201
|
Pasquereau S, Kumar A, Herbein G. Targeting TNF and TNF Receptor Pathway in HIV-1 Infection: from Immune Activation to Viral Reservoirs. Viruses 2017; 9:v9040064. [PMID: 28358311 PMCID: PMC5408670 DOI: 10.3390/v9040064] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Several cellular functions such as apoptosis, cellular proliferation, inflammation, and immune regulation involve the tumor necrosis factor-α (TNF)/TNF receptor (TNFR) pathway. Human immunodeficiency virus 1 (HIV-1) interacts with the TNF/TNFR pathway. The activation of the TNF/TNFR pathway impacts HIV-1 replication, and the TNF/TNFR pathway is the target of HIV-1 proteins. A hallmark of HIV-1 infection is immune activation and inflammation with increased levels of TNF in the plasma and the tissues. Therefore, the control of the TNF/TNFR pathway by new therapeutic approaches could participate in the control of immune activation and impact both viral replication and viral persistence. In this review, we will describe the intricate interplay between HIV-1 proteins and TNF/TNFR signaling and how TNF/TNFR activation modulates HIV-1 replication and discuss new therapeutic approaches, especially anti-TNF therapy, that could control this pathway and ultimately favor the clearance of infected cells to cure HIV-infected patients.
Collapse
Affiliation(s)
- Sébastien Pasquereau
- Department of Virology, University of Franche-Comte, University of Bourgogne-Franche-Comté (UBFC), CHRU Besançon, UPRES EA4266 Pathogens & Inflammation/EPILAB, SFR FED 4234, F-25030 Besançon, France.
| | - Amit Kumar
- Department of Virology, University of Franche-Comte, University of Bourgogne-Franche-Comté (UBFC), CHRU Besançon, UPRES EA4266 Pathogens & Inflammation/EPILAB, SFR FED 4234, F-25030 Besançon, France.
| | - Georges Herbein
- Department of Virology, University of Franche-Comte, University of Bourgogne-Franche-Comté (UBFC), CHRU Besançon, UPRES EA4266 Pathogens & Inflammation/EPILAB, SFR FED 4234, F-25030 Besançon, France.
| |
Collapse
|
202
|
Niu K, Xu J, Cao Y, Hou Y, Shan M, Wang Y, Xu Y, Sun M, Wang B. BAP31 is involved in T cell activation through TCR signal pathways. Sci Rep 2017; 7:44809. [PMID: 28333124 PMCID: PMC5363085 DOI: 10.1038/srep44809] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/15/2017] [Indexed: 12/15/2022] Open
Abstract
BAP31 is a ubiquitously expressed endoplasmic reticulum (ER) membrane protein. The functions of BAP31 in the immune system have not been investigated due to the lack of animal models. Therefore we created a BAP31 conditional knockdown mouse by performing a knockdown of BAP31 in the thymus. In doing so, we demonstrate that the maturation of T cells is normal but the number of T cells is less in the thymus of the knockout mouse. In addition, the spleen and lymph nodes of peripheral immune organs contained a lesser proportion of the mature T cells in the thymus specific BAP31 knockout mice. The BAP31 knockout T cells decreased the proliferation activated by TCR signal pathways. Further studies clarified that BAP31 affects the phosphorylation levels of both Zap70/Lck/Lat of the upstream members and Akt/GSK/Jnk/Erk of the downstream members of TCR signal pathways. Furthermore, BAP31 can regulate the expression of some markers such as CD3/TCRα/TCRβ and some cytokines like IL-2/IFN-γ/IL-6/TNF-α which are important for T cell activation. Taken together, these results demonstrate that BAP31 may play an important role in T cell activation by regulating TCR signaling.
Collapse
Affiliation(s)
- Kunwei Niu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Jialin Xu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Yuhua Cao
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Yue Hou
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Mu Shan
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Yanqing Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Yang Xu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Mingyi Sun
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| | - Bing Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang 110169, China
| |
Collapse
|
203
|
Herpes Simplex Virus 1 UL24 Abrogates the DNA Sensing Signal Pathway by Inhibiting NF-κB Activation. J Virol 2017; 91:JVI.00025-17. [PMID: 28100608 DOI: 10.1128/jvi.00025-17] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/22/2022] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is a newly identified DNA sensor that recognizes foreign DNA, including the genome of herpes simplex virus 1 (HSV-1). Upon binding of viral DNA, cGAS produces cyclic GMP-AMP, which interacts with and activates stimulator of interferon genes (STING) to trigger the transcription of antiviral genes such as type I interferons (IFNs), and the production of inflammatory cytokines. HSV-1 UL24 is widely conserved among members of the herpesviruses family and is essential for efficient viral replication. In this study, we found that ectopically expressed UL24 could inhibit cGAS-STING-mediated promoter activation of IFN-β and interleukin-6 (IL-6), and UL24 also inhibited interferon-stimulatory DNA-mediated IFN-β and IL-6 production during HSV-1 infection. Furthermore, UL24 selectively blocked nuclear factor κB (NF-κB) but not IFN-regulatory factor 3 promoter activation. Coimmunoprecipitation analysis demonstrated that UL24 bound to the endogenous NF-κB subunits p65 and p50 in HSV-1-infected cells, and UL24 was also found to bind the Rel homology domains (RHDs) of these subunits. Furthermore, UL24 reduced the tumor necrosis factor alpha (TNF-α)-mediated nuclear translocation of p65 and p50. Finally, mutational analysis revealed that the region spanning amino acids (aa) 74 to 134 of UL24 [UL24(74-134)] is responsible for inhibiting cGAS-STING-mediated NF-κB promoter activity. For the first time, UL24 was shown to play an important role in immune evasion during HSV-1 infection.IMPORTANCE NF-κB is a critical component of the innate immune response and is strongly induced downstream of most pattern recognition receptors (PRRs), leading to the production of IFN-β as well as a number of inflammatory chemokines and interleukins. To establish persistent infection, viruses have evolved various mechanisms to counteract the host NF-κB pathway. In the present study, for the first time, HSV-1 UL24 was demonstrated to inhibit the activation of NF-κB in the DNA sensing signal pathway via binding to the RHDs of the NF-κB subunits p65 and p50 and abolishing their nuclear translocation.
Collapse
|
204
|
Zeng YY, Feng L, Jiang WD, Liu Y, Wu P, Jiang J, Kuang SY, Tang L, Tang WN, Zhang YA, Zhou XQ. Dietary alpha-linolenic acid/linoleic acid ratios modulate immune response, physical barrier and related signaling molecules mRNA expression in the gills of juvenile grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2017; 62:1-12. [PMID: 28063950 DOI: 10.1016/j.fsi.2017.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/29/2016] [Accepted: 01/03/2017] [Indexed: 06/06/2023]
Abstract
This study was conducted to explore the possible effects of dietary ALA/LNA ratios on the gill immunity, tight junction and antioxidant capacity, and the related signaling factor mRNA levels of juvenile grass carp (Ctenopharyngodon idella). Fish were fed diets with different ALA/LNA ratios (0.01, 0.34, 0.68, 1.03, 1.41, 1.76 and 2.15) for 60 days. The present results showed that ALA/LNA ratio of 1.03 significantly enhanced lysozyme and acid phosphatase activities, complement 3 contents, promoted mRNA levels of antimicrobial peptides (Hepcidin and liver expression antimicrobial peptide-2), anti-inflammatory cytokines (interleukin 10 and transforming growth factor β1) and inhibitor protein κBα, whereas suppressed pro-inflammatory cytokines (interleukin 1β, interleukin 8, tumor necrosis factor a and interferon γ2), and signal molecules (IκB kinase β, IκB kines γ and nuclear factor κB p65) mRNA levels in the gill, indicating that optimal dietary ALA/LNA ratio improve gill immunity of juvenile fish. Besides, ALA/LNA ratio of 1.03 increased mRNA levels of the barrier functional proteins (occludin, zonula occludens-1, claudin-b, -c and -3), and reduced the pore-formation proteins (claudin-15a) and myosin light-chain kinase mRNA abundance in the gill of juvenile grass carp, indicating optimum ALA/LNA ratio strengthen gill tight junction of juvenile fish. Additionally, ALA/LNA ratio of 1.03 increased glutathione contents, copper/zinc superoxide dismutase, glutathione peroxidase, glutathione S-transferase and glutathione reductase activities and mRNA abundance, and nuclear factor erythoid 2-related factor 2 mRNA levels in the gill of fish, suggesting that optimal ALA/LNA ratio ameliorate gill antioxidant status of juvenile fish. Interestingly, dietary ALA/LNA ratios had no effect on IκB kinase α and catalase activities in fish gills. Collectively, optimal dietary ALA/LNA ratio could improve gill immunity and strengthen physical barrier of juvenile fish. Based on the quadratic regression analysis of complement 3 content in the gill, optimal dietary ALA/LNA ratio for maximum growth of juvenile grass carp was estimated to be 1.12.
Collapse
Affiliation(s)
- Yun-Yun Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
205
|
ZNF395 Is an Activator of a Subset of IFN-Stimulated Genes. Mediators Inflamm 2017; 2017:1248201. [PMID: 28316371 PMCID: PMC5339479 DOI: 10.1155/2017/1248201] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/05/2017] [Indexed: 11/17/2022] Open
Abstract
Activation of the interferon (IFN) pathway in response to infection with pathogens results in the induction of IFN-stimulated genes (ISGs) including proinflammatory cytokines, which mount the proper antiviral immune response. However, aberrant expression of these genes is pathogenic to the host. In addition to IFN-induced transcription factors non-IFN-regulated factors contribute to the transcriptional control of ISGs. Here, we show by genome wide expression analysis, siRNA-mediated suppression and Doxycycline-induced overexpression that the cellular transcription factor ZNF395 activates a subset of ISGs including the chemokines CXCL10 and CXCL11 in keratinocytes. We found that ZNF395 acts independently of IFN but enhances the IFN-induced expression of CXCL10 and CXCL11. Luciferase reporter assays revealed a requirement of intact NFκB-binding sites for ZNF395 to stimulate the CXCL10 promoter. The transcriptional activation of CXCL10 and CXCL11 by ZNF395 was abolished after inhibition of IKK by BMS-345541, which increased the stability of ZNF395. ZNF395 encodes at least two motifs that mediate the enhanced degradation of ZNF395 in response to IKK activation. Thus, IKK is required for ZNF395-mediated activation of transcription and enhances its turn-over to keep the activity of ZNF395 low. Our results support a previously unrecognized role of ZNF395 in the innate immune response and inflammation.
Collapse
|
206
|
Herpes Simplex Virus 1 Ubiquitin-Specific Protease UL36 Abrogates NF-κB Activation in DNA Sensing Signal Pathway. J Virol 2017; 91:JVI.02417-16. [PMID: 28031360 DOI: 10.1128/jvi.02417-16] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 01/09/2023] Open
Abstract
The DNA sensing pathway triggers innate immune responses against DNA virus infection, and NF-κB signaling plays a critical role in establishing innate immunity. We report here that the herpes simplex virus 1 (HSV-1) ubiquitin-specific protease (UL36USP), which is a deubiquitinase (DUB), antagonizes NF-κB activation, depending on its DUB activity. In this study, ectopically expressed UL36USP blocked promoter activation of beta interferon (IFN-β) and NF-κB induced by cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING). UL36USP restricted NF-κB activation mediated by overexpression of STING, TANK-binding kinase 1, IκB kinase α (IKKα), and IKKβ, but not p65. UL36USP was also shown to inhibit IFN-stimulatory DNA-induced IFN-β and NF-κB activation under conditions of HSV-1 infection. Furthermore, UL36USP was demonstrated to deubiquitinate IκBα and restrict its degradation and, finally, abrogate NF-κB activation. More importantly, the recombinant HSV-1 lacking UL36USP DUB activity, denoted as C40A mutant HSV-1, failed to cleave polyubiquitin chains on IκBα. For the first time, UL36USP was shown to dampen NF-κB activation in the DNA sensing signal pathway to evade host antiviral innate immunity.IMPORTANCE It has been reported that double-stranded-DNA-mediated NF-κB activation is critical for host antiviral responses. Viruses have established various strategies to evade the innate immune system. The N terminus of the HSV-1 UL36 gene-encoded protein contains the DUB domain and is conserved across all herpesviruses. This study demonstrates that UL36USP abrogates NF-κB activation by cleaving polyubiquitin chains from IκBα and therefore restricts proteasome-dependent degradation of IκBα and that DUB activity is indispensable for this process. This study expands our understanding of the mechanisms utilized by HSV-1 to evade the host antiviral innate immune defense induced by NF-κB signaling.
Collapse
|
207
|
Tse AKW, Chen YJ, Fu XQ, Su T, Li T, Guo H, Zhu PL, Kwan HY, Cheng BCY, Cao HH, Lee SKW, Fong WF, Yu ZL. Sensitization of melanoma cells to alkylating agent-induced DNA damage and cell death via orchestrating oxidative stress and IKKβ inhibition. Redox Biol 2017; 11:562-576. [PMID: 28107677 PMCID: PMC5247288 DOI: 10.1016/j.redox.2017.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/11/2017] [Accepted: 01/11/2017] [Indexed: 01/22/2023] Open
Abstract
Nitrosourea represents one of the most active classes of chemotherapeutic alkylating agents for metastatic melanoma. Treatment with nitrosoureas caused severe systemic side effects which hamper its clinical use. Here, we provide pharmacological evidence that reactive oxygen species (ROS) induction and IKKβ inhibition cooperatively enhance nitrosourea-induced cytotoxicity in melanoma cells. We identified SC-514 as a ROS-inducing IKKβ inhibitor which enhanced the function of nitrosoureas. Elevated ROS level results in increased DNA crosslink efficiency triggered by nitrosoureas and IKKβ inhibition enhances DNA damage signals and sensitizes nitrosourea-induced cell death. Using xenograft mouse model, we confirm that ROS-inducing IKKβ inhibitor cooperates with nitrosourea to reduce tumor size and malignancy in vivo. Taken together, our results illustrate a new direction in nitrosourea treatment, and reveal that the combination of ROS-inducing IKKβ inhibitors with nitrosoureas can be potentially exploited for melanoma therapy.
Collapse
Affiliation(s)
- Anfernee Kai-Wing Tse
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Ying-Jie Chen
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xiu-Qiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Tao Su
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ting Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hui Guo
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Pei-Li Zhu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hiu-Yee Kwan
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Brian Chi-Yan Cheng
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Hui-Hui Cao
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Sally Kin-Wah Lee
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Wang-Fun Fong
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| |
Collapse
|
208
|
Rodríguez-Calvo R, Tajes M, Vázquez-Carrera M. The NR4A subfamily of nuclear receptors: potential new therapeutic targets for the treatment of inflammatory diseases. Expert Opin Ther Targets 2017; 21:291-304. [PMID: 28055275 DOI: 10.1080/14728222.2017.1279146] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Prolonged inflammatory response contributes to the pathogenesis of chronic disease-related disturbances. Among nuclear receptors (NRs), the orphan NR4A subfamily, which includes Nur77 (NR4A1), Nurr1 (NR4A2) and NOR1 (NR4A3), has recently emerged as a therapeutic target for the treatment of inflammation. Areas covered: This review focuses on the capacity of NR4A receptors to counter-regulate the development of the inflammatory response, with a special focus on the molecular transrepression mechanisms. Expert opinion: Recent studies have highlighted the role of NR4A receptors as significant regulators of the inflammatory response. NR4A receptors are rapidly induced by inflammatory stimuli, thus suggesting that they are required for the initiation of inflammation. Nevertheless, NR4A anti-inflammatory properties indicate that this acute regulation could be a protective reaction aimed at resolving inflammation in the later stages. Therefore, NR4A receptors are involved in a negative feedback mechanism to maintain the inflammatory balance. However, the underlying mechanisms are not entirely clear. Only a small number of NR4A-target genes have been identified, and the transcriptional repression mechanisms are only beginning to emerge. Despite further research is needed to fully understand the role of NR4A receptors in inflammation, these NRs should be considered as targets for new therapeutic approaches to inflammatory diseases.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- a Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Pere Virgili Health Research Institute (IISPV) and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)-Instituto de Salud Carlos III, Faculty of Medicine and Health Sciences , Rovira i Virgili University , Reus , Spain
| | - Marta Tajes
- b Heart Diseases Biomedical Research Group, Inflammatory and Cardiovascular Disorders Program , Hospital del Mar Medical Research Institute (IMIM), Parc de Salut Mar , Barcelona , Spain
| | - Manuel Vázquez-Carrera
- c Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Pediàtrica-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)-Instituto de Salud Carlos III, Faculty of Pharmacy, Diagonal 643 , University of Barcelona , Barcelona , Spain
| |
Collapse
|
209
|
Durand JK, Baldwin AS. Targeting IKK and NF-κB for Therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2017; 107:77-115. [PMID: 28215229 DOI: 10.1016/bs.apcsb.2016.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In addition to regulating immune responses, the NF-κB family of transcription factors also promotes cellular proliferation and survival. NF-κB and its activating kinase, IKK, have become appealing therapeutic targets because of their critical roles in the progression of many diseases including chronic inflammation and cancer. Here, we discuss the conditions that lead to pathway activation, the effects of constitutive activation, and some of the strategies used to inhibit NF-κB signaling.
Collapse
Affiliation(s)
- J K Durand
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, United States; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - A S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
210
|
Cytokines in Hepatic Injury. LIVER PATHOPHYSIOLOGY 2017. [DOI: 10.1016/b978-0-12-804274-8.00027-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
211
|
Role for NF-κB inflammatory signalling pathway in tenofovir disoproxil fumarate (TDF) induced renal damage in rats. Food Chem Toxicol 2017; 99:103-118. [DOI: 10.1016/j.fct.2016.11.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 01/28/2023]
|
212
|
Kashyap T, Argueta C, Aboukameel A, Unger TJ, Klebanov B, Mohammad RM, Muqbil I, Azmi AS, Drolen C, Senapedis W, Lee M, Kauffman M, Shacham S, Landesman Y. Selinexor, a Selective Inhibitor of Nuclear Export (SINE) compound, acts through NF-κB deactivation and combines with proteasome inhibitors to synergistically induce tumor cell death. Oncotarget 2016; 7:78883-78895. [PMID: 27713151 PMCID: PMC5346685 DOI: 10.18632/oncotarget.12428] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/22/2016] [Indexed: 12/17/2022] Open
Abstract
The nuclear export protein, exportin-1 (XPO1/CRM1), is overexpressed in many cancers and correlates with poor prognosis. Selinexor, a first-in-class Selective Inhibitor of Nuclear Export (SINE) compound, binds covalently to XPO1 and blocks its function. Treatment of cancer cells with selinexor results in nuclear retention of major tumor suppressor proteins and cell cycle regulators, leading to growth arrest and apoptosis. Recently, we described the selection of SINE compound resistant cells and reported elevated expression of inflammation-related genes in these cells. Here, we demonstrated that NF-κB transcriptional activity is up-regulated in cells that are naturally resistant or have acquired resistance to SINE compounds. Resistance to SINE compounds was created by knockdown of the cellular NF-κB inhibitor, IκB-α. Combination treatment of selinexor with proteasome inhibitors decreased NF-κB activity, sensitized SINE compound resistant cells and showed synergistic cytotoxicity in vitro and in vivo. Furthermore, we showed that selinexor inhibited NF-κB activity by blocking phosphorylation of the IκB-α and the NF-κB p65 subunits, protecting IκB-α from proteasome degradation and trapping IκB-α in the nucleus to suppress NF-κB activity. Therefore, combination treatment of selinexor with a proteasome inhibitor may be beneficial to patients with resistance to either single-agent.
Collapse
Affiliation(s)
| | | | - Amro Aboukameel
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | - Ramzi M. Mohammad
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Irfana Muqbil
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Asfar S. Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Claire Drolen
- Karyopharm Therapeutics Inc., Newton, MA, 02459, USA
| | | | - Margaret Lee
- Karyopharm Therapeutics Inc., Newton, MA, 02459, USA
| | | | | | | |
Collapse
|
213
|
Lin B, Xu D, Leaman DW. X-linked inhibitor of apoptosis-associated factor 1 regulates TNF receptor 1 complex stability. FEBS Lett 2016; 590:4381-4392. [PMID: 27768232 DOI: 10.1002/1873-3468.12467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/17/2016] [Accepted: 08/30/2016] [Indexed: 11/06/2022]
Abstract
X-linked inhibitor of apoptosis (XIAP)-associated factor 1 (XAF1) is a cytokine-regulated, tumor necrosis factor (TNF) receptor-associated factor (TRAF) domain-containing protein that has a poorly defined cellular function. Here, we show that ectopically expressed XAF1 inhibits TNF-ɑ-induced NF-κB activation, whereas shRNA silencing of endogenous XAF1 augments it. Our data suggest that XAF1 may inhibit TNF-ɑ-induced NF-κB activation by disrupting the assembly of the TRADD/TRAF2/RIP1 complex (complex I) downstream of TNF receptor activation. XAF1 interacts with TRAF2 and inhibits TRAF2-dependent NF-κB activation, in part, by blocking TRAF2 polyubiquitination. Our findings also indicate that although XAF1 does not directly inhibit RIP1-dependent NF-κB activation, it binds RIP1 and disrupts RIP1/TRADD association. Our data suggest that XAF1 acts as a feedback regulator of the TNF receptor signaling pathway to suppress NF-κB activation.
Collapse
Affiliation(s)
- Boren Lin
- Department of Biological Sciences, The University of Toledo, OH, USA
| | - Da Xu
- Department of Biological Sciences, The University of Toledo, OH, USA
| | - Douglas W Leaman
- Department of Biological Sciences, The University of Toledo, OH, USA
| |
Collapse
|
214
|
Funakoshi-Tago M, Ohsawa K, Ishikawa T, Nakamura F, Ueda F, Narukawa Y, Kiuchi F, Tamura H, Tago K, Kasahara T. Inhibitory effects of flavonoids extracted from Nepalese propolis on the LPS signaling pathway. Int Immunopharmacol 2016; 40:550-560. [DOI: 10.1016/j.intimp.2016.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/11/2016] [Accepted: 10/11/2016] [Indexed: 01/27/2023]
|
215
|
Cnidilide, an alkylphthalide isolated from the roots of Cnidium officinale , suppresses LPS-induced NO, PGE 2 , IL-1β, IL-6 and TNF-α production by AP-1 and NF-κB inactivation in RAW 264.7 macrophages. Int Immunopharmacol 2016; 40:146-155. [DOI: 10.1016/j.intimp.2016.08.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/16/2016] [Accepted: 08/19/2016] [Indexed: 01/03/2023]
|
216
|
Corso-Díaz X, de Leeuw CN, Alonso V, Melchers D, Wong BKY, Houtman R, Simpson EM. Co-activator candidate interactions for orphan nuclear receptor NR2E1. BMC Genomics 2016; 17:832. [PMID: 27782803 PMCID: PMC5080790 DOI: 10.1186/s12864-016-3173-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 10/18/2016] [Indexed: 12/21/2022] Open
Abstract
Background NR2E1 (Tlx) is an orphan nuclear receptor that regulates the maintenance and self-renewal of neural stem cells, and promotes tumourigenesis. Nr2e1-null mice exhibit reduced cortical and limbic structures and pronounced retinal dystrophy. NR2E1 functions mainly as a repressor of gene transcription in association with the co-repressors atrophin-1, LSD1, HDAC and BCL11A. Recent evidence suggests that NR2E1 also acts as an activator of gene transcription. However, co-activator complexes that interact with NR2E1 have not yet been identified. In order to find potential novel co-regulators for NR2E1, we used a microarray assay for real-time analysis of co-regulator–nuclear receptor interaction (MARCoNI) that contains peptides representing interaction motifs from potential co-regulatory proteins, including known co-activator nuclear receptor box sequences (LxxLL motif). Results We found that NR2E1 binds strongly to an atrophin-1 peptide (Atro box) used as positive control and to 19 other peptides that constitute candidate NR2E1 partners. Two of these proteins, p300 and androgen receptor (AR), were further validated by reciprocal pull-down assays. The specificity of NR2E1 binding to peptides in the array was evaluated using two single amino acid variants, R274G and R276Q, which disrupted the majority of the binding interactions observed with wild-type NR2E1. The decreased binding affinity of these variants to co-regulators was further validated by pull-down assays using atrophin1 as bait. Despite the high conservation of arginine 274 in vertebrates, its reduced interactions with co-regulators were not significant in vivo as determined by retinal phenotype analysis in single-copy Nr2e1-null mice carrying the variant R274G. Conclusions We showed that MARCoNI is a specific assay to test interactions of NR2E1 with candidate co-regulators. In this way, we unveiled 19 potential co-regulator partners for NR2E1, including eight co-activators. All the candidates here identified need to be further validated using in vitro and in vivo models. This assay was sensitive to point mutations in NR2E1 ligand binding domain making it useful to identify mutations and/or small molecules that alter binding of NR2E1 to protein partners. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3173-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ximena Corso-Díaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
| | - Charles N de Leeuw
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Vivian Alonso
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | | | - Bibiana K Y Wong
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - René Houtman
- PamGene International B.V., Den Bosch, The Netherlands
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, BC, V6T 2A1, Canada. .,Department of Ophthalmology and Visual Science, University of British Columbia, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
217
|
Hedl M, Yan J, Abraham C. IRF5 and IRF5 Disease-Risk Variants Increase Glycolysis and Human M1 Macrophage Polarization by Regulating Proximal Signaling and Akt2 Activation. Cell Rep 2016; 16:2442-55. [PMID: 27545875 DOI: 10.1016/j.celrep.2016.07.060] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 04/21/2016] [Accepted: 07/22/2016] [Indexed: 12/19/2022] Open
Abstract
Interferon regulatory factor 5 (IRF5) regulates inflammatory M1 macrophage polarization, and disease-associated IRF5 genetic variants regulate pattern-recognition-receptor (PRR)-induced cytokines. PRR-stimulated macrophages and M1 macrophages exhibit enhanced glycolysis, a central mediator of inflammation. We find that IRF5 is needed for PRR-enhanced glycolysis in human macrophages and in mice in vivo. Upon stimulation of the PRR nucleotide binding oligomerization domain containing 2 (NOD2) in human macrophages, IRF5 binds RIP2, IRAK1, and TRAF6. IRF5, in turn, is required for optimal Akt2 activation, which increases expression of glycolytic pathway genes and HIF1A as well as pro-inflammatory cytokines and M1 polarization. Furthermore, pro-inflammatory cytokines and glycolytic pathways co-regulate each other. Rs2004640/rs2280714 TT/TT IRF5 disease-risk-carrier cells demonstrate increased IRF5 expression and increased PRR-induced Akt2 activation, glycolysis, pro-inflammatory cytokines, and M1 polarization relative to GG/CC carrier macrophages. Our findings identify that IRF5 disease-associated polymorphisms regulate diverse immunological and metabolic outcomes and provide further insight into mechanisms contributing to the increasingly recognized important role for glycolysis in inflammation.
Collapse
Affiliation(s)
- Matija Hedl
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Jie Yan
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Clara Abraham
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
218
|
Drube S, Weber F, Göpfert C, Loschinski R, Rothe M, Boelke F, Diamanti MA, Löhn T, Ruth J, Schütz D, Häfner N, Greten FR, Stumm R, Hartmann K, Krämer OH, Dudeck A, Kamradt T. TAK1 and IKK2, novel mediators of SCF-induced signaling and potential targets for c-Kit-driven diseases. Oncotarget 2016; 6:28833-50. [PMID: 26353931 PMCID: PMC4745695 DOI: 10.18632/oncotarget.5008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/20/2015] [Indexed: 12/25/2022] Open
Abstract
NF-κB activation depends on the IKK complex consisting of the catalytically active IKK1 and 2 subunits and the scaffold protein NEMO. Hitherto, IKK2 activation has always been associated with IκBα degradation, NF-κB activation, and cytokine production. In contrast, we found that in SCF-stimulated primary bone marrow-derived mast cells (BMMCs), IKK2 is alternatively activated. Mechanistically, activated TAK1 mediates the association between c-Kit and IKK2 and therefore facilitates the Lyn-dependent IKK2 activation which suffices to mediate mitogenic signaling but, surprisingly, does not result in NF-κB activation. Moreover, the c-Kit-mediated and Lyn-dependent IKK2 activation is targeted by MyD88-dependent pathways leading to enhanced IKK2 activation and therefore to potentiated effector functions. In neoplastic cells, expressing constitutively active c-Kit mutants, activated TAK1 and IKKs do also not induce NF-κB activation but mediate uncontrolled proliferation, resistance to apoptosis and enables IL-33 to mediate c-Kit-dependent signaling. Together, we identified the formation of the c-Kit-Lyn-TAK1 signalosome which mediates IKK2 activation. Unexpectedly, this IKK activation is uncoupled from the NF-κB-machinery but is critical to modulate functional cell responses in primary-, and mediates uncontrolled proliferation and survival of tumor-mast cells. Therefore, targeting TAK1 and IKKs might be a novel approach to treat c-Kit-driven diseases.
Collapse
Affiliation(s)
- Sebastian Drube
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Franziska Weber
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | | | - Romy Loschinski
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Mandy Rothe
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Franziska Boelke
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Michaela A Diamanti
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, Frankfurt, Germany
| | - Tobias Löhn
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Julia Ruth
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| | - Dagmar Schütz
- Institut für Pharmakologie, Universitätsklinikum Jena, Jena, Germany
| | - Norman Häfner
- Gynäkologische Molekularbiologie, Klinik für Frauenheilkunde und Geburtshilfe, Jena, Germany
| | - Florian R Greten
- Georg-Speyer-Haus, Institute for Tumorbiology and Experimental Therapy, Frankfurt, Germany
| | - Ralf Stumm
- Institut für Pharmakologie, Universitätsklinikum Jena, Jena, Germany
| | - Karin Hartmann
- Klinik und Poliklinik für Dermatologie und Venerologie, Universität zu Köln, Köln, Germany
| | - Oliver H Krämer
- Institut für Toxikologie, Universitätsmedizin Mainz, Mainz, Germany
| | - Anne Dudeck
- Institute for Immunology, Technische Universität Dresden, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Thomas Kamradt
- Institut für Immunologie, Universitätsklinikum Jena, Jena, Germany
| |
Collapse
|
219
|
Park HS, Vick EJ, Gao Y, He C, Almosnid NM, Farone M, Farone AL. Cis- and Trans-gnetin H from Paeonia suffruticosa suppress inhibitor kappa B kinase phosphorylation in LPS-stimulated human THP-1 cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 189:202-209. [PMID: 27196294 DOI: 10.1016/j.jep.2016.05.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/12/2016] [Accepted: 05/16/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The inflammatory response is an important mechanism in host defense; however, overstimulation and chronic inflammation are involved in many important human diseases. Currently, tumor necrosis factor-alpha blockers such as infliximab and adalimumab along with methotrexate are used in cases of severe and chronic disease. However, there are severe side effects and limitations associated with these treatments. Cis- and trans-gnetin H are compounds isolated from the seeds of Paeonia suffruticosa, a medicinal plant used in traditional Chinese medicine for the treatment of many conditions, including inflammatory diseases. In this study, we investigated possible anti-inflammatory mechanisms of cis- and trans-gnetin H against LPS-stimulated human THP-1 cells. MATERIAL AND METHODS PMA-differentiated THP-1 cells were pretreated with increasing concentrations of cis- and trans-gnetin H with or without LPS. Following treatment, cytotoxicity and the TNF-α, IL-1β, and IL-8 response were measured. We also characterized the nuclear translocation of NF-κB subunit p65 (RelA) by immunofluorescence and then investigated NF-κB activation by measuring the phosphorylation of NF-κB mediators, IKK-β, IκB α, and p65 by western blotting. RESULTS We found that cis- and trans-gnetin H significantly inhibited the cytokine response in a concentration-dependent manner without affecting cell viability. Cis- and trans-gnetin H effectively inhibited nuclear translocation of p65 and phosphorylation of IKK-β, IκB α, and p65. While both compounds showed promising anti-inflammatory effects, trans-gnetin H was determined to be more effective in suppressing cytokine responses. CONCLUSION We demonstrated that cis- and trans-gnetin H suppress cytokine response in LPS-stimulated THP-1 cells by preventing activation of key signaling molecules, IKK-β, IκB α, and p65, involved in the NF-κB pathway and suggest the use of cis- and trans-gnetin H in potential therapies for conditions and diseases associated with chronic inflammation.
Collapse
Affiliation(s)
- Hyo S Park
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| | - Eric J Vick
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| | - Ying Gao
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| | - Chunnian He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, No. 151 Malianwa, North Road, Haidian District, Beijing 100193, PR China.
| | - Nadin Marwan Almosnid
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| | - Mary Farone
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| | - Anthony L Farone
- Middle Tennessee State University, 1301 East Main Street, Murfreesboro, Tennessee 37132, USA.
| |
Collapse
|
220
|
Müller-Edenborn K, Léger K, Glaus Garzon JF, Oertli C, Mirsaidi A, Richards PJ, Rehrauer H, Spielmann P, Hoogewijs D, Borsig L, Hottiger MO, Wenger RH. Hypoxia attenuates the proinflammatory response in colon cancer cells by regulating IκB. Oncotarget 2016; 6:20288-301. [PMID: 25978030 PMCID: PMC4653005 DOI: 10.18632/oncotarget.3961] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 04/10/2015] [Indexed: 12/12/2022] Open
Abstract
Two main features common to all solid tumors are tissue hypoxia and inflammation, both of which cause tumor progression, metastasis, therapy resistance and increased mortality. Chronic inflammation is associated with increased cancer risk, as demonstrated for inflammatory bowel disease patients developing colon cancer. However, the interplay between hypoxia and inflammation on the molecular level remains to be elucidated. We found that MC-38 mouse colon cancer cells contain functional hypoxic (HIF-1α) and inflammatory (p65/RelA) signaling pathways. In contrast to cells of the myeloid lineage, HIF-1α levels remained unaffected in MC-38 cells treated with LPS, and hypoxia failed to induce NF-κB. A similar regulation of canonical HIF and NF-κB target genes confirmed these results. RNA deep sequencing of HIF-1α and p65/RelA knock-down cells revealed that a surprisingly large fraction of HIF target genes required p65/RelA for hypoxic regulation and a number of p65/RelA target genes required HIF-1α for proinflammatory regulation, respectively. Hypoxia attenuated the inflammatory response to LPS by inhibiting nuclear translocation of p65/RelA independently of HIF-1α, which was associated with enhanced IκBα levels and decreased IKKβ phosphorylation. These data demonstrate that the interaction between hypoxic and inflammatory signaling pathways needs to be considered when designing cancer therapies targeting HIF or NF-κB.
Collapse
Affiliation(s)
- Kamila Müller-Edenborn
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Karolin Léger
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| | - Jesus F Glaus Garzon
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Carole Oertli
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Ali Mirsaidi
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| | - Peter J Richards
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center, University of Zurich, Zurich, Switzerland
| | - Patrick Spielmann
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - David Hoogewijs
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Michael O Hottiger
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
221
|
Liu Q, Zhang Z, Zheng Z, Zheng C, Liu Y, Hu Q, Ke X, Wang H. Human Bocavirus NS1 and NS1-70 Proteins Inhibit TNF-α-Mediated Activation of NF-κB by targeting p65. Sci Rep 2016; 6:28481. [PMID: 27329558 PMCID: PMC4916443 DOI: 10.1038/srep28481] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/25/2022] Open
Abstract
Human bocavirus (HBoV), a parvovirus, is a single-stranded DNA etiologic agent causing lower respiratory tract infections in young children worldwide. Nuclear factor kappa B (NF-κB) transcription factors play crucial roles in clearance of invading viruses through activation of many physiological processes. Previous investigation showed that HBoV infection could significantly upregulate the level of TNF-α which is a strong NF-κB stimulator. Here we investigated whether HBoV proteins modulate TNF-α–mediated activation of the NF-κB signaling pathway. We showed that HBoV NS1 and NS1-70 proteins blocked NF-κB activation in response to TNF-α. Overexpression of TNF receptor-associated factor 2 (TRAF2)-, IκB kinase alpha (IKKα)-, IκB kinase beta (IKKβ)-, constitutively active mutant of IKKβ (IKKβ SS/EE)-, or p65-induced NF-κB activation was inhibited by NS1 and NS1-70. Furthermore, NS1 and NS1-70 didn’t interfere with TNF-α-mediated IκBα phosphorylation and degradation, nor p65 nuclear translocation. Coimmunoprecipitation assays confirmed the interaction of both NS1 and NS1-70 with p65. Of note, NS1 but not NS1-70 inhibited TNF-α-mediated p65 phosphorylation at ser536. Our findings together indicate that HBoV NS1 and NS1-70 inhibit NF-κB activation. This is the first time that HBoV has been shown to inhibit NF-κB activation, revealing a potential immune-evasion mechanism that is likely important for HBoV pathogenesis.
Collapse
Affiliation(s)
- Qingshi Liu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenfeng Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zhenhua Zheng
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Caishang Zheng
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yan Liu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xianliang Ke
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hanzhong Wang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
222
|
Wang L, Zhu S, Xu G, Feng J, Han T, Zhao F, She YL, Liu S, Ye L, Zhu Y. Gene Expression and Antiviral Activity of Interleukin-35 in Response to Influenza A Virus Infection. J Biol Chem 2016; 291:16863-76. [PMID: 27307042 DOI: 10.1074/jbc.m115.693101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Indexed: 01/22/2023] Open
Abstract
Interleukin-35 (IL-35) is a newly described member of the IL-12 family. It has been reported to inhibit inflammation and autoimmune inflammatory disease and can increase apoptotic sensitivity. Little is known about the role of IL-35 during viral infection. Herein, high levels of IL-35 were found in peripheral blood mononuclear cells and throat swabs from patients with seasonal influenza A virus (IAV) relative to healthy individuals. IAV infection of human lung epithelial and primary cells increased levels of IL-35 mRNA and protein. Further studies demonstrated that IAV-induced IL-35 transcription is regulated by NF-κB. IL-35 expression was significantly suppressed by selective inhibitors of cyclooxygenase-2 (COX-2) and inducible nitric-oxide synthase, indicating their involvement in IL-35 expression. Interestingly, IL-35 production may have suppressed IAV RNA replication and viral protein synthesis via induction of type I and III interferons (IFN), leading to activation of downstream IFN effectors, including double-stranded RNA-dependent protein kinase, 2',5'-oligoadenylate synthetase, and myxovirus resistance protein. IL-35 exhibited extensive antiviral activity against the hepatitis B virus, enterovirus 71, and vesicular stomatitis virus. Our results demonstrate that IL-35 is a novel IAV-inducible cytokine, and its production elicits antiviral activity.
Collapse
Affiliation(s)
- Li Wang
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shengli Zhu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Gang Xu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Jian Feng
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Tao Han
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Fanpeng Zhao
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying-Long She
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shi Liu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Linbai Ye
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Ying Zhu
- From the The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
223
|
Pierce EN, Piyankarage SC, Dunlap T, Litosh V, Siklos MI, Wang YT, Thatcher GRJ. Prodrugs Bioactivated to Quinones Target NF-κB and Multiple Protein Networks: Identification of the Quinonome. Chem Res Toxicol 2016; 29:1151-9. [PMID: 27258437 DOI: 10.1021/acs.chemrestox.6b00115] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Electrophilic reactive intermediates resulting from drug metabolism have been associated with toxicity and off-target effects and in some drug discovery programs trigger NO-GO decisions. Many botanicals and dietary supplements are replete with such reactive electrophiles, notably Michael acceptors, which have been demonstrated to elicit chemopreventive mechanisms; and Michael acceptors are gaining regulatory approval as contemporary cancer therapeutics. Identifying protein targets of these electrophiles is central to understanding potential therapeutic benefit and toxicity risk. NO-donating NSAID prodrugs (NO-NSAIDs) have been the focus of extensive clinical and preclinical studies in inflammation and cancer chemoprevention and therapy: a subset exemplified by pNO-ASA, induces chemopreventive mechanisms following bioactivation to an electrophilic quinone methide (QM) Michael acceptor. Having previously shown that these NO-independent, QM-donors activated Nrf2 via covalent modification of Keap-1, we demonstrate that components of canonical NF-κB signaling are also targets, leading to the inhibition of NF-κB signaling. Combining bio-orthogonal probes of QM-donor ASA prodrugs with mass spectrometric proteomics and pathway analysis, we proceeded to characterize the quinonome: the protein cellular targets of QM-modification by pNO-ASA and its ASA pro-drug congeners. Further comparison was made using a biorthogonal probe of the "bare-bones", Michael acceptor, and clinical anti-inflammatory agent, dimethyl fumarate, which we have shown to inhibit NF-κB signaling. Identified quinonome pathways include post-translational protein folding, cell-death regulation, protein transport, and glycolysis; and identified proteins included multiple heat shock elements, the latter functionally confirmed by demonstrating activation of heat shock response.
Collapse
Affiliation(s)
- Emily N Pierce
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Sujeewa C Piyankarage
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Tareisha Dunlap
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Vladislav Litosh
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Marton I Siklos
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Yue-Ting Wang
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| |
Collapse
|
224
|
The Ubiquitination of NF-κB Subunits in the Control of Transcription. Cells 2016; 5:cells5020023. [PMID: 27187478 PMCID: PMC4931672 DOI: 10.3390/cells5020023] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/03/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023] Open
Abstract
Nuclear factor (NF)-κB has evolved as a latent, inducible family of transcription factors fundamental in the control of the inflammatory response. The transcription of hundreds of genes involved in inflammation and immune homeostasis require NF-κB, necessitating the need for its strict control. The inducible ubiquitination and proteasomal degradation of the cytoplasmic inhibitor of κB (IκB) proteins promotes the nuclear translocation and transcriptional activity of NF-κB. More recently, an additional role for ubiquitination in the regulation of NF-κB activity has been identified. In this case, the ubiquitination and degradation of the NF-κB subunits themselves plays a critical role in the termination of NF-κB activity and the associated transcriptional response. While there is still much to discover, a number of NF-κB ubiquitin ligases and deubiquitinases have now been identified which coordinate to regulate the NF-κB transcriptional response. This review will focus the regulation of NF-κB subunits by ubiquitination, the key regulatory components and their impact on NF-κB directed transcription.
Collapse
|
225
|
Zhang L, Zhang J, Chen Z, Wang L, Wu X, Ou M. Epidermal growth factor (EGF) triggers the malignancy of hemangioma cells via activation of NF-κB signals. Biomed Pharmacother 2016; 82:133-40. [PMID: 27470348 DOI: 10.1016/j.biopha.2016.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 04/30/2016] [Accepted: 05/02/2016] [Indexed: 11/16/2022] Open
Abstract
Hemangioma (HA) is tumor formed by hyper-proliferation of vascular endothelial cells. However, the role and mechanisms of epidermal growth factor (EGF) on the progression of HA are not well illustrated. Our present study revealed that EGF can significantly promote the in vitro proliferation and motility of HA cells, which was confirmed by the up regulation of Bcl-2, proliferating cell nuclear antigen (PCNA), and metalloproteinase-2 (MMP-2) and MMP-9. The pharmacological inhibition of NF-κB, while not ERK1/2 or PI3K/Akt, attenuated EGF induced cell proliferation and expression of MMP-2 and MMP-9. EGF treatment also increased the phosphorylation, nuclear translocation and transcriptional activities of NF-κB in HA cells. These data suggested that NF-κB plays an essential role in EGF induced malignancy of HA cells. Furthermore, EGF treatment also increased the phosphorylation of IκB and IKKα, while not IKKβ or IKKγ. The knockdown of IKKα reversed EGF induced activation of NF-κB. EGF treatment also decreased the phosphorylation of GSK-3β and increased its activities in both HDEC and CRL-2586 EOMA cells. LiCl, a potent GSK-3β inhibitor, can obviously reverse EGF induced up regulation of p65 phosphorylation. Collectively, our study revealed that EGF can trigger the malignancy of HA cells via induction of proliferation and invasion. The activation of NF-κB through IKKα/IκBα and GSK-3β signal is essential for this process. It suggested that EGF/NF-κB signal may represent a novel therapeutic target for the treatment of human HA.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Emergency, Peolpe's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, China
| | - Jingming Zhang
- Department of Cardiology, The People's Hospital of Zhongwei City of Ningxia Hui Autonomous Region, Zhongwei 755000, China
| | - Zhanlong Chen
- Department of Emergency, Peolpe's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, China
| | - Liqin Wang
- Department of Vascular Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, China
| | - Xiaomin Wu
- Department of Vascular Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, China
| | - Minghui Ou
- Department of Vascular Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750002, China.
| |
Collapse
|
226
|
Choi SI, Lee SY, Jung WJ, Lee SH, Lee EJ, Min KH, Hur GY, Lee SH, Lee SY, Kim JH, Shin C, Shim JJ, In KH, Kang KH, Lee MG. The effect of an IκB-kinase-β(IKKβ) inhibitor on tobacco smoke-induced pulmonary inflammation. Exp Lung Res 2016; 42:182-9. [PMID: 27144414 DOI: 10.1080/01902148.2016.1174749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF THE STUDY Inactivation of NF-κB with IKKβ knockout mice reduces tobacco smoke-induced pulmonary inflammation. In this study, we investigated whether the IKKβ inhibitor PS-1145 could attenuate the pulmonary inflammation induced by tobacco smoke. MATERIALS AND METHODS We divided 30 mice into three groups: a control group, a smoking group, and a PS-1145 group. Mice from the smoking and PS-1145 groups were exposed for 2 weeks to tobacco smoke. PS-1145 was injected intraperitoneally before every tobacco smoke exposure. After 2 weeks, bronchoalveolar lavage (BAL) was performed for cell counting and measuring of inflammatory chemokines. We analyzed the correlation between NF-κB and NF-κB-regulated chemokines in BAL fluid and measured the neutrophils and macrophages by immunostaining in lung tissues. RESULTS The PS-1145 group showed a significant reduction in the number of total cells, neutrophils, and macrophages, as well as the KC and MCP-1 level, in the BAL fluid compared to the smoking group. There was no significant difference in the level of MIP-1α. The level of NF-κB in BAL fluid was significantly positively correlated with KC and MCP-1 levels, but not with MIP-1α level. The PS-1145 group also showed a significant fewer neutrophils and macrophages in the lung tissue. CONCLUSIONS We conclude that the IKKβ inhibitor PS-1145 suppressed the NF-κB signaling pathway and reduced the recruitment of inflammatory cells and chemokines in pulmonary inflammation induced by tobacco smoke. IKKβ inhibition offers a potential therapeutic target for tobacco smoke-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Sue In Choi
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Sang Yeub Lee
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Won Jai Jung
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Seung Hyeun Lee
- b Division of Pulmonary and Critical Care Medicine , Department of Internal Medicine, Kyung Hee University School of Medicine , Seoul , Korea
| | - Eun Joo Lee
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Kyung Hoon Min
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Gyu Young Hur
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Seung Heon Lee
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Sung Yong Lee
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Je Hyeong Kim
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Chol Shin
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Jae Jeong Shim
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Kwang Ho In
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Kyung Ho Kang
- a Division of Respiratory and Critical Care Medicine , Department of Internal Medicine, College of Medicine, Korea University , Seoul , Korea
| | - Min-Goo Lee
- c Department of Physiology , College of Medicine, Korea University , Seoul , Korea
| |
Collapse
|
227
|
Al-Sadi R, Guo S, Ye D, Rawat M, Ma TY. TNF-α Modulation of Intestinal Tight Junction Permeability Is Mediated by NIK/IKK-α Axis Activation of the Canonical NF-κB Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1151-1165. [PMID: 26948423 PMCID: PMC4861759 DOI: 10.1016/j.ajpath.2015.12.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022]
Abstract
Tumor necrosis factor (TNF)-α, a key mediator of intestinal inflammation, causes an increase in intestinal epithelial tight junction (TJ) permeability by activating myosin light chain kinase (MLCK; official name MYLK3) gene. However, the precise signaling cascades that mediate the TNF-α-induced activation of MLCK gene and increase in TJ permeability remain unclear. Our aims were to delineate the upstream signaling mechanisms that regulate the TNF-α modulation of intestinal TJ barrier function with the use of in vitro and in vivo intestinal epithelial model systems. TNF-α caused a rapid activation of both canonical and noncanonical NF-κB pathway. NF-κB-inducing kinase (NIK) and mitogen-activated protein kinase kinase-1 (MEKK-1) were activated in response to TNF-α. NIK mediated the TNF-α activation of inhibitory κB kinase (IKK)-α, and MEKK1 mediated the activation of IKK complex, including IKK-β. NIK/IKK-α axis regulated the activation of both NF-κB p50/p65 and RelB/p52 pathways. Surprisingly, the siRNA induced knockdown of NIK, but not MEKK-1, prevented the TNF-α activation of both NF-κB p50/p65 and RelB/p52 and the increase in intestinal TJ permeability. Moreover, NIK/IKK-α/NF-κB p50/p65 axis mediated the TNF-α-induced MLCK gene activation and the subsequent MLCK increase in intestinal TJ permeability. In conclusion, our data show that NIK/IKK-α/regulates the activation of NF-κB p50/p65 and plays an integral role in the TNF-α-induced activation of MLCK gene and increase in intestinal TJ permeability.
Collapse
Affiliation(s)
- Rana Al-Sadi
- Department of Internal Medicine, University of New Mexico School of Medicine and Albuquerque Veterans Affairs Medical Center, Albuquerque, New Mexico
| | - Shuhong Guo
- Department of Internal Medicine, University of New Mexico School of Medicine and Albuquerque Veterans Affairs Medical Center, Albuquerque, New Mexico
| | - Dongmei Ye
- Department of Internal Medicine, University of New Mexico School of Medicine and Albuquerque Veterans Affairs Medical Center, Albuquerque, New Mexico
| | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine and Albuquerque Veterans Affairs Medical Center, Albuquerque, New Mexico
| | - Thomas Y Ma
- Department of Internal Medicine, University of New Mexico School of Medicine and Albuquerque Veterans Affairs Medical Center, Albuquerque, New Mexico.
| |
Collapse
|
228
|
Kafle B, Khadka D, Park BH, Kim CK, Cho H. Antiobesity Effect of SA37 in High-Fat Diet-induced Obese C57BL/6J Mice. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Bhooshan Kafle
- Department of Chemistry, College of Natural Science; Inha University; Incheon 22212 Korea
| | - Deegendra Khadka
- Department of Chemistry, College of Natural Science; Inha University; Incheon 22212 Korea
| | - Byung Ho Park
- Department of Chemistry, College of Natural Science; Inha University; Incheon 22212 Korea
| | - Chan Kyung Kim
- Department of Chemistry, College of Natural Science; Inha University; Incheon 22212 Korea
| | - Hyeongjin Cho
- Department of Chemistry, College of Natural Science; Inha University; Incheon 22212 Korea
| |
Collapse
|
229
|
Zhu G, Huang Y, Wu C, Wei D, Shi Y. Activation of G-Protein-Coupled Estrogen Receptor Inhibits the Migration of Human Nonsmall Cell Lung Cancer Cells via IKK-β/NF-κB Signals. DNA Cell Biol 2016; 35:434-42. [PMID: 27082459 DOI: 10.1089/dna.2016.3235] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Estrogen signals have been suggested to modulate the progression and metastasis of nonsmall cell lung cancer (NSCLC), which is one of the leading causes of cancer deaths worldwide. While there are limited data concerning the roles and effects of G-protein-coupled estrogen receptor (GPER) on the progression of NSCLC, our present study reveals that the expression of GPER in NSCLC cells is obviously greater than that in lung fibroblast cell line MRC-5. Activation of GPER via its specific agonist G-1 decreases the in vitro motility of A549 and H358 cells and the expression of matrix metalloproteinase 2 (MMP-2) and MMP-9. Further, G-1 treatment can rapidly decrease the phosphorylation, nuclear translocation, and promoter activities of NF-κB in NSCLC cells. BAY 11-7082, the inhibitor of NF-κB, also inhibits the expression of MMP-2/9, while overexpression of p65 significantly attenuates G-1-induced downregulation of MMP-2/9. It suggests that inhibition of NF-κB mediates G-1-induced MMP-2/9 downregulation. G-1 treatment significantly down regulates the phosphorylation of IκB kinase β (IKK-β) and IκBα, while not IKK-α, in both 549 and H358 cells. ACHP, the specific inhibitor of IKK-β, can reinforce G-1-induced MMP-2/9 downregulation and invasion suppression of A549 cells. Collectively, our results suggest that activation of GPER can inhibit the migration of human NSCLC cells via suppression of IKK-β/NF-κB signals. These findings will help to better understand the roles and mechanisms of GPER as a potential therapy target for NSCLC patients.
Collapse
Affiliation(s)
- Guangfa Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University , Beijing, People's Republic of China
| | - Yan Huang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University , Beijing, People's Republic of China
| | - Chunting Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University , Beijing, People's Republic of China
| | - Dong Wei
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University , Beijing, People's Republic of China
| | - Yingxin Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University , Beijing, People's Republic of China
| |
Collapse
|
230
|
Latanoprost-induced Cytokine and Chemokine Release From Human Tenon's Capsule Fibroblasts: Role of MAPK and NF-κB Signaling Pathways. J Glaucoma 2016; 24:635-41. [PMID: 25715004 DOI: 10.1097/ijg.0000000000000140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE Long-term topical antiglaucoma therapy is considered a significant risk factor for failure of trabeculectomy. We investigated the effects of antiglaucoma drugs on proinflammatory cytokine and chemokine release from cultured human Tenon's capsule fibroblasts (HTFs) as well as the signaling pathways that underlie such effects. MATERIALS AND METHODS Release of the proinflammatory cytokine interleukin (IL)-6 and the chemokines IL-8 and monocyte chemotactic protein (MCP)-1 was measured with enzyme-linked immunosorbent assays. The phosphorylation (activation) of mitogen-activated protein kinases (MAPKs) as well as the phosphorylation and degradation of the nuclear factor-κB (NF-κB) inhibitor IκB-α were assessed by immunoblot analysis. RESULTS Latanoprost stimulated the release of IL-6, IL-8, and MCP-1 from HTFs in a concentration-dependent and time-dependent manner, whereas timolol maleate and pilocarpine had no such effects. Latanoprost also activated the MAPKs extracellular signal-regulated kinase, p38, and c-Jun NH2-terminal kinase as well as induced the phosphorylation and degradation of IκB-α in these cells. The latanoprost-induced release of IL-6, IL-8, and MCP-1 was attenuated by inhibitors of MAPK (PD98059, SB203580, or JNK inhibitor II) or NF-κB (IκB kinase 2 inhibitor) signaling pathways. CONCLUSIONS Latanoprost induced proinflammatory cytokine and chemokine release from HTFs in a manner dependent on MAPK and NF-κB signaling. These effects of latanoprost might influence bleb scarring after filtration surgery.
Collapse
|
231
|
Wakita M, Edamatsu H, Li M, Emi A, Kitazawa S, Kataoka T. Phospholipase Cϵ Activates Nuclear Factor-κB Signaling by Causing Cytoplasmic Localization of Ribosomal S6 Kinase and Facilitating Its Phosphorylation of Inhibitor κB in Colon Epithelial Cells. J Biol Chem 2016; 291:12586-12600. [PMID: 27053111 DOI: 10.1074/jbc.m116.717561] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Indexed: 12/30/2022] Open
Abstract
Phospholipase Cϵ (PLCϵ), an effector of Ras and Rap small GTPases, plays a crucial role in inflammation by augmenting proinflammatory cytokine expression. This proinflammatory function of PLCϵ is implicated in its facilitative role in tumor promotion and progression during skin and colorectal carcinogenesis, although their direct link remains to be established. Moreover, the molecular mechanism underlying these functions of PLCϵ remains unknown except that PKD works downstream of PLCϵ. Here we show by employing the colitis-induced colorectal carcinogenesis model, where Apc(Min) (/+) mice are administered with dextran sulfate sodium, that PLCϵ knock-out alleviates the colitis and suppresses the following tumorigenesis concomitant with marked attenuation of proinflammatory cytokine expression. In human colon epithelial Caco2 cells, TNF-α induces sustained expression of proinflammatory molecules and sustained activation of nuclear factor-κB (NF-κB) and PKD, the late phases of which are suppressed by not only siRNA-mediated PLCϵ knockdown but also treatment with a lysophosphatidic acid (LPA) receptor antagonist. Also, LPA stimulation induces these events in an early time course, suggesting that LPA mediates TNF-α signaling in an autocrine manner. Moreover, PLCϵ knockdown results in inhibition of phosphorylation of IκB by ribosomal S6 kinase (RSK) but not by IκB kinases. Subcellular fractionation suggests that enhanced phosphorylation of a scaffolding protein, PEA15 (phosphoprotein enriched in astrocytes 15), downstream of the PLCϵ-PKD axis causes sustained cytoplasmic localization of phosphorylated RSK, thereby facilitating IκB phosphorylation in the cytoplasm. These results suggest the crucial role of the TNF-α-LPA-LPA receptor-PLCϵ-PKD-PEA15-RSK-IκB-NF-κB pathway in facilitating inflammation and inflammation-associated carcinogenesis in the colon.
Collapse
Affiliation(s)
- Masahiro Wakita
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hironori Edamatsu
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Mingzhen Li
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Aki Emi
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Sohei Kitazawa
- Department of Molecular Pathology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Tohru Kataoka
- Division of Molecular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| |
Collapse
|
232
|
Nowarski R, Jackson R, Gagliani N, de Zoete MR, Palm NW, Bailis W, Low JS, Harman CCD, Graham M, Elinav E, Flavell RA. Epithelial IL-18 Equilibrium Controls Barrier Function in Colitis. Cell 2016; 163:1444-56. [PMID: 26638073 DOI: 10.1016/j.cell.2015.10.072] [Citation(s) in RCA: 427] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/03/2015] [Accepted: 10/23/2015] [Indexed: 12/17/2022]
Abstract
The intestinal mucosal barrier controlling the resident microbiome is dependent on a protective mucus layer generated by goblet cells, impairment of which is a hallmark of the inflammatory bowel disease, ulcerative colitis. Here, we show that IL-18 is critical in driving the pathologic breakdown of barrier integrity in a model of colitis. Deletion of Il18 or its receptor Il18r1 in intestinal epithelial cells (Δ/EC) conferred protection from colitis and mucosal damage in mice. In contrast, deletion of the IL-18 negative regulator Il18bp resulted in severe colitis associated with loss of mature goblet cells. Colitis and goblet cell loss were rescued in Il18bp(-/-);Il18r(Δ/EC) mice, demonstrating that colitis severity is controlled at the level of IL-18 signaling in intestinal epithelial cells. IL-18 inhibited goblet cell maturation by regulating the transcriptional program instructing goblet cell development. These results inform on the mechanism of goblet cell dysfunction that underlies the pathology of ulcerative colitis.
Collapse
Affiliation(s)
- Roni Nowarski
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruaidhrí Jackson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Nicola Gagliani
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marcel R de Zoete
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Will Bailis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jun Siong Low
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Christian C D Harman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Morven Graham
- Center for Cellular and Molecular Imaging, Yale School of Medicine, New Haven, CT 06520, USA
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
233
|
Inhibitory effect of baicalin on allergic response in ovalbumin-induced allergic rhinitis guinea pigs and lipopolysaccharide-stimulated human mast cells. Inflamm Res 2016; 65:603-12. [PMID: 27043920 DOI: 10.1007/s00011-016-0943-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/23/2016] [Accepted: 03/25/2016] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Baicalin, a flavonoid compound purified from the dry roots of Scutellaria baicalensis Georgi, has generally been used for the treatment of various allergic diseases. However, there is little information about the anti-inflammatory effects of baicalin for allergic rhinitis. This study aims to investigate the anti-allergic effect of baicalin on allergic response in ovalbumin (OVA)-induced allergic rhinitis guinea pigs and lipopolysaccharide (LPS)-stimulated human mast cells. METHODS Using in vivo models, we evaluated the effect of baicalin on allergic rhinitis symptoms via recording the number of nasal rubs and sneezes. The levels of histamine, OVA-specific immunoglobulin E(IgE), eosinophil cationic protein (ECP) and inflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA). The histological changes of nasal mucosa were observed by light microscope after HE staining. In vitro, the release of histamine and β-hexosaminidase of compound 48/80-induced human mast cells were measured by ELISA and PNP-NAG colorimetry, respectively. The productions of inflammatory cytokines of LPS-stimulated human mast cells were determined using ELISA. Western blot was used to test the protein expression of JAK2, p-JAK2, STAT5, p-STAT5, IKKβ, p-IKKβ, IκBα, p-IκBα and NF-κB (p65) of LPS-stimulated human mast cells. RESULTS The oral administration of baicalin at doses of 50 and 200 mg/kg improved allergic rhinitis symptoms and the histological changes of nasal mucosa and decreased the serum levels of histamine, ECP, interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor (TNF)-α and OVA-specific IgE in OVA-induced allergic rhinitis guinea pigs. In vitro, baicalin suppressed the release of histamine and β-hexosaminidase in compound 48/80-induced human mast cells. In addition, baicalin also inhibited the productions of inflammatory cytokines such as IL-1β, IL-6, IL-8 and TNF-α and suppressed the phosphorylation of JAK2, STAT5, IKKβ, IκBα and the nuclear translocation of NF-κB (p65) subunit in LPS-stimulated human mast cells. CONCLUSIONS These results suggest that baicalin can effectively prevent allergic response in OVA-induced allergic rhinitis guinea pigs and inhibit inflammatory response via blocking JAK2-STAT5 and NF-κB signaling pathways in LPS-stimulated human mast cells. Considered together,the results show that baicalin may be a useful drug in the treatment of allergic rhinitis.
Collapse
|
234
|
Chen J, Zhang J, Cao J, Xia Z, Gan J. Inflammatory MAPK and NF-κB signaling pathways differentiated hepatitis potential of two agglomerated titanium dioxide particles. JOURNAL OF HAZARDOUS MATERIALS 2016; 304:370-378. [PMID: 26590873 DOI: 10.1016/j.jhazmat.2015.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/30/2015] [Accepted: 11/02/2015] [Indexed: 06/05/2023]
Abstract
TiO2 nanoparticles (TiO2NPs) consumption and deposit in liver have possible implications for hepatitis risks. Specific signal dysregulation at early inflammatory responses needs to be characterized in TiO2NP-induced hepatopathy. MAPK and NF-κB signaling pathways are known to participate in inflammation and respond sensitively to chemical agents, making them preferable biomarkers for hepatitis. In the present study, dynamic activation of MAPK and NF-κB pathways were explored by immunoblotting and NF-κB luciferase reporter assay depending on characterization of TiO2NP agglomeration in human HepG2 cells. Inflammatory and cytotoxic potential of TiO2NPs were determined by qRT-PCR and WST-1 assay. AFM and TEM analyses uncovered ultrastructure damages underlying hepatotoxicity of TiO2NPs. Rod-like rutile agglomerated smaller and induced more pronounced cytotoxicity and immunogenicity than anatase. Correspondingly, though both rutile and anatase significantly activated p38, ERK1/2 and NF-κB pathways, rutile accelerated the maximum phosphorylation of ERK1/2 and elevated the potency of IκBα phosphorylation to its bell curve shape in comparison with a lower and sigmoid type of IκBα phosphorylation for anatase. Furthermore, cell elasticity indicated by Young's modulus and adhesion force increased accompanied with mitochondria damage, contributing to signal dysregulation and hepatotoxicity. The results suggested that differential activation of MAPK and NF-κB pathways could be early predictors for distinct hepatitis risks of two agglomerated TiO2NPs.
Collapse
Affiliation(s)
- Jin Chen
- Environmental Science Institute, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianying Zhang
- Environmental Science Institute, Zhejiang University, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang, China.
| | - Junmei Cao
- Environmental Science Institute, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Zongping Xia
- Life Sciences Institute, Zhejiang University, Hangzhou, China.
| | - Jay Gan
- Department of Environmental Sciences, University of California, Riverside, CA, USA.
| |
Collapse
|
235
|
Ichikawa K, Ohshima D, Sagara H. Regulation of signal transduction by spatial parameters: a case in NF-κB oscillation. IET Syst Biol 2016; 9:41-51. [PMID: 26672147 DOI: 10.1049/iet-syb.2013.0020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
NF-κB is a transcription factor regulating expression of more than 500 genes, and its dysfunction leads to the autoimmune and inflammatory diseases. In malignant cancer cells, NF-κB is constitutively activated. Thus the elucidation of mechanisms for NF-κB regulation is important for the establishment of therapeutic treatment caused by incorrect NF-κB responses. Cytoplasmic NF-κB translocates to the nucleus by the application of extracellular stimuli such as cytokines. Nuclear NF-κB is known to oscillate with the cycle of 1.5-4.5 h, and it is thought that the oscillation pattern regulates the expression profiles of genes. In this review, first we briefly describe regulation mechanisms of NF-κB. Next, published computational simulations on the oscillation of NF-κB are summarised. There are at least 60 reports on the computational simulation and analysis of NF-κB oscillation. Third, the importance of a 'space' for the regulation of oscillation pattern of NF-κB is discussed, showing altered oscillation pattern by the change in spatial parameters such as diffusion coefficient, nuclear to cytoplasmic volume ratio (N/C ratio), and transport through nuclear membrane. Finally, simulations in a true intracellular space (TiCS), which is an intracellular 3D space reconstructed in a computer with organelles such as nucleus and mitochondria are discussed.
Collapse
|
236
|
Hong FF, Wang YF, Liu H, Yang MW, Yang SL. V-PYRRO/NO downregulates mRNA expression levels of leukotriene C4 synthase during hepatic ischemia reperfusion injury in rats via inhibition of the nuclear factor-κB activation pathway. Biomed Rep 2016; 4:112-116. [PMID: 26870346 DOI: 10.3892/br.2015.533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/18/2015] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to explore the mechanism underlying the effects of a selective liver nitric oxide (NO) donor, O2-vinyl1-(pyrrolidin-1-yl)-diazen-1-ium-1,2-diolate (V-PYRRO/NO), on the gene expression of leukotriene C4 synthase (LTC4S) during hepatic ischemia/reperfusion (I/R). Adult male Sprague-Dawley rats were divided into 3 groups: Sham (control), I/R and V-PYRRO/NO + I/R groups. The liver was subjected to 1 h of partial hepatic ischemia followed by 5 h of reperfusion, saline or V-PYRRO/NO (1.06 µmol/kg/h) administered intravenously. The mRNA expression levels of LTC4S in rat liver tissue were examined by the reverse transcription-polymerase chain reaction method, the protein expression levels of nuclear factor-κB (NF-κB) p65, p50 and IκBα in liver cell lysates and nuclear extracts were detected by western blot analysis. Hepatic mRNA expression of LTC4S was lower in V-PYRRO/NO + I/R group compared to the I/R group. In addition, the protein expression levels of NF-κB p65 and p50 in the nucleus extract were lower in the V-PYRRO/NO + I/R group when compared with the I/R group. However, the IκBα protein in the 3 groups was not changed. Immunohistochemistry staining revealed that the I/R liver exhibited strong cytoplasmic and nuclear staining for NF-κB p65; however, the V-PYRRO/NO + I/R group liver presented slight cytoplasmic and nuclear staining. In conclusion, V-PYRRO/NO may downregulate LTC4S mRNA expression by inhibiting NF-κB activation independent of IκBα during hepatic I/R injury.
Collapse
Affiliation(s)
- Feng-Fang Hong
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China; Department of Medical Experimental Teaching, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi-Fan Wang
- Institute of Cancer Research, Jiangxi Academy of Medical Science, Nanchang, Jiangxi 330006, P.R. China
| | - Hui Liu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Mei-Wen Yang
- Fuzhou Medical College, Nanchang University, Fuzhou 344000, P.R. China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
237
|
Maldonado MD, García-Moreno H, González-Yanes C, Calvo JR. Possible Involvement of the Inhibition of NF-κB Factor in Anti-Inflammatory Actions That Melatonin Exerts on Mast Cells. J Cell Biochem 2016; 117:1926-33. [PMID: 26756719 DOI: 10.1002/jcb.25491] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/08/2016] [Indexed: 12/23/2022]
Abstract
Melatonin is a molecule endogenously produced in a wide variety of immune cells, including mast cells (RBL-2H3). It exhibits immunomodulatory, anti-inflammatory and anti-apoptotic properties. The physiologic mechanisms underlying these activities of melatonin have not been clarified in mast cells. This work is designed to determine the anti-inflammatory effect and mechanism of action of melatonin on activated mast cells. RBL-2H3 were pre-treated with exogenous melatonin (MELx) at physiological (100nM) and pharmacological (1 mM) doses for 30 min, washed and activated with PMACI (phorbol 12-myristate 13-acetate plus calcium ionophore A23187) for 2 h and 12 h. The data shows that pre-treatment of MELx in stimulated mast cells, significantly reduced the levels of endogenous melatonin production (MELn), TNF-α and IL-6. These effects are directly related with the MELx concentration used. MELx also inhibited IKK/NF-κB signal transduction pathway in stimulated mast cells. These results indicate a molecular basis for the ability of melatonin to prevent inflammation and for the treatment of allergic inflammatory diseases through the down-regulation of mast cell activation. J. Cell. Biochem. 117: 1926-1933, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M D Maldonado
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain
| | - H García-Moreno
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain
| | - C González-Yanes
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain
| | - J R Calvo
- Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Spain
| |
Collapse
|
238
|
Mamińska A, Bartosik A, Banach-Orłowska M, Pilecka I, Jastrzębski K, Zdżalik-Bielecka D, Castanon I, Poulain M, Neyen C, Wolińska-Nizioł L, Toruń A, Szymańska E, Kowalczyk A, Piwocka K, Simonsen A, Stenmark H, Fürthauer M, González-Gaitán M, Miaczynska M. ESCRT proteins restrict constitutive NF-κB signaling by trafficking cytokine receptors. Sci Signal 2016; 9:ra8. [PMID: 26787452 DOI: 10.1126/scisignal.aad0848] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Because signaling mediated by the transcription factor nuclear factor κB (NF-κB) is initiated by ligands and receptors that can undergo internalization, we investigated how endocytic trafficking regulated this key physiological pathway. We depleted all of the ESCRT (endosomal sorting complexes required for transport) subunits, which mediate receptor trafficking and degradation, and found that the components Tsg101, Vps28, UBAP1, and CHMP4B were essential to restrict constitutive NF-κB signaling in human embryonic kidney 293 cells. In the absence of exogenous cytokines, depletion of these proteins led to the activation of both canonical and noncanonical NF-κB signaling, as well as the induction of NF-κB-dependent transcriptional responses in cultured human cells, zebrafish embryos, and fat bodies in flies. These effects depended on cytokine receptors, such as the lymphotoxin β receptor (LTβR) and tumor necrosis factor receptor 1 (TNFR1). Upon depletion of ESCRT subunits, both receptors became concentrated on and signaled from endosomes. Endosomal accumulation of LTβR induced its ligand-independent oligomerization and signaling through the adaptors TNFR-associated factor 2 (TRAF2) and TRAF3. These data suggest that ESCRTs constitutively control the distribution of cytokine receptors in their ligand-free state to restrict their signaling, which may represent a general mechanism to prevent spurious activation of NF-κB.
Collapse
Affiliation(s)
- Agnieszka Mamińska
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Anna Bartosik
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | | | - Iwona Pilecka
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kamil Jastrzębski
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | | | - Irinka Castanon
- Department of Biochemistry, University of Geneva, 1211 Geneva, Switzerland
| | - Morgane Poulain
- Institut de Biologie Valrose, CNRS UMR 7277, INSERM 1091, University of Nice Sophia Antipolis, 06108 Nice, France
| | - Claudine Neyen
- École Polytechnique Fédérale de Lausanne (EPFL), Global Health Institute, 1015 Lausanne, Switzerland
| | | | - Anna Toruń
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Ewelina Szymańska
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Agata Kowalczyk
- Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | | | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | - Maximilian Fürthauer
- Institut de Biologie Valrose, CNRS UMR 7277, INSERM 1091, University of Nice Sophia Antipolis, 06108 Nice, France
| | | | - Marta Miaczynska
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland.
| |
Collapse
|
239
|
Lycopene acts through inhibition of IκB kinase to suppress NF-κB signaling in human prostate and breast cancer cells. Tumour Biol 2016; 37:9375-85. [PMID: 26779636 DOI: 10.1007/s13277-016-4798-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/06/2016] [Indexed: 01/03/2023] Open
Abstract
We studied the effect of the potent dietary antioxidant lycopene on multiple points along the nuclear factor kappa B (NF-κB) signaling pathway in prostate and breast cancer cells. Lycopene significantly inhibited prostate and breast cancer cell growth at physiologically relevant concentrations of ≥1.25 μM. Similar concentrations also caused a 30-40 % reduction in inhibitor of kappa B (IκB) phosphorylation in the cells, as determined by western blotting. Furthermore, the same degree of inhibition by lycopene was observed for NF-κB transcriptional activity, as determined by reporter gene assay. Concomitant with this, immunofluorescence staining of lycopene-treated cells showed a significant suppression (≥25 %) of TNF-induced NF-κB p65 subunit nuclear translocation. Further probing of lycopene's effects on upstream elements of the NF-κB pathway showed a 25 % inhibition of both activity of recombinant IκB kinase β (IKKβ) kinase in a cell-free in vitro assay, as well as activity of IKKβ immunoprecipitated from MDA-MB-231 cells treated with lycopene. In conclusion, the anticancer properties of lycopene may occur through inhibition of the NF-κB signaling pathway, beginning at the early stage of cytoplasmic IKK kinase activity, which then leads to reduced NF-κB-responsive gene regulation. Furthermore, these effects in cancer cells were observed at concentrations of lycopene that are relevant and achievable in vivo.
Collapse
|
240
|
Kastrati I, Siklos MI, Calderon-Gierszal EL, El-Shennawy L, Georgieva G, Thayer EN, Thatcher GRJ, Frasor J. Dimethyl Fumarate Inhibits the Nuclear Factor κB Pathway in Breast Cancer Cells by Covalent Modification of p65 Protein. J Biol Chem 2015; 291:3639-47. [PMID: 26683377 DOI: 10.1074/jbc.m115.679704] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/08/2023] Open
Abstract
In breast tumors, activation of the nuclear factor κB (NFκB) pathway promotes survival, migration, invasion, angiogenesis, stem cell-like properties, and resistance to therapy--all phenotypes of aggressive disease where therapy options remain limited. Adding an anti-inflammatory/anti-NFκB agent to breast cancer treatment would be beneficial, but no such drug is approved as either a monotherapy or adjuvant therapy. To address this need, we examined whether dimethyl fumarate (DMF), an anti-inflammatory drug already in clinical use for multiple sclerosis, can inhibit the NFκB pathway. We found that DMF effectively blocks NFκB activity in multiple breast cancer cell lines and abrogates NFκB-dependent mammosphere formation, indicating that DMF has anti-cancer stem cell properties. In addition, DMF inhibits cell proliferation and significantly impairs xenograft tumor growth. Mechanistically, DMF prevents p65 nuclear translocation and attenuates its DNA binding activity but has no effect on upstream proteins in the NFκB pathway. Dimethyl succinate, the inactive analog of DMF that lacks the electrophilic double bond of fumarate, is unable to inhibit NFκB activity. Also, the cell-permeable thiol N-acetyl l-cysteine, reverses DMF inhibition of the NFκB pathway, supporting the notion that the electrophile, DMF, acts via covalent modification. To determine whether DMF interacts directly with p65, we synthesized and used a novel chemical probe of DMF by incorporating an alkyne functionality and found that DMF covalently modifies p65, with cysteine 38 being essential for the activity of DMF. These results establish DMF as an NFκB inhibitor with anti-tumor activity that may add therapeutic value in the treatment of aggressive breast cancers.
Collapse
Affiliation(s)
| | - Marton I Siklos
- Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois 60612
| | | | | | | | - Emily N Thayer
- Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Gregory R J Thatcher
- Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jonna Frasor
- From the Departments of Physiology and Biophysics and
| |
Collapse
|
241
|
Shih RH, Wang CY, Yang CM. NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review. Front Mol Neurosci 2015; 8:77. [PMID: 26733801 PMCID: PMC4683208 DOI: 10.3389/fnmol.2015.00077] [Citation(s) in RCA: 617] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022] Open
Abstract
The NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) transcription factor family is a pleiotropic regulator of many cellular signaling pathways, providing a mechanism for the cells in response to a wide variety of stimuli linking to inflammation. The stimulated cells will be regulated by not only the canonical but also non-canonical NF-κB pathways. To initiate both of these pathways, IκB-degradation triggers NF-κB release and the nuclear translocated-heterodimer (or homodimer) can associate with the κB sites of promoter to regulate the gene transcriptions. NF-κB ubiquitously expresses in neurons and the constitutive NF-κB activation is associated with processing of neuronal information. NF-κB can regulate the transcription of genes such as chemokines, cytokines, proinflammatory enzymes, adhesion molecules, proinflammatory transcription factors, and other factors to modulate the neuronal survival. In neuronal insult, NF-κB constitutively active in neuron cell bodies can protect neurons against different injuries and regulate the neuronal inflammatory reactions. Besides neurons, NF-κB transcription factors are abundant in glial cells and cerebral blood vessels and the diverse functions of NF-κB also regulate the inflammatory reaction around the neuronal environment. NF-κB transcription factors are abundant in the brain and exhibit diverse functions. Several central nerve system (CNS) diseases are linked to NF-κB activated by inflammatory mediators. The RelA and c-Rel expression produce opposite effects on neuronal survival. Importantly, c-Rel expression in CNS plays a critical role in anti-apoptosis and reduces the age-related behaviors. Moreover, the different subunits of NF-κB dimer formation can modulate the neuroninflammation, neuronal protection, or neurotoxicity. The diverse functions of NF-κB depend on the subunits of the NF-κB dimer-formation which enable us to develop a therapeutic approach to neuroinflammation based on a new concept of inflammation as a strategic tool in neuronal cells. However, the detail role of NF-κB in neuroinflammation, remains to be clarified. In the present article, we provide an updated review of the current state of our knowledge about relationship between NF-κB and neuroinflammation.
Collapse
Affiliation(s)
- Ruey-Horng Shih
- Institute of Neuroscience, National Chengchi University Taipei, Taiwan
| | - Chen-Yu Wang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| |
Collapse
|
242
|
Necrosis-Induced Sterile Inflammation Mediated by Interleukin-1α in Retinal Pigment Epithelial Cells. PLoS One 2015; 10:e0144460. [PMID: 26641100 PMCID: PMC4671579 DOI: 10.1371/journal.pone.0144460] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/18/2015] [Indexed: 12/20/2022] Open
Abstract
Endogenous danger signals released from necrotic cells contribute to retinal inflammation. We have now investigated the effects of necrotic cell extracts prepared from ARPE-19 human retinal pigment epithelial cells (ANCE) on the release of proinflammatory cytokines and chemokines by healthy ARPE-19 cells. ANCE were prepared by subjection of ARPE-19 cells to freeze-thaw cycles. The release of various cytokines and chemokines from ARPE-19 cells was measured with a multiplex assay system or enzyme-linked immunosorbent assays. The expression of interleukin (IL)–1α and the phosphorylation and degradation of the endogenous nuclear factor–κB (NF-κB) inhibitor IκB-α were examined by immunoblot analysis. Among the various cytokines and chemokines examined, we found that ANCE markedly stimulated the release of the proinflammatory cytokine IL-6 and the chemokines IL-8 and monocyte chemoattractant protein (MCP)–1 by ARPE-19 cells. ANCE-induced IL-6, IL-8, and MCP-1 release was inhibited by IL-1 receptor antagonist and by an IKK2 inhibitor (a blocker of NF-κB signaling) in a concentration-dependent manner, but was not affected by a pan-caspase inhibitor (Z-VAD-FMK). Recombinant IL-1α also induced the secretion of IL-6, IL-8, and MCP-1 from ARPE-19 cells, and IL-1α was detected in ANCE. Furthermore, ANCE induced the phosphorylation and degradation of IκB-α in ARPE-19 cells. Our findings thus suggest that IL-1α is an important danger signal that is released from necrotic retinal pigment epithelial cells and triggers proinflammatory cytokine and chemokine secretion from intact cells in a manner dependent on NF-κB signaling. IL-1α is therefore a potential therapeutic target for amelioration of sterile inflammation in the retina.
Collapse
|
243
|
Inhibition of NF-κB by deoxycholic acid induces miR-21/PDCD4-dependent hepatocellular apoptosis. Sci Rep 2015; 5:17528. [PMID: 26621219 PMCID: PMC4664913 DOI: 10.1038/srep17528] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are key regulators of liver metabolism, while toxic bile acids participate in the development of several liver diseases. We previously demonstrated that deoxycholic acid (DCA), a cytotoxic bile acid implicated in the pathogenesis of non-alcoholic fatty liver disease, inhibits miR-21 expression in hepatocytes. Here, we investigated the mechanisms by which DCA modulates miR-21 and whether miR-21 contributes for DCA-induced cytotoxicity. DCA inhibited miR-21 expression in primary rat hepatocytes in a dose-dependent manner, and increased miR-21 pro-apoptotic target programmed cell death 4 (PDCD4) and apoptosis. Both miR-21 overexpression and PDCD4 silencing hampered DCA-induced cell death. Further, DCA decreased NF-κB activity, shown to represent an upstream mechanism leading to modulation of the miR-21/PDCD4 pathway. In fact, NF-κB overexpression or constitutive activation halted miR-21-dependent apoptosis by DCA while opposite results were observed upon NF-κB inhibition. In turn, DCA-induced oxidative stress resulted in caspase-2 activation and NF-κB/miR-21 inhibition, in a PIDD-dependent manner. Finally, modulation of the NF-κB/miR-21/PDCD4 pro-apoptotic pathway by DCA was also shown to occur in the rat liver in vivo. These signalling circuits may constitute appealing targets for bile acid-associated liver pathologies.
Collapse
|
244
|
White CH, Johnston HE, Moesker B, Manousopoulou A, Margolis DM, Richman DD, Spina CA, Garbis SD, Woelk CH, Beliakova-Bethell N. Mixed effects of suberoylanilide hydroxamic acid (SAHA) on the host transcriptome and proteome and their implications for HIV reactivation from latency. Antiviral Res 2015; 123:78-85. [PMID: 26343910 PMCID: PMC5606336 DOI: 10.1016/j.antiviral.2015.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 08/22/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023]
Abstract
Suberoylanilide hydroxamic acid (SAHA) has been assessed in clinical trials as part of a "shock and kill" strategy to cure HIV-infected patients. While it was effective at inducing expression of HIV RNA ("shock"), treatment with SAHA did not result in a reduction of reservoir size ("kill"). We therefore utilized a combined analysis of effects of SAHA on the host transcriptome and proteome to dissect its mechanisms of action that may explain its limited success in "shock and kill" strategies. CD4+ T cells from HIV seronegative donors were treated with 1μM SAHA or its solvent dimethyl sulfoxide (DMSO) for 24h. Protein expression and post-translational modifications were measured with iTRAQ proteomics using ultra high-precision two-dimensional liquid chromatography-tandem mass spectrometry. Gene expression was assessed by Illumina microarrays. Using limma package in the R computing environment, we identified 185 proteins, 18 phosphorylated forms, 4 acetylated forms and 2982 genes, whose expression was modulated by SAHA. A protein interaction network integrating these 4 data types identified the HIV transcriptional repressor HMGA1 to be upregulated by SAHA at the transcript, protein and acetylated protein levels. Further functional category assessment of proteins and genes modulated by SAHA identified gene ontology terms related to NFκB signaling, protein folding and autophagy, which are all relevant to HIV reactivation. In summary, SAHA modulated numerous host cell transcripts, proteins and post-translational modifications of proteins, which would be expected to have very mixed effects on the induction of HIV-specific transcription and protein function. Proteome profiling highlighted a number of potential counter-regulatory effects of SAHA with respect to viral induction, which transcriptome profiling alone would not have identified. These observations could lead to a more informed selection and design of other HDACi with a more refined targeting profile, and prioritization of latency reversing agents of other classes to be used in combination with SAHA to achieve more potent induction of HIV expression.
Collapse
Affiliation(s)
- Cory H White
- Graduate Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA 92093, USA; San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA
| | - Harvey E Johnston
- Cancer Sciences Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK; Centre for Proteomic Research, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, UK
| | - Bastiaan Moesker
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK
| | - Antigoni Manousopoulou
- Centre for Proteomic Research, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, UK; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK
| | - David M Margolis
- Departments of Medicine, Microbiology and Immunology, Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Douglas D Richman
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA; Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Celsa A Spina
- San Diego VA Medical Center and Veterans Medical Research Foundation, San Diego, CA 92161, USA; Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Spiros D Garbis
- Cancer Sciences Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK; Centre for Proteomic Research, Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton, UK; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK.
| | - Christopher H Woelk
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hants SO16 6YD, UK.
| | | |
Collapse
|
245
|
Hochrainer K, Pejanovic N, Olaseun VA, Zhang S, Iadecola C, Anrather J. The ubiquitin ligase HERC3 attenuates NF-κB-dependent transcription independently of its enzymatic activity by delivering the RelA subunit for degradation. Nucleic Acids Res 2015; 43:9889-904. [PMID: 26476452 PMCID: PMC4787756 DOI: 10.1093/nar/gkv1064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/06/2015] [Indexed: 11/21/2022] Open
Abstract
Activation of NF-κB-dependent transcription represents an important hallmark of inflammation. While the acute inflammatory response is per se beneficial, it can become deleterious if its spatial and temporal profile is not tightly controlled. Classically, NF-κB activity is limited by cytoplasmic retention of the NF-κB dimer through binding to inhibitory IκB proteins. However, increasing evidence suggests that NF-κB activity can also be efficiently contained by direct ubiquitination of NF-κB subunits. Here, we identify the HECT-domain ubiquitin ligase HERC3 as novel negative regulator of NF-κB activity. We find that HERC3 restricts NF-κB nuclear import and DNA binding without affecting IκBα degradation. Instead HERC3 indirectly binds to the NF-κB RelA subunit after liberation from IκBα inhibitor leading to its ubiquitination and protein destabilization. Remarkably, the regulation of RelA activity by HERC3 is independent of its inherent ubiquitin ligase activity. Rather, we show that HERC3 and RelA are part of a multi-protein complex containing the proteasome as well as the ubiquitin-like protein ubiquilin-1 (UBQLN1). We present evidence that HERC3 and UBQLN1 provide a link between NF-κB RelA and the 26S proteasome, thereby facilitating RelA protein degradation. Our findings establish HERC3 as novel candidate regulating the inflammatory response initiated by NF-κB.
Collapse
Affiliation(s)
- Karin Hochrainer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| | - Nadja Pejanovic
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA Instituto Gulbenkian de Ciência, Apartado 14, Oeiras, Portugal
| | | | - Sheng Zhang
- Institute of Biotechnology and Life Sciences Biotechnologies, Cornell University, Ithaca, NY14853, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY10065, USA
| |
Collapse
|
246
|
Ampofo E, Rudzitis-Auth J, Dahmke IN, Rössler OG, Thiel G, Montenarh M, Menger MD, Laschke MW. Inhibition of protein kinase CK2 suppresses tumor necrosis factor (TNF)-α-induced leukocyte–endothelial cell interaction. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2123-36. [DOI: 10.1016/j.bbadis.2015.07.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 06/29/2015] [Accepted: 07/15/2015] [Indexed: 12/22/2022]
|
247
|
Mia MM, Bank RA. The IκB kinase inhibitor ACHP strongly attenuates TGFβ1-induced myofibroblast formation and collagen synthesis. J Cell Mol Med 2015; 19:2780-92. [PMID: 26337045 PMCID: PMC4687706 DOI: 10.1111/jcmm.12661] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/23/2015] [Indexed: 01/07/2023] Open
Abstract
Excessive accumulation of a collagen‐rich extracellular matrix (ECM) by myofibroblasts is a characteristic feature of fibrosis, a pathological state leading to serious organ dysfunction. Transforming growth factor beta1 (TGFβ1) is a strong inducer of myofibroblast formation and subsequent collagen production. Currently, there are no remedies for the treatment of fibrosis. Activation of the nuclear factor kappa B (NF‐κB) pathway by phosphorylating IκB with the enzyme IκB kinase (IKK) plays a major role in the induction of fibrosis. ACHP {2‐Amino‐6‐[2‐(cyclopropylmethoxy)‐6‐hydroxyphenyl]‐4‐(4‐piperidinyl)‐3 pyridinecarbonitrile}, a selective inhibitor of IKK, prohibits the activation of the NF‐κB pathway. It is not known whether ACHP has potential anti‐fibrotic properties. Using adult human dermal and lung fibroblasts we have investigated whether ACHP has the ability to inhibit the TGFβ1‐induced transition of fibroblasts into myofibroblasts and its excessive synthesis of ECM. The presence of ACHP strongly suppressed the induction of the myofibroblast markers alpha‐smooth muscle actin (αSMA) and SM22α, as well as the deposition of the ECM components collagen type I and fibronectin. Furthermore, post‐treatment with ACHP partly reversed the expression of αSMA and collagen type I production. Finally, ACHP suppressed the expression of the three collagen‐modifying enzymes lysyl hydroxylase (PLOD1,PLOD2 and PLOD3) in dermal fibroblasts, but did not do so in lung fibroblasts. We conclude that the IKK inhibitor ACHP has potent antifibrotic properties, and that the NF‐κB pathway plays an important role in myofibroblast biology.
Collapse
Affiliation(s)
- Masum M Mia
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ruud A Bank
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
248
|
Karki P, Kim C, Smith K, Son DS, Aschner M, Lee E. Transcriptional Regulation of the Astrocytic Excitatory Amino Acid Transporter 1 (EAAT1) via NF-κB and Yin Yang 1 (YY1). J Biol Chem 2015; 290:23725-37. [PMID: 26269591 DOI: 10.1074/jbc.m115.649327] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Indexed: 12/19/2022] Open
Abstract
Astrocytic glutamate transporter excitatory amino acid transporter (EAAT) 1, also known as glutamate aspartate transporter (GLAST) in rodents, is one of two glial glutamate transporters that are responsible for removing excess glutamate from synaptic clefts to prevent excitotoxic neuronal death. Despite its important role in neurophysiological functions, the molecular mechanisms of EAAT1 regulation at the transcriptional level remain to be established. Here, we report that NF-κB is a main positive transcription factor for EAAT1, supported by the following: 1) EAAT1 contains two consensus sites for NF-κB, 2) mutation of NF-κB binding sites decreased EAAT1 promoter activity, and 3) activation of NF-κB increased, whereas inhibition of NF-κB decreased EAAT1 promoter activity and mRNA/protein levels. EGF increased EAAT1 mRNA/protein levels and glutamate uptake via NF-κB. The transcription factor yin yang 1 (YY1) plays a role as a critical negative regulator of EAAT1, supported by the following: 1) the EAAT1 promoter contains multiple consensus sites for YY1, 2) overexpression of YY1 decreased EAAT1 promoter activity and mRNA/protein levels, and 3) knockdown of YY1 increased EAAT1 promoter activity and mRNA/protein levels. Manganese decreased EAAT1 expression via YY1. Epigenetic modifiers histone deacetylases (HDACs) served as co-repressors of YY1 to further decrease EAAT1 promoter activity, whereas inhibition of HDACs reversed manganese-induced decrease of EAAT1 expression. Taken together, our findings suggest that NF-κB is a critical positive regulator of EAAT1, mediating the stimulatory effects of EGF, whereas YY1 is a negative regulator of EAAT1 with HDACs as co-repressors, mediating the inhibitory effects of manganese on EAAT1 regulation.
Collapse
Affiliation(s)
- Pratap Karki
- From the Department of Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Clifford Kim
- the Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Keisha Smith
- From the Department of Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Deok-Soo Son
- From the Department of Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208
| | - Michael Aschner
- the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Eunsook Lee
- From the Department of Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee 37208,
| |
Collapse
|
249
|
MTMR3 risk allele enhances innate receptor-induced signaling and cytokines by decreasing autophagy and increasing caspase-1 activation. Proc Natl Acad Sci U S A 2015; 112:10461-6. [PMID: 26240347 DOI: 10.1073/pnas.1501752112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by dysregulated host:microbial interactions and cytokine production. Host pattern recognition receptors (PRRs) are critical in regulating these interactions. Multiple genetic loci are associated with IBD, but altered functions for most, including in the rs713875 MTMR3/HORMAD2/LIF/OSM region, are unknown. We identified a previously undefined role for myotubularin-related protein 3 (MTMR3) in amplifying PRR-induced cytokine secretion in human macrophages and defined MTMR3-initiated mechanisms contributing to this amplification. MTMR3 decreased PRR-induced phosphatidylinositol 3-phosphate (PtdIns3P) and autophagy levels, thereby increasing PRR-induced caspase-1 activation, autocrine IL-1β secretion, NFκB signaling, and, ultimately, overall cytokine secretion. This MTMR3-mediated regulation required the N-terminal pleckstrin homology-GRAM domain and Cys413 within the phosphatase domain of MTMR3. In MTMR3-deficient macrophages, reducing the enhanced autophagy or restoring NFκB signaling rescued PRR-induced cytokines. Macrophages from rs713875 CC IBD risk carriers demonstrated increased MTMR3 expression and, in turn, decreased PRR-induced PtdIns3P and autophagy and increased PRR-induced caspase-1 activation, signaling, and cytokine secretion. Thus, the rs713875 IBD risk polymorphism increases MTMR3 expression, which modulates PRR-induced outcomes, ultimately leading to enhanced PRR-induced cytokines.
Collapse
|
250
|
Lee SG, Jung JY, Shin JS, Shin KS, Cho CW, Rhee YK, Hong HD, Lee KT. Immunostimulatory polysaccharide isolated from the leaves of Diospyros kaki Thumb modulate macrophage via TLR2. Int J Biol Macromol 2015; 79:971-82. [DOI: 10.1016/j.ijbiomac.2015.06.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/28/2015] [Accepted: 06/14/2015] [Indexed: 01/22/2023]
|