic Diagnostics and Cell Biology, Institute of Pediatrics, Research Center for Children's Health, Moscow, Russia
| | - Veronika A Myasoedova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Victor V Revin
- Biological Faculty, N.P. Ogaryov Mordovian State University, Republic of Mordovia, Saransk 430005, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, NSW, Sydney, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.
| |
Collapse
|
16
|
Murphy W, Muroi M, Zhang C, Suzuki T, Russell S. Both basal and enhancer κB elements are required for full induction of the mouse inducible nitric oxide synthase gene. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300502] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transcriptional regulatory region of the mouse inducible nitric oxide synthase (iNOS) gene has two KB elements, one enhancer-linked (KBII) and the other (KBI) proximal to its core promoter. Mutation of κBII substantially reduced the extent to which the iNOS promoter could be induced by LPS and interfered with augmented responsiveness of the promoter to LPS+IFN-γ. Mutation of KBI had a quantitatively less dramatic negative effect on LPS responsiveness and this construct still showed augmented responsiveness to LPS+IFN-γ. When both KB elements were mutated, inducibility by LPS and, in particular, by LPS+IFN-γ was paradoxically restored, compared with the mutated KBII alone, suggesting cooperative interactions among the transcription factors that trans-activate the iNOS gene. In vivo footprint analysis showed that both KB elements were bound by protein complexes when macrophages were stimulated with LPS ± IFN-γ. Furthermore, KBI was bound even in untreated cells, suggesting that KB binding proteins might also have a negative influence on expression of the gene. Both KBI and KBII were bound by NF-KB/Rel proteins found in nuclear extracts prepared from macrophages treated with LPS ± IFN-γ, although the specificity of binding to each element was different. Our results show that, while NF-KB/Rel proteins are required for maximal expression of the iNOS gene, alone they are not alone sufficient. Furthermore, the results reported here show that the augmentative effect of IFN-γ on the LPS-induced expression of the iNOS gene is not mediated through increased activation of NF-KB/Rel.
Collapse
Affiliation(s)
- W.J. Murphy
- The Wilkinson Laboratory of the Kansas Cancer Institute, and the Departments of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA, Wilkinson Laboratory of the Kansas Cancer Institute, 1008 Wahl Hall West, 3901 Rainbow Boulevard, Kansas City, KS 66160-7184, USA,
| | - M. Muroi
- ABL-Basic Research Program, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland, USA
| | - C.X. Zhang
- The Wilkinson Laboratory of the Kansas Cancer Institute, and the Departments of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - T. Suzuki
- Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - S.W. Russell
- The Wilkinson Laboratory of the Kansas Cancer Institute, and the Departments of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
17
|
Do KH, Park SH, Kim J, Yu M, Moon Y. Ribosome Inactivation Leads to Attenuation of Intestinal Polymeric Ig Receptor Expression via Differential Regulation of Human Antigen R. THE JOURNAL OF IMMUNOLOGY 2016; 197:847-58. [DOI: 10.4049/jimmunol.1502047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 05/12/2016] [Indexed: 02/07/2023]
|
18
|
Fantetti KN, Gray EL, Ganesan P, Kulkarni A, O'Donnell LA. Interferon gamma protects neonatal neural stem/progenitor cells during measles virus infection of the brain. J Neuroinflammation 2016; 13:107. [PMID: 27178303 PMCID: PMC4867982 DOI: 10.1186/s12974-016-0571-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 05/06/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND In the developing brain, self-renewing neural stem/progenitor cells (NSPC) give rise to neuronal and glial lineages. NSPC survival and differentiation can be altered by neurotropic viruses and by the anti-viral immune response. Several neurotropic viruses specifically target and infect NSPCs, in addition to inducing neuronal loss, which makes it difficult to distinguish between effects on NSPCs that are due to direct viral infection or due to the anti-viral immune response. METHODS We have investigated the impact of anti-viral immunity on NSPCs in measles virus (MV)-infected neonates. A neuron-restricted viral infection model was used, where NSPCs remain uninfected. Thus, an anti-viral immune response was induced without the confounding issue of NSPC infection. Two-transgenic mouse lines were used: CD46+ mice express the human isoform of CD46, the MV entry receptor, under the control of the neuron-specific enolase promoter; CD46+/IFNγ-KO mice lack the key anti-viral cytokine IFNγ. Multi-color flow cytometry and Western Blot analysis were used to quantify effects on NSPC, neuronal, and glial cell number, and quantify effects on IFNγ-mediated signaling and cell markers, respectively. RESULTS Flow cytometric analysis revealed that NSPCs were reduced in CD46+/IFNγ-KO mice at 3, 7, and 10 days post-infection (dpi), but were unaffected in CD46+ mice. Early neurons showed the greatest cell loss at 7 dpi in both genotypes, with no effect on mature neurons and glial cells. Thus, IFNγ protected against NSPC loss, but did not protect young neurons. Western Blot analyses on hippocampal explants showed reduced nestin expression in the absence of IFNγ, and reduced doublecortin and βIII-tubulin in both genotypes. Phosphorylation of STAT1 and STAT2 occurred independently of IFNγ in the hippocampus, albeit with distinct regulation of activation. CONCLUSIONS This is the first study to demonstrate bystander effects of anti-viral immunity on NSPC function. Our results show IFNγ protects the NSPC population during a neonatal viral CNS infection. Significant loss of NSPCs in CD46+/IFNγ-KO neonates suggests that the adaptive immune response is detrimental to NSPCs in the absence of IFNγ. These results reveal the importance and contribution of the anti-viral immune response to neuropathology and may be relevant to other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Kristen N Fantetti
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Erica L Gray
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Priya Ganesan
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Apurva Kulkarni
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Lauren A O'Donnell
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
19
|
Quirós Y, Blanco-Gozalo V, Sanchez-Gallego JI, López-Hernandez FJ, Ruiz J, Perez de Obanos MP, López-Novoa JM. Cardiotrophin-1 therapy prevents gentamicin-induced nephrotoxicity in rats. Pharmacol Res 2016; 107:137-146. [PMID: 26996880 DOI: 10.1016/j.phrs.2016.02.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/10/2023]
Abstract
Aminoglycosides are very effective antibiotics for the treatment of severe infections, but they rank among the most frequent causes of drug-induced nephrotoxicity. Thus, prevention of aminoglycoside nephrotoxicity is an unmet therapeutic objective. Cardiotrophin-1 (CT-1), a member of the IL-6 family of cytokines, has been reported to protect the kidney against toxic and ischemic acute kidney injury (AKI). We have assessed the effect of rat CT-1 in the severity of gentamicin (G)-induced AKI. Groups of male Wistar rats received the following for 6 consecutive days: i) isotonic saline solution (group CONT), ii) G, 150mg/kg/day, i.p. (group G), iii) CT-1, 100μg/kg/day i.v. (group CT-1), or iv) G and CT-1 at the doses described above. The G group showed a manifest AKI characterized by low creatinine clearance, high plasma creatinine and urea levels, increased urinary excretion of proteins, glucose and AKI markers such as N-acetyl-glucosaminidase, neutrophil gelatinase-associated lipocalin, kidney-injury molecule-1 and T-gelsolin, increased kidney levels of CD-68, iNOS, IL-1β and TNF-α, and markedly higher histological renal damage and leukocyte infiltration than the CONT and CT-1 groups. Administration of CT-1 together with G reduced almost all of the above-described manifestations of G-induced AKI. The results of this study have potential clinical application, as CT-1 is near to being used as a drug for organ protection.
Collapse
Affiliation(s)
| | | | | | - Francisco J López-Hernandez
- Unidad de Fisiopatología Renal y Cardiovascular, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Instituto de Estudios de Ciencias de la Salud de Castilla y León (IESCYL), Salamanca, Spain
| | | | | | - José M López-Novoa
- Unidad de Fisiopatología Renal y Cardiovascular, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
20
|
Talaei N, Yu T, Manion K, Bremner R, Wither JE. Identification of the SLAM Adapter Molecule EAT-2 as a Lupus-Susceptibility Gene That Acts through Impaired Negative Regulation of Dendritic Cell Signaling. THE JOURNAL OF IMMUNOLOGY 2015; 195:4623-31. [PMID: 26432891 DOI: 10.4049/jimmunol.1500552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/23/2015] [Indexed: 01/06/2023]
Abstract
We showed previously that C57BL/6 congenic mice with an introgressed homozygous 70 cM (125.6 Mb) to 100 cM (179.8 Mb) interval on c1 from the lupus-prone New Zealand Black (NZB) mouse develop high titers of antinuclear Abs and severe glomerulonephritis. Using subcongenic mice, we found that a genetic locus in the 88-96 cM region was associated with altered dendritic cell (DC) function and synergized with T cell functional defects to promote expansion of pathogenic proinflammatory T cell subsets. In this article, we show that the promoter region of the NZB gene encoding the SLAM signaling pathway adapter molecule EWS-activated transcript 2 (EAT-2) is polymorphic, which results in an ∼ 70% reduction in EAT-2 in DC. Silencing of the EAT-2 gene in DC that lacked this polymorphism led to increased production of IL-12 and enhanced differentiation of T cells to a Th1 phenotype in T cell-DC cocultures, reproducing the phenotype observed for DC from congenic mice with the NZB c1 70-100 cM interval. SLAM signaling was shown to inhibit production of IL-12 by CD40L-activated DCs. Consistent with a role for EAT-2 in this inhibition, knockdown of EAT-2 resulted in increased production of IL-12 by CD40-stimulated DC. Assessment of downstream signaling following CD40 cross-linking in the presence or absence of SLAM cross-linking revealed that SLAM coengagement blocked activation of p38 MAPK and JNK signaling pathways in DC, which was reversed in DC with the NZB EAT-2 allele. We conclude that EAT-2 negatively regulates cytokine production in DC downstream of SLAM engagement and that a genetic polymorphism that disturbs this process promotes the development of lupus.
Collapse
Affiliation(s)
- Nafiseh Talaei
- Arthritis Centre of Excellence, Toronto Western Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tao Yu
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Kieran Manion
- Arthritis Centre of Excellence, Toronto Western Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario M5S 1A1, Canada; and
| | - Joan E Wither
- Arthritis Centre of Excellence, Toronto Western Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
21
|
Shan SJ, Liu DZ, Wang L, Zhu YY, Zhang FM, Li T, An LG, Yang GW. Identification and expression analysis of irak1 gene in common carp Cyprinus carpio L.: indications for a role of antibacterial and antiviral immunity. JOURNAL OF FISH BIOLOGY 2015; 87:241-255. [PMID: 26099328 DOI: 10.1111/jfb.12714] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/23/2015] [Indexed: 06/04/2023]
Abstract
In this study, the full-length complementary (c)DNA of interleukin-1 receptor-associated kinase 1 gene (irak1) was cloned from common carp Cyprinus carpio. The complete open reading frame of irak1 contained 2109 bp encoding a protein of 702 amino acid residues that comprised a death domain, a ProST region, a serine-threonine-specific protein kinase catalytic domain and a C-terminal domain. The amino-acid sequence of C. carpio Irak1 protein shared sequence homology with grass carp Ctenopharyngodon idellus (84.5%). The phylogenetic tree of IRAKs separated the polypeptides into four clades, comprising IRAK1s, IRAK2s, IRAK3s and IRAK4s. Cyprinus carpio Irak1 fell into the cluster with previously reported IRAK1s including teleost Irak1s. The irak1 gene was highly expressed in gills, followed by brain, skin, hindgut, buccal epithelium, spleen, foregut, head kidney and liver, and was expressed at lowest levels in gonad and muscle. The irak1 messenger (m)RNA expression was up-regulated in liver, spleen, head kidney, foregut, hindgut, gills and skin after stimulation with Vibrio anguillarum and poly(I:C), and significantly high up-regulated expression was observed in liver and spleen. These results implied that irak1 might participate in antibacterial and antiviral innate immunity. These findings gave the indications that irak1 may participate in antibacterial and antiviral immunity.
Collapse
Affiliation(s)
- S J Shan
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - D Z Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - L Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - Y Y Zhu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - F M Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - T Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - L G An
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| | - G W Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, Jinan, 250014, People's Republic of China
| |
Collapse
|
22
|
van Berlo D, Hullmann M, Schins RPF. Toxicology of ambient particulate matter. ACTA ACUST UNITED AC 2015; 101:165-217. [PMID: 22945570 DOI: 10.1007/978-3-7643-8340-4_7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is becoming increasingly clear that inhalation exposure to particulate matter (PM) can lead to or exacerbate various diseases, which are not limited to the lung but extend to the cardiovascular system and possibly other organs and tissues. Epidemiological studies have provided strong evidence for associations with chronic obstructive pulmonary disease (COPD), asthma, bronchitis and cardiovascular disease, while the evidence for a link with lung cancer is less strong. Novel research has provided first hints that exposure to PM might lead to diabetes and central nervous system (CNS) pathology. In the current review, an overview is presented of the toxicological basis for adverse health effects that have been linked to PM inhalation. Oxidative stress and inflammation are discussed as central processes driving adverse effects; in addition, profibrotic and allergic processes are implicated in PM-related diseases. Effects of PM on key cell types considered as regulators of inflammatory, fibrotic and allergic mechanisms are described.
Collapse
Affiliation(s)
- Damiën van Berlo
- Particle Research, Institut für Umweltmedizinische Forschung (IUF), Heinrich-Heine University Düsseldorf, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | | | | |
Collapse
|
23
|
Chow NA, Jasenosky LD, Goldfeld AE. A distal locus element mediates IFN-γ priming of lipopolysaccharide-stimulated TNF gene expression. Cell Rep 2014; 9:1718-1728. [PMID: 25482561 PMCID: PMC4268019 DOI: 10.1016/j.celrep.2014.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 10/16/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022] Open
Abstract
Interferon γ (IFN-γ) priming sensitizes monocytes and macrophages to lipopolysaccharide (LPS) stimulation, resulting in augmented expression of a set of genes including TNF. Here, we demonstrate that IFN-γ priming of LPS-stimulated TNF transcription requires a distal TNF/LT locus element 8 kb upstream of the TNF transcription start site (hHS-8). IFN-γ stimulation leads to increased DNase I accessibility of hHS-8 and its recruitment of interferon regulatory factor 1 (IRF1), and subsequent LPS stimulation enhances H3K27 acetylation and induces enhancer RNA synthesis at hHS-8. Ablation of IRF1 or targeting the hHS-8 IRF1 binding site in vivo with Cas9 linked to the KRAB repressive domain abolishes IFN-γ priming, but does not affect LPS induction of the gene. Thus, IFN-γ poises a distal enhancer in the TNF/LT locus by chromatin remodeling and IRF1 recruitment, which then drives enhanced TNF gene expression in response to a secondary toll-like receptor (TLR) stimulus.
Collapse
Affiliation(s)
- Nancy A Chow
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Luke D Jasenosky
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Anne E Goldfeld
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Circadian rhythmicity, variability and correlation of interleukin-6 levels in plasma and cerebrospinal fluid of healthy men. Psychoneuroendocrinology 2014; 44:71-82. [PMID: 24767621 DOI: 10.1016/j.psyneuen.2014.02.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 01/02/2023]
Abstract
BACKGROUND Interleukin-6 (IL-6) is a cytokine with pleiotropic actions in both the periphery of the body and the central nervous system (CNS). Altered IL-6 secretion has been associated with inflammatory dysregulation and several adverse health consequences. However, little is known about the physiological circadian characteristics and dynamic inter-correlation between circulating and CNS IL-6 levels in humans, or their significance. METHODS Simultaneous assessment of plasma and cerebrospinal fluid (CSF) IL-6 levels was performed hourly in 11 healthy male volunteers over 24h, to characterize physiological IL-6 secretion levels in both compartments. RESULTS IL-6 levels showed considerable within- and between-subject variability in both plasma and CSF, with plasma/CSF ratios revealing consistently higher levels in the CSF. Both CSF and plasma IL-6 levels showed a distinctive circadian variation, with CSF IL-6 levels exhibiting a main 24h, and plasma a biphasic 12h, circadian component. Plasma peaks were roughly at 4 p.m. and 4 a.m., while the CSF peak was at around 7 p.m. There was no correlation between coincident CSF and plasma IL-6 values, but evidence for significant correlations at a negative 7-8h time lag. CONCLUSIONS This study provides evidence in humans for a circadian IL-6 rhythm in CSF and confirms prior observations reporting a plasma biphasic circadian pattern. Our results indicate differential IL-6 regulation across the two compartments and are consistent with local production of IL-6 in the CNS. Possible physiological significance is discussed and implications for further research are highlighted.
Collapse
|
25
|
Park S, Choi JJ, Park BK, Yoon SJ, Choi JE, Jin M. Pheophytin a and chlorophyll a suppress neuroinflammatory responses in lipopolysaccharide and interferon-γ-stimulated BV2 microglia. Life Sci 2014; 103:59-67. [DOI: 10.1016/j.lfs.2014.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/21/2014] [Accepted: 03/31/2014] [Indexed: 01/05/2023]
|
26
|
Haghikia A, Ricke-Hoch M, Stapel B, Gorst I, Hilfiker-Kleiner D. STAT3, a key regulator of cell-to-cell communication in the heart. Cardiovasc Res 2014; 102:281-9. [PMID: 24518140 DOI: 10.1093/cvr/cvu034] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) is fundamental for physiological homeostasis and stress-induced remodelling of the heart as deregulated STAT3 circuits are sufficient to induce dilated and peripartum cardiomyopathy and adverse remodelling after myocardial infarction. STAT3 activity depends on multiple post-translational modifications (phosphorylation, acetylation, and dimerization). It is regulated by multiple receptor systems, which are coupled to positive and negative feedback loops to ensure physiological and beneficial action. Its intracellular functions are diverse as it acts as a signalling protein, a transcription factor but also participates in mitochondria energy production and protection. STAT3 modulates proliferation, differentiation, survival, oxidative stress, and/or metabolism in cardiomyocytes, fibroblasts, endothelial cells, progenitor cells, and various inflammatory cells. By regulating the secretome of these cardiac cells, STAT3 influences a broad range of intercellular communication systems. It thereby impacts on the communication between cardiomyocytes, the plasticity of the cardiac microenvironment, the vasculature, the extracellular matrix, and the inflammation in response to physiological and pathophysiological stress. Here, we sum up current knowledge on STAT3-mediated intra- and intercellular communication within the heterogeneous cellular network of the myocardium to co-ordinate complex biological processes and discuss STAT3-dependent targets as novel therapeutic concepts to treat various forms of heart disease.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | | | | | | | | |
Collapse
|
27
|
Interferon-regulatory factors determine macrophage phenotype polarization. Mediators Inflamm 2013; 2013:731023. [PMID: 24379524 PMCID: PMC3863528 DOI: 10.1155/2013/731023] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 01/09/2023] Open
Abstract
The mononuclear phagocyte system regulates tissue homeostasis as well as all phases of tissue injury and repair. To do so changing tissue environments alter the phenotype of tissue macrophages to assure their support for sustaining and amplifying their respective surrounding environment. Interferon-regulatory factors are intracellular signaling elements that determine the maturation and gene transcription of leukocytes. Here we discuss how several among the 9 interferon-regulatory factors contribute to macrophage polarization.
Collapse
|
28
|
Carroll CJ, Sayan BS, Bailey SG, McCormick J, Stephanou A, Latchman DS, Townsend PA. Regulation of myocardial interleukin-6 expression by p53 and STAT1. J Interferon Cytokine Res 2013; 33:542-8. [PMID: 23675777 DOI: 10.1089/jir.2012.0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cardiovascular diseases are a major cause of morbidity and mortality worldwide. The interferon inducible transcriptional activator signal transducer and activator of transcription-1 (STAT1) and p53 are two critical transcriptional factors that have pivotal roles in cardiac biology and pathology. Here we describe a novel interplay between these two key players that critically regulate the levels of the pleiotropic interleukin 6 (IL6) in the heart. We provide in vivo evidence to demonstrate that, in cardiac tissues, STAT1 is a positive regulator of IL6 expression and it competes with the suppressive effect of p53 to sustain basal IL6 levels. Induction of IL6 expression in response to interferon gamma (IFNγ), a well-characterized activator of STAT1, parallels that of STAT1 phosphorylation and induction of STAT1 target genes, such as the interferon regulatory factor-1 (IRF-1), major histocompatibility complex class II transactivator (C2ta), and β2-microglobulin (B2m). Furthermore, hearts from STAT1 knockout mice fail to induce IL6 expression in response to IFNγ. More importantly, we showed that this regulatory system is not functional in mouse embryonic fibroblasts, suggesting that activation of IL6 expression by STAT1 may be tissue specific. IL6 is a major effector of inflammation and cardiac hypertrophy, two major processes involved in heart failure, and therefore, understanding the molecular mechanisms regulating IL6 expression will enable better therapies and treatments for cardiovascular disease patients.
Collapse
Affiliation(s)
- Christopher J Carroll
- Research Programs Unit, Molecular Neurology, Biomedicum-Helsinki, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|
29
|
Collino M, Massimo C, Pini A, Alessandro P, Mastroianni R, Rosanna M, Benetti E, Elisa B, Lanzi C, Cecilia L, Bani D, Daniele B, Jacopo C, Manoni M, Marco M, Fantozzi R, Roberto F, Masini E, Emanuela M. The non-anticoagulant heparin-like K5 polysaccharide derivative K5-N,OSepi attenuates myocardial ischaemia/reperfusion injury. J Cell Mol Med 2013; 16:2196-207. [PMID: 22248092 PMCID: PMC3822989 DOI: 10.1111/j.1582-4934.2012.01530.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Heparin and low molecular weight heparins have been demonstrated to reduce myocardial ischaemia/reperfusion (I/R) injury, although their use is hampered by the risk of haemorrhagic and thrombotic complications. Chemical and enzymatic modifications of K5 polysaccharide have shown the possibility of producing heparin-like compounds with low anticoagulant activity and strong anti-inflammatory effects. Using a rat model of regional myocardial I/R, we investigated the effects of an epimerized N-,O-sulphated K5 polysaccharide derivative, K5-N,OSepi, on infarct size and histological signs of myocardial injury caused by 30 min. ligature of the left anterior descending coronary artery followed by 1 or 24 h reperfusion. K5-N,OSepi (0.1-1 mg/kg given i.v. 15 min. before reperfusion) significantly reduced the extent of myocardial damage in a dose-dependent manner. Furthermore, we investigated the potential mechanism(s) of the cardioprotective effect(s) afforded by K5-N,OSepi. In left ventricular samples, I/R induced mast cell degranulation and a robust increase in lipid peroxidation, free radical-induced DNA damage and calcium overload. Markers of neutrophil infiltration and activation were also induced by I/R in rat hearts, specifically myeloperoxidase activity, intercellular-adhesion-molecule-1 expression, prostaglandin-E(2) and tumour-necrosis-factor-α production. The robust increase in oxidative stress and inflammatory markers was blunted by K5-N,OSepi, in a dose-dependent manner, with maximum at 1 mg/kg. Furthermore, K5-N,OSepi administration attenuated the increase in caspase 3 activity, Bid and Bax activation and ameliorated the decrease in expression of Bcl-2 within the ischaemic myocardium. In conclusion, we demonstrate that the cardioprotective effect of the non-anticoagulant K5 derivative K5-N,OSepi is secondary to a combination of anti-apoptotic and anti-inflammatory effects.
Collapse
Affiliation(s)
| | - Collino Massimo
- Department of Anatomy, Pharmacology and Forensic Medicine, University of Turin, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Southworth T, Metryka A, Lea S, Farrow S, Plumb J, Singh D. IFN-γ synergistically enhances LPS signalling in alveolar macrophages from COPD patients and controls by corticosteroid-resistant STAT1 activation. Br J Pharmacol 2012; 166:2070-83. [PMID: 22352763 DOI: 10.1111/j.1476-5381.2012.01907.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE IFN-γ levels are increased in chronic obstructive airway disease (COPD) patients compared with healthy subjects and are further elevated during viral exacerbations. IFN-γ can 'prime' macrophages to enhance the response to toll-like receptor (TLR) ligands, such as LPS. The aim of this study was to examine the effect IFN-γ on corticosteroid sensitivity in alveolar macrophages (AM). EXPERIMENTAL APPROACH AM from non-smokers, smokers and COPD patients were stimulated with IFN-γ and/or LPS with or without dexamethasone. IL-6, TNF-α and IFN-γ-induced protein 10 kDa (IP-10) levels were measured by elisa, and Western blots were used to investigate the IFN-γ-stimulated Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway. Real-time PCR and flow cytometry were used to investigate TLR levels following IFN-γ treatment. KEY RESULTS In all three subject groups, IFN-γ alone had no effect on IL-6 and TNF-α production but enhanced the effects of LPS on these cytokines. In contrast, IFN-γ alone increased the production of IP-10. IFN-γ increased TLR2 and TLR4 expression in AM. Cytokine induction and STAT1 activation by IFN-γ were insensitive to dexamethasone for all groups. The inhibition of JAK and STAT1 repressed all these IFN-γ effects. CONCLUSIONS AND IMPLICATIONS Our results demonstrate that IFN-γ-induced STAT-1 signalling is corticosteroid resistant in AMs, and that targeting IFN-γ signalling by JAK inhibitors is a potentially novel anti-inflammatory strategy in COPD.
Collapse
Affiliation(s)
- T Southworth
- Manchester Academic Health Centre, NIHR Translational Research Facility, University Hospital of South Manchester Foundation Trust, Manchester, UK.
| | | | | | | | | | | |
Collapse
|
31
|
Subacute inflammatory activation in subjects with acute coronary syndrome and left ventricular dysfunction. Inflammation 2012; 35:363-70. [PMID: 21487907 DOI: 10.1007/s10753-011-9326-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Several lines of evidence indicate that increased inflammatory cytokine levels can be used for risk prediction in patients with acute coronary syndrome (ACS). This study therefore aimed to evaluate correlations between levels of soluble interleukin (IL)-2 receptor (sIL-2r), IL-6, and IL-8 and in-hospital incidence of acute heart failure (AHF) and left ventricular (LV) systolic dysfunction in the subacute phase of ACS. In 48 consecutive patients with ACS, circulating levels of sIL-2r, IL-6, and IL-8 were ascertained 72-96 h after onset of symptoms. Clinical data, LV function, and in-hospital incidence of AHF were also evaluated. IL-8 levels were significantly higher in patients with pulmonary edema (1,829 ± 2,496 vs 456 ± 624 pg/ml, p < 0.05); sIL-2r, IL-6, and IL-8 levels were increased proportionally to Killip class (r = 0.35, p < 0.05; r = 0.48, r = 0.47, p < 0.01) and in patients with LV ejection fraction (LVEF) < 30%. Levels of sIL-2r were inversely related to LVEF in subjects with acute myocardial infarction (r = -0.51, p < 0.05). Soluble IL-2r and IL-8 levels were related to mitral regurgitation severity (r = 0.34, p < 0.05; r = 0.37, p < 0.05). Levels of sIL-2 were proportional to LV end-diastolic diameter (r = 0.49, p < 0.001) and LV end-systolic diameter (r = 0.58, p < 0.001). Number of cytokines with circulating values above upper level of normal was significantly correlated with Killip class and LVEF (r = 0.40, r = -0.38, p < 0.05). sIL-2r, IL-6, and IL-8 are increased in patients with ACS and systolic dysfunction or AHF. These data suggest that inflammatory cytokine activity detectable in peripheral blood may be useful in identifying subjects with a worse clinical course.
Collapse
|
32
|
Lee K, Na W, Lee JY, Na J, Cho H, Wu H, Yune TY, Kim WS, Ju BG. Molecular mechanism of Jmjd3-mediated interleukin-6 gene regulation in endothelial cells underlying spinal cord injury. J Neurochem 2012; 122:272-82. [PMID: 22578249 DOI: 10.1111/j.1471-4159.2012.07786.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The inflammatory response contributes substantially to secondary injury cascades after spinal cord injury, with both neurotoxic and protective effects. However, epigenetic regulations of inflammatory genes following spinal cord injury have yet to be characterized thoroughly. In this study, we found that histone H3K27me3 demethylase Jmjd3 expression is acutely up-regulated in blood vessels of the injured spinal cord. We also observed up-regulation of Jmjd3 gene expression in bEnd.3 endothelial cells that were subjected to oxygen-glucose deprivation/reperfusion injury. When Jmjd3 was depleted by siRNA, oxygen-glucose deprivation/reperfusion injury-induced up-regulation of IL-6 was significantly inhibited. In addition, Jmjd3 associated with NF-κB (p65/p50) and CCAAT-enhancer-binding protein β at the IL-6 gene promoter. The recruitment of Jmjd3 coincided with decreased levels of tri-methylated H3K27 as well as increased levels of mono-methylated H3K27 at the IL-6 gene promoter. Furthermore, Jmjd3 depletion did not result in significant changes of methylation level of H3K27 at the IL-6 gene promoter. Collectively, our findings imply that Jmjd3-mediated H3K27me3 demethylation is crucial for IL-6 gene activation in endothelial cells, and this molecular event may regulate acute inflammatory response and integrity of the blood-spinal cord barrier following spinal cord injury.
Collapse
Affiliation(s)
- Kwanghyun Lee
- Department of Life Science, Sogang University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Batra S, Balamayooran G, Sahoo MK. Nuclear factor-κB: a key regulator in health and disease of lungs. Arch Immunol Ther Exp (Warsz) 2011; 59:335-51. [PMID: 21786215 PMCID: PMC7079756 DOI: 10.1007/s00005-011-0136-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 03/02/2011] [Indexed: 12/27/2022]
Abstract
Rel/NF-κB transcription factors play a key role in modulating the response of immunoregulatory genes including cytokines and chemokines, cell adhesion molecules, acute phase proteins, and anti-microbial peptides. Furthermore, an array of genes important for angiogenesis, tumor invasion and metastasis is also regulated by nuclear factor-κB (NF-κB). Close association of NF-κB with inflammation and tumorigenesis makes it an attractive target for basic research as well as for pharmaceutical industries. Studies involving various animal and cellular models have revealed the importance of NF-κB in pathobiology of lung diseases. This review (a) describes structures, activities, and regulation of NF-κB family members; (b) provides information which implicates NF-κB in pathogenesis of pulmonary inflammation and cancer; and (c) discusses information about available synthetic and natural compounds which target NF-κB or specific components of NF-κB signal transduction pathway and which may provide the foundation for development of effective therapy for lung inflammation and bronchogenic carcinomas.
Collapse
Affiliation(s)
- Sanjay Batra
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, 70803, USA.
| | | | | |
Collapse
|
34
|
Shen M, Bunaciu RP, Congleton J, Jensen HA, Sayam LG, Varner JD, Yen A. Interferon regulatory factor-1 binds c-Cbl, enhances mitogen activated protein kinase signaling and promotes retinoic acid-induced differentiation of HL-60 human myelo-monoblastic leukemia cells. Leuk Lymphoma 2011; 52:2372-9. [PMID: 21740303 DOI: 10.3109/10428194.2011.603449] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
All-trans retinoic acid (RA) and interferons (IFNs) have efficacy in treating certain leukemias and lymphomas, respectively, motivating interest in their mechanism of action to improve therapy. Both RA and IFNs induce interferon regulatory factor-1 (IRF-1). We find that in HL-60 myeloblastic leukemia cells which undergo mitogen activated protien kinase (MAPK)-dependent myeloid differentiation in response to RA, IRF-1 propels differentiation. RA induces MAPK-dependent expression of IRF-1. IRF-1 binds c-Cbl, a MAPK related adaptor. Ectopic IRF-1 expression causes CD38 expression and activation of the Raf/MEK/ERK axis, and enhances RA-induced differentiation by augmenting CD38, CD11b, respiratory burst and G0 arrest. Ectopic IRF-1 expression also decreases the activity of aldehyde dehydrogenase 1, a stem cell marker, and enhances RA-induced ALDH1 down-regulation. Interestingly, expression of aryl hydrocarbon receptor (AhR), which is RA-induced and known to down-regulate Oct4 and drive RA-induced differentiation, also enhances IRF-1 expression. The data are consistent with a model whereby IRF-1 acts downstream of RA and AhR to enhance Raf/MEK/ERK activation and propel differentiation.
Collapse
Affiliation(s)
- Miaoqing Shen
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
NF-κB transcription factors are critical regulators of immunity, stress responses, apoptosis and differentiation. A variety of stimuli coalesce on NF-κB activation, which can in turn mediate varied transcriptional programs. Consequently, NF-κB-dependent transcription is not only tightly controlled by positive and negative regulatory mechanisms but also closely coordinated with other signaling pathways. This intricate crosstalk is crucial to shaping the diverse biological functions of NF-κB into cell type- and context-specific responses.
Collapse
|
36
|
Kasten-Jolly J, Heo Y, Lawrence DA. Central nervous system cytokine gene expression: modulation by lead. J Biochem Mol Toxicol 2011; 25:41-54. [PMID: 21322097 DOI: 10.1002/jbt.20358] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The environmental heavy metal toxicant, lead (Pb) has been shown to be more harmful to the central nervous system (CNS) of children than to adults, given that Pb exposure affects the neural system during development. Because growth factors and cytokines play very important roles in development of the CNS, we have examined the impact of Pb exposure on the expression of cytokines during CNS development. Cytokine expression was studied in post-natal-day 21 (pnd21) mice by microarray, real-time RT-PCR, Luminex, and ELISA methodologies. BALB/c mouse pups were exposed to Pb through the dam's drinking water (0.1 mM Pb acetate), from gestation-day 8 (gd8) to pnd21. Two cytokines, interleukin-6 (IL-6) and transforming growth factor-β1 (TGF-β1), displayed significantly changed transcript levels in the presence of Pb. IL-6 and TGF-β1 both have signal transduction cascades that can cooperatively turn on the gene for the astrocyte marker glial-fibrillary acidic protein (GFAP). Microarray results indicated that Pb exposure significantly increased expression of GFAP. Pb also modulated IL-6, TGF-β1, and IL-18 protein expression in select brain regions. The deleterious effects of Pb on learning and long-term memory are posited to result from excessive astrocyte growth and/or activation with concomitant interference with neural connections. Differential neural expression of cytokines in brain regions needs to be further investigated to mechanistically associate Pb and neuroinflammation with behavioral and cognitive changes.
Collapse
Affiliation(s)
- Jane Kasten-Jolly
- Laboratory of Clinical and Experimental Endocrinology and Immunology, Wadsworth Center, 120 New Scotland Avenue, Albany, NY 12208, USA
| | | | | |
Collapse
|
37
|
Borgatti M, Mancini I, Bianchi N, Guerrini A, Lampronti I, Rossi D, Sacchetti G, Gambari R. Bergamot (Citrus bergamia Risso) fruit extracts and identified components alter expression of interleukin 8 gene in cystic fibrosis bronchial epithelial cell lines. BMC BIOCHEMISTRY 2011; 12:15. [PMID: 21496221 PMCID: PMC3095539 DOI: 10.1186/1471-2091-12-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 04/15/2011] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) airway pathology is a fatal, autosomal, recessive genetic disease characterized by extensive lung inflammation. After induction by TNF-α, elevated concentrations of several pro-inflammatory cytokines (i.e. IL-6, IL-1β) and chemokines (i.e. IL-8) are released from airway epithelial cells. In order to reduce the excessive inflammatory response in the airways of CF patients, new therapies have been developed and in this respect, medicinal plant extracts have been studied. In this article we have investigated the possible use of bergamot extracts (Citrus bergamia Risso) and their identified components to alter the expression of IL-8 associated with the cystic fibrosis airway pathology. METHODS The extracts were chemically characterized by 1H-NMR (nuclear magnetic resonance), GC-FID (gas chromatography-flame ionization detector), GC-MS (gas chromatography-mass spectrometry) and HPLC (high pressure liquid chromatography). Both bergamot extracts and main detected chemical constituents were assayed for their biological activity measuring (a) cytokines and chemokines in culture supernatants released from cystic fibrosis IB3-1 cells treated with TNF-α by Bio-Plex cytokine assay; (b) accumulation of IL-8 mRNA by real-time PCR. RESULTS The extracts obtained from bergamot (Citrus bergamia Risso) epicarps contain components displaying an inhibitory activity on IL-8. Particularly, the most active molecules were bergapten and citropten. These effects have been confirmed by analyzing mRNA levels and protein release in the CF cellular models IB3-1 and CuFi-1 induced with TNF-α or exposed to heat-inactivated Pseudomonas aeruginosa. CONCLUSIONS These obtained results clearly indicate that bergapten and citropten are strong inhibitors of IL-8 expression and could be proposed for further studies to verify possible anti-inflammatory properties to reduce lung inflammation in CF patients.
Collapse
Affiliation(s)
- Monica Borgatti
- Department of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara 74, Ferrara, 44121, Italy
| | - Irene Mancini
- Department of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara 74, Ferrara, 44121, Italy
| | - Nicoletta Bianchi
- Department of Biology and Evolution, Agro-technological and pharmaceutical Resources (Agri-Unife), University of Ferrara, Corso Ercole I d'Este, Ferrara, 44121, Italy
| | - Alessandra Guerrini
- Department of Biology and Evolution, Agro-technological and pharmaceutical Resources (Agri-Unife), University of Ferrara, Corso Ercole I d'Este, Ferrara, 44121, Italy
| | - Ilaria Lampronti
- Department of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara 74, Ferrara, 44121, Italy
| | - Damiano Rossi
- Department of Biology and Evolution, Agro-technological and pharmaceutical Resources (Agri-Unife), University of Ferrara, Corso Ercole I d'Este, Ferrara, 44121, Italy
| | - Gianni Sacchetti
- Department of Biology and Evolution, Agro-technological and pharmaceutical Resources (Agri-Unife), University of Ferrara, Corso Ercole I d'Este, Ferrara, 44121, Italy
| | - Roberto Gambari
- Department of Biochemistry and Molecular Biology, University of Ferrara, Via Fossato di Mortara 74, Ferrara, 44121, Italy
| |
Collapse
|
38
|
The G to A polymorphism at -597 of the interleukin-6 gene is extremely rare in southern Han Chinese. Cytokine 2011; 55:1-3. [PMID: 21474330 DOI: 10.1016/j.cyto.2011.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 12/11/2010] [Accepted: 03/11/2011] [Indexed: 01/13/2023]
Abstract
Interleukin-6 (IL-6) is a cytokine involved in different physiologic and pathophysiologic processes including essential hypertension (EH). Associations of the IL-6 promoter region polymorphisms with circulating level of IL-6 have been reported in various studies. We detected the IL-6-597G/A polymorphism in 246 EH patients and 194 healthy controls from Jiangsu area (south of China). Individuals all carried the GG wild genotype, no GA or AA genotypes were found. Our results suggest that IL-6-597G/A polymorphism is extremely rare and unlikely to be contributing significantly to disease susceptibility in southern Han Chinese.
Collapse
|
39
|
NF-κB-mediated anti-inflammatory activity of the sesquiterpene lactone 7-hydroxyfrullanolide. Eur J Pharmacol 2011; 657:41-50. [DOI: 10.1016/j.ejphar.2011.01.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/06/2011] [Accepted: 01/20/2011] [Indexed: 10/18/2022]
|
40
|
Scharl M, Hruz P, McCole DF. Protein tyrosine phosphatase non-receptor Type 2 regulates IFN-γ-induced cytokine signaling in THP-1 monocytes. Inflamm Bowel Dis 2010; 16:2055-64. [PMID: 20848498 DOI: 10.1002/ibd.21325] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND We have previously shown that the Crohn's disease (CD)-associated gene protein tyrosine phosphatase non-receptor Type 2 (PTPN2) regulates interferon gamma (IFN-γ)-induced signaling and barrier function in intestinal epithelial cells. Overactivation of immature immune cells has been demonstrated in CD and elevated levels of proinflammatory cytokines, such as IFN-γ, play an important pathophysiological role in this disease. Here we studied the role of PTPN2 in the regulation of IFN-γ-induced signaling in THP-1 monocytic cells. METHODS Protein analysis was performed by Western blotting, PTPN2 knockdown was induced by siRNA, and cytokine levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS We demonstrated that IFN-γ (1000 U/mL) treatment of THP-1 cells elevates PTPN2 protein, reaching a peak by 24 hours. Increased PTPN2 expression, in turn, correlated with decreased activity of the signaling molecules, signal transducer and activator of transcription (STAT) 1 and STAT3. Loss of PTPN2 potentiated IFN-γ-induced phosphorylation of both of the STATs and of the mitogen-activated protein kinase (MAPK) family member, p38. However, PTPN2 loss did not affect the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 or c-Jun N-terminal kinase. As a functional consequence, PTPN2 knockdown elevated the IFN-γ-induced secretion of the proinflammatory cytokines interleukin-6 (IL-6) and macrophage chemoattractant protein 1 (MCP-1). CONCLUSIONS Our data demonstrate that IFN-γ enhances PTPN2 protein in THP-1 cells and loss of PTPN2 promotes IFN-γ-induced STAT signaling and secretion of IL-6 and MCP-1. Therefore, we show that PTPN2 regulates inflammation-related events and PTPN2 dysregulation may contribute to the onset as well as to the perpetuation of inflammatory events associated with CD.
Collapse
Affiliation(s)
- Michael Scharl
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California, USA
| | | | | |
Collapse
|
41
|
Schmid S, Mordstein M, Kochs G, García-Sastre A, Tenoever BR. Transcription factor redundancy ensures induction of the antiviral state. J Biol Chem 2010; 285:42013-22. [PMID: 20943654 DOI: 10.1074/jbc.m110.165936] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The transcriptional response to virus infection is thought to be predominantly induced by interferon (IFN) signaling. Here we demonstrate that, in the absence of IFN signaling, an IFN-like transcriptome is still maintained. This transcriptional activity is mediated from IFN-stimulated response elements (ISREs) that bind to both the IFN-stimulated gene factor 3 (ISGF3) as well as to IFN response factor 7 (IRF7). Through a combination of both in vitro biochemistry and in vivo transcriptional profiling, we have dissected what constitutes IRF-specific, ISGF3-specific, or universal ISREs. Taken together, the data presented here suggest that IRF7 can induce an IFN-like transcriptome in the absence of type-I or -III signaling and therefore provides a level of redundancy to cells to ensure the induction of the antiviral state.
Collapse
Affiliation(s)
- Sonja Schmid
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | |
Collapse
|
42
|
Madonna S, Scarponi C, Sestito R, Pallotta S, Cavani A, Albanesi C. The IFN-gamma-dependent suppressor of cytokine signaling 1 promoter activity is positively regulated by IFN regulatory factor-1 and Sp1 but repressed by growth factor independence-1b and Krüppel-like factor-4, and it is dysregulated in psoriatic keratinocytes. THE JOURNAL OF IMMUNOLOGY 2010; 185:2467-81. [PMID: 20644166 DOI: 10.4049/jimmunol.1001426] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epidermal keratinocytes can counteract the detrimental effects of IFN-gamma by inducing the expression of suppressor of cytokine signaling (SOCS)1, which plays an important anti-inflammatory and self-protective role. To date, limited information exists on its expression and regulation in human diseased keratinocytes. In this study, we compared the expression levels of SOCS1 in keratinocytes isolated from skin affected by psoriasis with cells obtained from healthy donors, unveiling that keratinocytes are more prone than healthy cells to upregulate SOCS1 mRNA expression in response to IFN-gamma. We explored the regulatory mechanisms involved in socs1 gene transcription, and found that Sp1 and IFN regulatory factor-1 transcription factors are, respectively, responsible for the basal and IFN-gamma-induced activity of human socs1 promoter. In parallel, we demonstrated that socs1 promoter is negatively regulated by two transcriptional repressors, namely, growth factor independence-1b and Krüppel-like factor 4, which tightly control SOCS1 transcription on IFN-gamma stimulation. Interestingly, although the expression of Sp1 and IFN regulatory factor-1 activators of socs1 promoter is unaltered, growth factor independence-1b and Krüppel-like factor 4 are significantly reduced in psoriatic compared with healthy keratinocytes. This reduction and the consequent unbalanced binding of transcriptional activators and repressors to socs1 promoter after IFN-gamma stimulation might be responsible for the enhanced expression of SOCS1 in psoriatic cells. We suggest that SOCS1 exaggerated upregulation in psoriatic keratinocytes could represent a mechanism through which these cells attempt to protect themselves from IFN-gamma effects. However, the SOCS1 increased levels in psoriatic keratinocytes are not sufficient to completely inhibit the expression of proinflammatory genes.
Collapse
Affiliation(s)
- Stefania Madonna
- Laboratorio di Immunologia Sperimentale, Istituto Dermopatico dell'Immacolata, Rome, Italy
| | | | | | | | | | | |
Collapse
|
43
|
Dagia NM, Agarwal G, Kamath DV, Chetrapal-Kunwar A, Gupte RD, Jadhav MG, Dadarkar SS, Trivedi J, Kulkarni-Almeida AA, Kharas F, Fonseca LC, Kumar S, Bhonde MR. A preferential p110alpha/gamma PI3K inhibitor attenuates experimental inflammation by suppressing the production of proinflammatory mediators in a NF-kappaB-dependent manner. Am J Physiol Cell Physiol 2010; 298:C929-41. [PMID: 20089935 DOI: 10.1152/ajpcell.00461.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A promising therapeutic approach to diminish pathological inflammation is to inhibit the increased production and/or biological activity of proinflammatory cytokines (e.g., TNF-alpha, IL-6). The production of proinflammatory cytokines is controlled at the gene level by the activity of transcription factors, such as NF-kappaB. Phosphatidylinositol 3-kinase (PI3K), a lipid kinase, is known to induce the activation of NF-kappaB. Given this, we hypothesized that inhibitors of PI3K activation would demonstrate anti-inflammatory potential. Accordingly, we studied the effects of a preferential p110alpha/gamma PI3K inhibitor (compound 8C; PIK-75) in inflammation-based assays. Mechanism-based assays utilizing human cells revealed that PIK-75-mediated inhibition of PI3K activation is associated with dramatic suppression of downstream signaling events, including AKT phosphorylation, IKK activation, and NF-kappaB transcription. Cell-based assays revealed that PIK-75 potently and dose dependently inhibits in vitro and in vivo production of TNF-alpha and IL-6, diminishes the induced expression of human endothelial cell adhesion molecules (E-selectin, ICAM-1, and VCAM-1), and blocks human monocyte-endothelial cell adhesion. Most importantly, PIK-75, when administered orally in a therapeutic regimen, significantly suppresses the macroscopic and histological abnormalities associated with dextran sulfate sodium-induced murine colitis. The efficacy of PIK-75 in attenuating experimental inflammation is mediated, at least in part, due to the downregulation of pertinent inflammatory mediators in the colon. Collectively, these results provide first evidence that PIK-75 possesses anti-inflammatory potential. Given that PIK-75 is known to exhibit anti-cancer activity, the findings from this study thus reinforce the cross-therapeutic functionality of potential drugs.
Collapse
Affiliation(s)
- Nilesh M Dagia
- Dept. of Pharmacology, Piramal Life Sciences Limited, 1 Nirlon Complex, Off. Western Express Highway, Goregaon (East), Mumbai 400063, Maharashtra, India.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Guruceaga E, Segura V, Corrales FJ, Rubio A. Genome-wide proximal promoter analysis and interpretation. Methods Mol Biol 2010; 593:157-174. [PMID: 19957149 DOI: 10.1007/978-1-60327-194-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
High-throughput gene expression technologies based on DNA microarrays allow the examination of biological systems. However, the interpretation of the complex molecular descriptions generated by these approaches is still challenging. The development of new methodologies to identify common regulatory mechanisms involved in the control of the expression of a set of co-expressed genes might enhance our capacity to extract functional information from genomic data sets. In this chapter, we describe a method that integrates different sources of information: gene expression data, genome sequence information, described transcription factor binding sites (TFBSs), functional information, and bibliographic data. The starting point of the analysis is the extraction of promoter sequences from a whole genome and the detection of TFBSs in each gene promoter. This information allows the identification of enriched TFBSs in the proximal promoter of differentially expressed genes. The functional and bibliographic interpretation of the results improves our biological insight into the regulatory mechanisms involved in a microarray experiment.
Collapse
Affiliation(s)
- Elizabeth Guruceaga
- CEIT, Centro de Estudios e Investigaciones Técnicas de Gipuzkoa, San Sebastian, Spain
| | | | | | | |
Collapse
|
45
|
Liu MY, Khachigian LM. Histone deacetylase-1 is enriched at the platelet-derived growth factor-D promoter in response to interleukin-1beta and forms a cytokine-inducible gene-silencing complex with NF-kappab p65 and interferon regulatory factor-1. J Biol Chem 2009; 284:35101-12. [PMID: 19843519 DOI: 10.1074/jbc.m109.061903] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Understanding the mechanisms governing cytokine control of growth factor expression in smooth muscle cells would provide invaluable insight into the molecular regulation of vascular phenotypes and create future opportunities for therapeutic intervention. Here, we report that the proinflammatory cytokine interleukin (IL)-1beta suppresses platelet-derived growth factor (PDGF)-D promoter activity and mRNA and protein expression in smooth muscle cells. NF-kappaB p65, induced by IL-1beta, interacts with a novel element in the PDGF-D promoter and inhibits PDGF-D transcription. Interferon regulatory factor-1 (IRF-1) is also induced by IL-1beta and binds to a different element upstream in the promoter. Immunoprecipitation and chromatin immunoprecipitation experiments showed that IL-1beta stimulates p65 interaction with IRF-1 and the accumulation of both factors at the PDGF-D promoter. Mutation of the IRF-1 and p65 DNA-binding elements relieved the promoter from IL-1beta-mediated repression. PDGF-D repression by IL-1beta involves histone deacetylation and interaction of HDAC-1 with IRF-1 and p65. HDAC-1 small interfering RNA ablates complex formation with IRF-1 and p65 and abrogates IRF-1 and p65 occupancy of the PDGF-D promoter. Thus, HDAC-1 is enriched at the PDGF-D promoter in cells exposed to IL-1beta and forms a cytokine-inducible gene-silencing complex with p65 and IRF-1.
Collapse
Affiliation(s)
- Mary Y Liu
- Centre for Vascular Research, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | |
Collapse
|
46
|
Molecular mechanisms underlying the pro-inflammatory synergistic effect of tumor necrosis factor alpha and interferon gamma in human microvascular endothelium. Eur J Cell Biol 2009; 88:731-42. [PMID: 19782427 DOI: 10.1016/j.ejcb.2009.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 07/14/2009] [Accepted: 07/14/2009] [Indexed: 11/22/2022] Open
Abstract
Tumor necrosis factor alpha (TNFalpha) and interferon gamma (IFNgamma) are among the most potent cytokines involved in orchestrating the inflammation response. The molecular mechanisms implicated in the synergism between cytokines are still poorly characterized. We demonstrate that both cytokines dose-dependently stimulate IFNgamma-inducible-protein-of-10-kDa (IP-10) secretion in human microvascular endothelial cells (HMEC-1), showing a potent synergism which is not restricted to IP-10, but is also evident for monokine-induced-by-IFNgamma (MIG) and IL-6 secretion. Immunofluorescence analysis reveals that TNFalpha and IFNgamma converge on a rapid phosphorylation of ERK, which however results in a different subcellular compartmentalization of the activated enzyme in response to the two cytokines. Differences in the subcellular recruitment of ERK in response to IFNgamma and TNFalpha are responsible for generating different ERK downstream signaling, which can thus synergize on the secretion of IP-10 as well as of other cytokines/chemokines. The importance of ERK activation in mediating the synergism of the two cytokines is further confirmed by the inhibitory effect of the anti-diabetic drug rosiglitazone and ERK blockers on IP-10, MIG and IL-6 secretion. A further mechanism of synergism involving the reciprocal upregulation of TNFalpha-RII and of IFNgamma-R, in response to IFNgamma and TNFalpha, respectively, was revealed by flow cytometry and quantitative real time RT-PCR analysis.
Collapse
|
47
|
Moxnes JF, Albert christophersen O. The Spanish flu as a worst case scenario? MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.1080/08910600701699067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
48
|
Wort SJ, Ito M, Chou PC, Mc Master SK, Badiger R, Jazrawi E, de Souza P, Evans TW, Mitchell JA, Pinhu L, Ito K, Adcock IM. Synergistic induction of endothelin-1 by tumor necrosis factor alpha and interferon gamma is due to enhanced NF-kappaB binding and histone acetylation at specific kappaB sites. J Biol Chem 2009; 284:24297-305. [PMID: 19592490 DOI: 10.1074/jbc.m109.032524] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Endothelin-1 (ET-1) is a potent vasoconstrictor and co-mitogen for vascular smooth muscle and is implicated in pulmonary vascular remodeling and the development of pulmonary arterial hypertension. Vascular smooth muscle is an important source of ET-1. Here we demonstrate synergistic induction of preproET-1 message RNA and release of mature peptide by a combination of tumor necrosis factor alpha (TNFalpha) and interferon gamma (IFNgamma) in primary human pulmonary artery smooth muscle cells. This induction was prevented by pretreatment with the histone acetyltransferase inhibitor anacardic acid. TNFalpha induced a rapid and prolonged pattern of nuclear factor (NF)-kappaB p65 subunit activation and binding to the native preproET-1 promoter. In contrast, IFNgamma induced a delayed activation of interferon regulatory factor-1 without any effect on NF-kappaB p65 nuclear localization or consensus DNA binding. However, we found cooperative p65 binding and histone H4 acetylation at distinct kappaB sites in the preproET-1 promoter after stimulation with both TNFalpha and IFNgamma. This was associated with enhanced recruitment of RNA polymerase II to the ATG start site and read-through of the ET-1 coding region. Understanding such mechanisms is crucial in determining the key control points in ET-1 release. This has particular relevance to developing novel treatments targeted at the inflammatory component of pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Stephen J Wort
- Department of Critical Care, National Heart and Lung Institute, Imperial College London, London SW3 6LY, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tanigawa T, Watanabe T, Higuchi K, Machida H, Okazaki H, Yamagami H, Watanabe K, Tominaga K, Fujiwara Y, Oshitani N, Arakawa T. Lansoprazole, a Proton Pump Inhibitor, Suppresses Production of Tumor Necrosis Factor-alpha and Interleukin-1beta Induced by Lipopolysaccharide and Helicobacter Pylori Bacterial Components in Human Monocytic Cells via Inhibition of Activation of Nuclear Factor-kappaB and Extracellular Signal-Regulated Kinase. J Clin Biochem Nutr 2009; 45:86-92. [PMID: 19590712 PMCID: PMC2704330 DOI: 10.3164/jcbn.08-267] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2008] [Accepted: 01/13/2009] [Indexed: 01/05/2023] Open
Abstract
Pathogenic bacterial components play critical roles in initiation of gastrointestinal inflammation via activation of intracellular signaling pathways which induce proinflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β. Lansoprazole (LANSO), a proton pump inhibitor, has been widely used for the treatment of peptic ulcers and reflux esophagitis due to its potent acid-suppressive effect. It has also been reported to have anti-inflammatory effects. In this study we investigated the effects of LANSO on the production of TNF-α and IL-1β induced by lipopolysaccharide (LPS) and Helicobacter pylori water-soluble extract (HpWE) in the human monocytic cell line (THP-1). LANSO (100 µM) significantly reduced mRNA expression and production of TNF-α and IL-1β by THP-1 cells stimulated by LPS and HpWE. LANSO inhibited phosphorylation and degradation of inhibitory factor κB-α (IκB-α) and phosphorylation of extracellular signal-regulated kinase (ERK) induced by LPS and HpWE in THP-1 cells. These findings suggest that LANSO exerts anti-inflammatory effects by suppressing induction of TNF-α and IL-1β via inhibition of nuclear factor (NF)-κB and ERK activation.
Collapse
Affiliation(s)
- Tetsuya Tanigawa
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Péant B, Diallo JS, Dufour F, Le Page C, Delvoye N, Saad F, Mes-Masson AM. Over-expression of IkappaB-kinase-epsilon (IKKepsilon/IKKi) induces secretion of inflammatory cytokines in prostate cancer cell lines. Prostate 2009; 69:706-18. [PMID: 19170126 DOI: 10.1002/pros.20912] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Elevated inflammatory cytokine levels in serum have been associated with advanced stage metastasis-related morbidity in prostate cancer. Several studies have shown that IL-6 and IL-8 can accelerate the growth of human prostate cancer cell lines. Previous studies, in murine embryonic fibroblasts, have shown that Ikappa-B kinase-epsilon (IKKepsilon/IKKi)-deficiency results in the reduction of lipopolysaccharide-mediated expression of IL-6. RESULTS In this study, we report that over-expression of IKKepsilon in hormone-sensitive 22Rv1 and LNCaP prostate cancer cells induces the secretion of several inflammatory cytokines including IL-6 and IL-8. Both of these cytokines are secreted by hormone-refractory PC-3 prostate cancer cells and IKKepsilon knock-down in these cells correlates with a strong decrease in IL-6 secretion. Furthermore, we demonstrate that IKKepsilon over-expression does not induce the activation of the IKKepsilon classical targets NF-kappaB and IRF-3, two transcription factors involved in the regulation of several cytokines. Finally, we observe that high IKKepsilon expression results in its nuclear translocation, a phenomena that is TBK1-independent. CONCLUSIONS This study identifies IKKepsilon as a potential prostate cancer gene that may favor chronic inflammation and create a tumor-supporting microenvironment that promotes prostate cancer progression, particularly by the induction of IL-6 secretion that may act as a positive growth factor in prostate cancer.
Collapse
Affiliation(s)
- Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|