1
|
Wang Z, Su X, Zhan Z, Wang H, Zhou S, Mao J, Xu H, Duan S. miR-660: A novel regulator in human cancer pathogenesis and therapeutic implications. Gene 2025; 953:149434. [PMID: 40120868 DOI: 10.1016/j.gene.2025.149434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression. Among these, miR-660, located on chromosome Xp11.23, is increasingly studied for its role in cancer due to its abnormal expression in various biological contexts. It is regulated by 8 competing endogenous RNAs (ceRNAs), which adds complexity to its function. miR- 660 targets 19 genes involved in 6 pathways such as PI3K/AKT/mTOR, STAT3, Wnt/β-catenin, p53, NF‑κB, and RAS, influencing cell cycle, proliferation, apoptosis, and invasion/migration. It also plays a role in resistance to chemotherapies like cisplatin, gemcitabine, and sorafenib in lung adenocarcinoma (LUAD), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC), thus highlighting its clinical importance. Additionally, leveraging liposomes as nanocarriers presents a promising avenue for enhancing cancer drug delivery. Our comprehensive study not only elucidates the aberrant expression patterns, biological functions, and regulatory networks of miR-660 and its ceRNAs but also delves into the intricate signaling pathways implicated. We envisage that our findings will furnish a robust framework and serve as a seminal reference for future investigations of miR-660, fostering advancements in cancer research and potentially catalyzing breakthroughs in cancer diagnosis and treatment paradigms.
Collapse
Affiliation(s)
- Zehua Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhiqing Zhan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hangxuan Wang
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shuhan Zhou
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Jiasheng Mao
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Qin TX, Zhu YY, Ng WH, Ng SK, Chek MF, Tang KD. NF-kappa-B inhibitor alpha mediates cancer stemness characteristics in oral squamous cell carcinoma by interacting with cathepsin B. Int J Biol Macromol 2025; 311:143690. [PMID: 40316102 DOI: 10.1016/j.ijbiomac.2025.143690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
The role of NF-kappa-B inhibitor alpha (IκBα), a well-established negative regulator of the NF-κB signalling pathway in cancer is complex, as evidenced by either promoting or suppressing tumourigenesis, depending on the cancer type, however, in oral squamous cell carcinoma (OSCC) remains unelucidated. Here, for the first time, we report that the elevated levels of both NKFBIA mRNA and IκBα protein in OSCC tumour tissues and OSCC cell lines. Meanwhile, IκBα silencing resulted in the suppression of cell proliferation, migration and invasion in OSCC. In addition, we demonstrated that IκBα can mediate the OSCC stemness and epithelial-mesenchymal transition (EMT) characteristics by directly interacting with cathepsin B (CTSB) and thus, facilitating the progression toward carcinogenesis. More importantly, we identified Psammaplin A (PsA), a natural metabolite derived from marine sponges that would disrupt the IκBα-CTSB interaction via competition, thereby reducing oral spheres formation, cell viability and OSCC tumour growth in vivo, clearly demonstrating its potential as an effective therapeutic agent that specifically targets this oncogenic complex. In summary, we have unveiled a novel mechanism underlying the oncogenic role of IκBα-CTSB complex in OSCC, which may offer the therapeutic potential of targeting this complex with PsA for the treatment of OSCC.
Collapse
Affiliation(s)
- Tian Xu Qin
- Nankai University, TEDA School of Biological Sciences and Biotechnology, Tianjin 300457, PR China; Nankai University, Nankai International Advanced Research Institute (Shenzhen Futian), Shenzhen, Guangdong 518045, PR China
| | - Ying Ying Zhu
- Nankai University, TEDA School of Biological Sciences and Biotechnology, Tianjin 300457, PR China; Nankai University, Nankai International Advanced Research Institute (Shenzhen Futian), Shenzhen, Guangdong 518045, PR China
| | - Wai Hoe Ng
- Queen's University Belfast, School of Medicine, Dentistry and Biomedical Sciences, Belfast BT7 1NN, UK
| | - Siew Kit Ng
- Universiti Sains Malaysia, Department of Biomedical Science, Advanced Medical and Dental Institute, Bertam 13200, Kepala Batas, Pulau Pinang, Malaysia
| | - Min Fey Chek
- Nara Institute of Science and Technology, Structural Biology and Protein Engineering Laboratory, Institute for Research Initiatives, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Kai Dun Tang
- Nankai University, TEDA School of Biological Sciences and Biotechnology, Tianjin 300457, PR China; Nankai University, Nankai International Advanced Research Institute (Shenzhen Futian), Shenzhen, Guangdong 518045, PR China.
| |
Collapse
|
3
|
Chen S, Zhu L, Fang X, Appiah C, Ji Y, Chen Z, Qiao S, Gong C, Li J, Zhao Y. Alloferon Mitigates LPS-Induced Endometritis by Attenuating the NLRP3/CASP1/IL-1β/IL-18 Signaling Cascade. Inflammation 2025; 48:730-746. [PMID: 38913143 DOI: 10.1007/s10753-024-02083-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024]
Abstract
Endometritis is an inflammatory reaction of the uterine lining that can lead to infertility. Alloferon, a linear non-glycosylated oligopeptide, has been recognized for its potent anti-inflammatory and immunomodulatory effects. In light of these attributes, this study aims to explore the potential therapeutic effects of alloferon in alleviating endometrial inflammation induced by lipopolysaccharide (LPS), while elucidating the underlying protective mechanisms. Two conditions representing pre- and post-menopause states were simulated using an ovariectomized (Ovx) murine model. The findings underscore alloferon's remarkable capacity to alleviate cardinal signs of endometritis, including redness, swelling, and congestion, while concurrently restoring the structural integrity of the endometrial tissue. Moreover, alloferon effectively modulates the expression of key inflammatory mediators, such as nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), cysteine aspartate-specific protease 1 (CASP1), interleukin-1β (IL-1β), and interleukin-18 (IL-18). In vitro experiments were conducted to further corroborate and validate these findings. In conclusion, alloferon shows promising potential in mitigating LPS-induced inflammation by attenuating the NLRP3/CASP1/IL-1β/IL-18 signaling cascade.
Collapse
Affiliation(s)
- Shitian Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Xinyu Fang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Clara Appiah
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Yuanbo Ji
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Ziyi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Shuai Qiao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Chen Gong
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Jian Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China.
| |
Collapse
|
4
|
Butera F, Sero JE, Dent LG, Bakal C. Actin networks modulate heterogeneous NF-κB dynamics in response to TNFα. eLife 2024; 13:e86042. [PMID: 39110005 PMCID: PMC11524587 DOI: 10.7554/elife.86042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 08/05/2024] [Indexed: 11/01/2024] Open
Abstract
The canonical NF-κB transcription factor RELA is a master regulator of immune and stress responses and is upregulated in pancreatic ductal adenocardinoma (PDAC) tumours. In this study, we characterised previously unexplored endogenous RELA-GFP dynamics in PDAC cell lines through live single-cell imaging. Our observations revealed that TNFα stimulation induces rapid, sustained, and non-oscillatory nuclear translocation of RELA. Through Bayesian analysis of single-cell datasets with variation in nuclear RELA, we predicted that RELA heterogeneity in PDAC cell lines is dependent on F-actin dynamics. RNA-seq analysis identified distinct clusters of RELA-regulated gene expression in PDAC cells, including TNFα-induced RELA upregulation of the actin regulators NUAK2 and ARHGAP31. Further, siRNA-mediated depletion of ARHGAP31 and NUAK2 altered TNFα-stimulated nuclear RELA dynamics in PDAC cells, establishing a novel negative feedback loop that regulates RELA activation by TNFα. Additionally, we characterised the NF-κB pathway in PDAC cells, identifying how NF-κB/IκB proteins genetically and physically interact with RELA in the absence or presence of TNFα. Taken together, we provide computational and experimental support for interdependence between the F-actin network and the NF-κB pathway with RELA translocation dynamics in PDAC.
Collapse
Affiliation(s)
- Francesca Butera
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| | - Julia E Sero
- Department of Life Sciences, University of BathBathUnited Kingdom
| | - Lucas G Dent
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| | - Chris Bakal
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondonUnited Kingdom
| |
Collapse
|
5
|
Hosseini H, Abbasi A, Sabahi S, Akrami S, Yousefi-Avarvand A. Assessing the Potential Biological Activities of Postbiotics Derived from Saccharomyces cerevisiae: An In Vitro Study. Probiotics Antimicrob Proteins 2024; 16:1348-1364. [PMID: 37402072 DOI: 10.1007/s12602-023-10117-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 07/05/2023]
Abstract
A new biotherapeutic strategy involves the use of microbial bioactive substances (postbiotics) that exhibit optimum compatibility and intimate contact with the immune system of the host. This study was aimed at investigating the potential biological activities of postbiotics derived from Saccharomyces cerevisiae (PTCC 5269) (PSC) under in vitro circumstances. Based on the outcomes, the synthesized PSC possessing a high level of phenolic (102.46 ± 0.25 mg GAE/g) and flavonoid (19.87 ± 75.32 mg QE/g) content demonstrated significant radical scavenging activity (87.34 ± 0.56%); antibacterial action towards Listeria monocytogenes, Streptococcus mutans, Salmonella typhi, and Escherichia coli (in order of effectiveness) in both in vitro and food models (whole milk and ground meat); probiotics' growth-promoting activity in the fermentation medium; α-glucosidase enzyme-inhibiting and cholesterol-lowering properties in a concentration- and pH-dependent manner; reduction in the cell viability (with the significant IC50 values of 34.27 and 23.58 μg/mL after 24 and 48 h, respectively); suppressed the initial (G0/G1) phase of the cell's division; induced apoptosis; and increased the expression of PTEN gene, while the IkB, RelA, and Bcl-XL genes indicated diminished expression in treated SW480 cancer cells. These multiple health-promoting functions of PSC can be extended to medical, biomedical, and food scopes, as novel biotherapeutic approaches, in order to design efficient and optimized functional food formulations or/and supplementary medications to use as adjuvant agents for preventing or/and treating chronic/acute disorders.
Collapse
Affiliation(s)
- Hedayat Hosseini
- Department of Food Science and Technology, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Abbasi
- Department of Food Science and Technology, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Sabahi
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Sousan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arshid Yousefi-Avarvand
- Department of Laboratory Sciences, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
Kochel B. Negative feedback systems for modelling NF-κB transcription factor oscillatory activity. Transcription 2024; 15:65-96. [PMID: 38739365 PMCID: PMC11810101 DOI: 10.1080/21541264.2024.2331887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 05/14/2024] Open
Abstract
Low-dimensional negative feedback systems (NFSs) were developed within a signal flow model to describe the oscillatory activities of NF-κB caused by interactions with its inhibitor IκBα. The NFSs were established as 3rd- and 4th-order linear systems containing unperturbed and perturbed negative feedback (NF) loops with constant or time-varying NF strengths and a feed-forward loop. NF-related analytical solutions to the NFSs representing the time courses of NF-κB and IκBα were determined and their exact mathematical relationship was found. The NFS's parameters were determined to fit the experimental time courses of NF-κB in TNF-α-stimulated embryonic fibroblasts, rela-/- embryonic fibroblasts reconstituted with RelA, C9L cells, GFP-p65 knock-in embryonic fibroblasts and embryogenic fibroblasts lacking Iκβ and IκBε, LPS-stimulated IC-21 macrophages treated or not with DCPA, and anti-IgM-stimulated DT40 B-lymphocytes. The unperturbed and perturbed NFSs describing the above biosystems generated isochronous and non-isochronous solutions, depending on a constant or time-varying NF strength, respectively. The oscillation period of the NF-coupled solutions, the phase difference between them and the time delays in the appearance of cytoplasmic IκBα after stimulation of NF-κB were determined. A significant divergence between the IκBα solutions to the NFSs and the IκBα experimental courses led to a rejection of the NF coupling between NF-κB and IκBα in the above biosystems. It was shown that neither the linearity nor the low dimensionality of the NFSs altered the NF relationship and the divergence between the IκBα solutions to the NFS and IκBα experimental time courses. Although the NF relationship between IκBα and NF-κB was not confirmed in all the experimental data analyzed, delayed negative feedback was found in some cases.
Collapse
Affiliation(s)
- Bonawentura Kochel
- Immunotherapy Central Europe, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
7
|
Wiggins DA, Maxwell JN, Nelson DE. Exploring the role of CITED transcriptional regulators in the control of macrophage polarization. Front Immunol 2024; 15:1365718. [PMID: 38646545 PMCID: PMC11032013 DOI: 10.3389/fimmu.2024.1365718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Macrophages are tissue resident innate phagocytic cells that take on contrasting phenotypes, or polarization states, in response to the changing combination of microbial and cytokine signals at sites of infection. During the opening stages of an infection, macrophages adopt the proinflammatory, highly antimicrobial M1 state, later shifting to an anti-inflammatory, pro-tissue repair M2 state as the infection resolves. The changes in gene expression underlying these transitions are primarily governed by nuclear factor kappaB (NF-κB), Janus kinase (JAK)/signal transducer and activation of transcription (STAT), and hypoxia-inducible factor 1 (HIF1) transcription factors, the activity of which must be carefully controlled to ensure an effective yet spatially and temporally restricted inflammatory response. While much of this control is provided by pathway-specific feedback loops, recent work has shown that the transcriptional co-regulators of the CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxy-terminal domain (CITED) family serve as common controllers for these pathways. In this review, we describe how CITED proteins regulate polarization-associated gene expression changes by controlling the ability of transcription factors to form chromatin complexes with the histone acetyltransferase, CBP/p300. We will also cover how differences in the interactions between CITED1 and 2 with CBP/p300 drive their contrasting effects on pro-inflammatory gene expression.
Collapse
Affiliation(s)
| | | | - David E. Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
8
|
Lee D, V AADLR, Kim Y. Optimal strategies of oncolytic virus-bortezomib therapy via the apoptotic, necroptotic, and oncolysis signaling network. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:3876-3909. [PMID: 38549312 DOI: 10.3934/mbe.2024173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Bortezomib and oncolytic virotherapy are two emerging targeted cancer therapies. Bortezomib, a proteasome inhibitor, disrupts protein degradation in cells, leading to the accumulation of unfolded proteins that induce apoptosis. On the other hand, virotherapy uses genetically modified oncolytic viruses (OVs) to infect cancer cells, trigger cell lysis, and activate anti-tumor response. Despite progress in cancer treatment, identifying administration protocols for therapeutic agents remains a significant concern, aiming to strike a balance between efficacy, minimizing toxicity, and administrative costs. In this work, optimal control theory was employed to design a cost-effective and efficient co-administration protocols for bortezomib and OVs that could significantly diminish the population of cancer cells via the cell death program with the NF$ \kappa $B-BAX-RIP1 signaling network. Both linear and quadratic control strategies were explored to obtain practical treatment approaches by adapting necroptosis protocols to efficient cell death programs. Our findings demonstrated that a combination therapy commencing with the administration of OVs followed by bortezomib infusions yields an effective tumor-killing outcome. These results could provide valuable guidance for the development of clinical administration protocols in cancer treatment.
Collapse
Affiliation(s)
- Donggu Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Aurelio A de Los Reyes V
- Institute of Mathematics, University of the Philippines Diliman, Quezon City 1101, Philippines
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Zhou S, Chen M, Yuan Y, Xu Y, Pu Q, Ai X, Liu S, Du F, Huang X, Dong J, Cui X, Tang Z. Trans-acting aptazyme for conditional gene knockdown in eukaryotic cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:367-375. [PMID: 37547296 PMCID: PMC10400872 DOI: 10.1016/j.omtn.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Trans-acting hammerhead ribozyme inherits the advantages of being the smallest and best-characterized RNA-cleaving ribozyme, offering high modularity and the ability to cleave any desired sequence without the aid of any protein, as long as the target sequence contains a cleavage site. However, achieving precise control over the trans-acting hammerhead ribozyme would enable safer and more accurate regulation of gene expression. Herein, we described an intracellular selection of hammerhead aptazyme that contains a theophylline aptamer on stem II based on toxin protein IbsC. Based on the intracellular selection, we obtained three new cis-acting hammerhead aptazymes. Moreover, the corresponding trans-acting aptazymes could be efficiently induced by theophylline to knock down different targeted genes in eukaryotic cells. Notably, the best one, T195, exhibited a ligand-dependent and dose-dependent response to theophylline, and the cleavage efficiency could be enhanced by incorporating multiplex aptazymes.
Collapse
Affiliation(s)
- Shan Zhou
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Meiyi Chen
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Yi Yuan
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Yan Xu
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Qinlin Pu
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Xilei Ai
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Shuai Liu
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Feng Du
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Xin Huang
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Juan Dong
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Xin Cui
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| | - Zhuo Tang
- Natural Products Research Center, Chengdu Institution of Biology, Chinese Academy of Sciences, Chengdu 610041, P.R. China
| |
Collapse
|
10
|
Kelly-Laubscher R, Somers S, Lacerda L, Lecour S. Role of nuclear factor kappa-B in TNF-induced cytoprotection. Cardiovasc J Afr 2023; 34:74-80. [PMID: 35687060 PMCID: PMC10512038 DOI: 10.5830/cvja-2022-023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 05/10/2022] [Indexed: 06/15/2023] Open
Abstract
Ischaemic heart disease (IHD) is a leading cause of death worldwide. Understanding prosurvival signalling pathways that protect against ischaemia-reperfusion injury (IRI) may assist in the development of novel cardioprotective strategies against IHD. In this regard, the transcription factor, nuclear factor kappa-B (NFκB) is activated by tumour necrosis factor (TNF), but its role in TNF-induced cytoprotection is unknown. Therefore, to investigate the role of NFκB in TNF-induced cytoprotection, C2C12 cells were pretreated with TNF (0.5 ng/ml) in the presence and absence of an NFκB inhibitor, pyrrolidine derivative of dithiocarbamate (PDTC; 100 µM). Cells were subjected to simulated IRI and treated with PDTC, either during TNF exposure or at reperfusion. Phosphorylation of IkB was measured after the TNF stimulus. Cytoprotection by TNF in cells subjected to IRI (cell viability: 43.7 ± 8.1% in control vs 70.6 ± 6.1% with TNF, p < 0.001) was abrogated by co-administration of PDTC (40.6 ± 1.9%, p < 0.001 vs TNF) but not by exposure to PDTC at reperfusion (70.7 ± 1.7%). Cytosolic IkB phosphorylation [1.5 ± 0.2 arbitrary units (AU) for TNF vs 1.0 ± 0.0 for untreated, p < 0.01]) was increased after TNF exposure and this increase was abolished by co-administration with PDTC (0.8 ± 0.3 AU, p < 0 01 vs TNF). Our data suggest that NFκB acts as a key component in TNF-induced cytoprotection. These findings may pave the way for the development of novel therapeutic drugs that target TNF/NFκB signalling to protect against IHD.
Collapse
Affiliation(s)
- Roisin Kelly-Laubscher
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, University of Cape Town, South Africa; and Department of Pharmacology and Therapeutics, College of Medicine and Health, University College Cork, Ireland.
| | - Sarin Somers
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, University of Cape Town, South Africa.
| | - Lydia Lacerda
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
11
|
Deciphering signal transduction networks in the liver by mechanistic mathematical modelling. Biochem J 2022; 479:1361-1374. [PMID: 35748700 PMCID: PMC9246346 DOI: 10.1042/bcj20210548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022]
Abstract
In health and disease, liver cells are continuously exposed to cytokines and growth factors. While individual signal transduction pathways induced by these factors were studied in great detail, the cellular responses induced by repeated or combined stimulations are complex and less understood. Growth factor receptors on the cell surface of hepatocytes were shown to be regulated by receptor interactions, receptor trafficking and feedback regulation. Here, we exemplify how mechanistic mathematical modelling based on quantitative data can be employed to disentangle these interactions at the molecular level. Crucial is the analysis at a mechanistic level based on quantitative longitudinal data within a mathematical framework. In such multi-layered information, step-wise mathematical modelling using submodules is of advantage, which is fostered by sharing of standardized experimental data and mathematical models. Integration of signal transduction with metabolic regulation in the liver and mechanistic links to translational approaches promise to provide predictive tools for biology and personalized medicine.
Collapse
|
12
|
Dependency of EGFR activation in vanadium-based sensitization to oncolytic virotherapy. Mol Ther Oncolytics 2022; 25:146-159. [PMID: 35572196 PMCID: PMC9065483 DOI: 10.1016/j.omto.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Oncolytic virotherapy is a clinically validated approach to treat cancers such as melanoma; however, tumor resistance to virus makes its efficacy variable. Compounds such as sodium orthovanadate (vanadate) can overcome viral resistance and synergize with RNA-based oncolytic viruses. In this study, we explored the basis of vanadate mode of action and identified key cellular components in vanadate’s oncolytic virus-enhancing mechanism using a high-throughput kinase inhibitor screen. We found that several kinase inhibitors affecting signaling downstream of the epidermal growth factor receptor (EGFR) pathway abrogated the oncolytic virus-enhancing effects of vanadate. EGFR pathway inhibitors such as gefitinib negated vanadate-associated changes in the phosphorylation and localization of STAT1/2 as well as NF-κB signaling. Moreover, gefitinib treatment could abrogate the viral sensitizing response of vanadium compounds in vivo. Together, we demonstrate that EGFR signaling plays an integral role in vanadium viral sensitization and that pharmacological EGFR blockade can counteract vanadium/oncolytic virus combination therapy.
Collapse
|
13
|
Maladaptation after a virus host switch leads to increased activation of the pro-inflammatory NF-κB pathway. Proc Natl Acad Sci U S A 2022; 119:e2115354119. [PMID: 35549551 PMCID: PMC9171774 DOI: 10.1073/pnas.2115354119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Myxoma virus (MYXV) is benign in the natural brush rabbit host but causes a fatal disease in European rabbits. Here, we demonstrate that MYXV M156 inhibited brush rabbit protein kinase R (bPKR) more efficiently than European rabbit PKR (ePKR). Because ePKR was not completely inhibited by M156, there was a depletion of short–half-life proteins like the nuclear factor kappa B (NF-κB) inhibitor IκBα, concomitant NF-κB activation and NF-κB target protein expression in ePKR-expressing cells. NF-κB pathway activation was blocked by either hypoactive or hyperactive M156 mutants. This demonstrates that maladaptation of viral immune antagonists can result in substantially different immune responses in aberrant hosts. These different host responses may contribute to altered viral dissemination and may influence viral pathogenesis. Myxoma virus (MYXV) causes localized cutaneous fibromas in its natural hosts, tapeti and brush rabbits; however, in the European rabbit, MYXV causes the lethal disease myxomatosis. Currently, the molecular mechanisms underlying this increased virulence after cross-species transmission are poorly understood. In this study, we investigated the interaction between MYXV M156 and the host protein kinase R (PKR) to determine their crosstalk with the proinflammatory nuclear factor kappa B (NF-κB) pathway. Our results demonstrated that MYXV M156 inhibits brush rabbit PKR (bPKR) more strongly than European rabbit PKR (ePKR). This moderate ePKR inhibition could be improved by hyperactive M156 mutants. We hypothesized that the moderate inhibition of ePKR by M156 might incompletely suppress the signal transduction pathways modulated by PKR, such as the NF-κB pathway. Therefore, we analyzed NF-κB pathway activation with a luciferase-based promoter assay. The moderate inhibition of ePKR resulted in significantly higher NF-κB–dependent reporter activity than complete inhibition of bPKR. We also found a stronger induction of the NF-κB target genes TNFα and IL-6 in ePKR-expressing cells than in bPKR-expressing cells in response to M156 in both transfection and infections assays. Furthermore, a hyperactive M156 mutant did not cause ePKR-dependent NF-κB activation. These observations indicate that M156 is maladapted for ePKR inhibition, only incompletely blocking translation in these hosts, resulting in preferential depletion of short–half-life proteins, such as the NF-κB inhibitor IκBα. We speculate that this functional activation of NF-κB induced by the intermediate inhibition of ePKR by M156 may contribute to the increased virulence of MYXV in European rabbits.
Collapse
|
14
|
Yang H, Wang Y, Liu M, Liu X, Jiao Y, Jin S, Shan A, Feng X. Effects of Dietary Resveratrol Supplementation on Growth Performance and Anti-Inflammatory Ability in Ducks ( Anas platyrhynchos) through the Nrf2/HO-1 and TLR4/NF-κB Signaling Pathways. Animals (Basel) 2021; 11:3588. [PMID: 34944363 PMCID: PMC8698092 DOI: 10.3390/ani11123588] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to explore the effect of dietary resveratrol on the growth performance and anti-inflammatory mechanism in ducks. A total of 280 one-day-old specific pathogen-free male ducklings (Anas platyrhynchos) with an average body weight of 35 ± 1 g were randomly divided into two dietary treatment groups with different supplementation levels of resveratrol for growth performance experiments: R0 and R400 (0 and, 400 mg kg-1 resveratrol, respectively). At the age of 28 days, 16 ducks were selected from each treatment group and divided into four subgroups for a 2 × 2 factorial pathological experiment: R0; R400; R0 + LPS; R400 + LPS, (0 mg kg-1 resveratrol, 400 mg kg-1 resveratrol, 0 mg kg-1 resveratrol, 400 mg kg-1 resveratrol + 5 mg lipopolysaccharide/kg body weight). The results showed that resveratrol significantly improved final body weight and average daily gain (p < 0.01) and alleviated the lipopolysaccharide-induced inflammatory response with a reduction in IL-1β and IL-6 in the plasma and the liver (p < 0.05). Resveratrol improved mRNA levels of Nrf2 and HO-1 and decreased the mRNA levels of TLR4 and NF-κB in duck liver (p < 0.05). Dietary resveratrol can improve growth performance and reduce inflammation through the Nrf2/HO-1 and TLR4/NF-κB signaling pathways in duck.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xingjun Feng
- Laboratory of Molecular Nutrition, Institute of Animal Nutrition, Northeast Agricultural University, Changjiang Street 600#, Xiangfang District, Harbin 150030, China; (H.Y.); (Y.W.); (M.L.); (X.L.); (Y.J.); (S.J.); (A.S.)
| |
Collapse
|
15
|
The Methyltransferase Smyd1 Mediates LPS-Triggered Up-Regulation of IL-6 in Endothelial Cells. Cells 2021; 10:cells10123515. [PMID: 34944023 PMCID: PMC8700543 DOI: 10.3390/cells10123515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
The lysine methyltransferase Smyd1 with its characteristic catalytic SET-domain is highly enriched in the embryonic heart and skeletal muscles, participating in cardiomyogenesis, sarcomere assembly and chromatin remodeling. Recently, significant Smyd1 levels were discovered in endothelial cells (ECs) that responded to inflammatory cytokines. Based on these biochemical properties, we hypothesized that Smyd1 is involved in inflammation-triggered signaling in ECs and therefore, investigated its role within the LPS-induced signaling cascade. Human endothelial cells (HUVECs and EA.hy926 cells) responded to LPS stimulation with higher intrinsic Smyd1 expression. By transfection with expression vectors containing gene inserts encoding either intact Smyd1, a catalytically inactive Smyd1-mutant or Smyd1-specific siRNAs, we show that Smyd1 contributes to LPS-triggered expression and secretion of IL-6 in EA.hy926 cells. Further molecular analysis revealed this process to be based on two signaling pathways: Smyd1 increased the activity of NF-κB and promoted the trimethylation of lysine-4 of histone-3 (H3K4me3) within the IL-6 promoter, as shown by ChIP-RT-qPCR combined with IL-6-promoter-driven luciferase reporter gene assays. In summary, our experimental analysis revealed that LPS-binding to ECs leads to the up-regulation of Smyd1 expression to transduce the signal for IL-6 up-regulation via activation of the established NF-κB pathway as well as via epigenetic trimethylation of H3K4.
Collapse
|
16
|
Milanesi E, Dobre M, Cucos CA, Rojo AI, Jiménez-Villegas J, Capetillo-Zarate E, Matute C, Piñol-Ripoll G, Manda G, Cuadrado A. Whole Blood Expression Pattern of Inflammation and Redox Genes in Mild Alzheimer's Disease. J Inflamm Res 2021; 14:6085-6102. [PMID: 34848989 PMCID: PMC8612672 DOI: 10.2147/jir.s334337] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/29/2021] [Indexed: 12/21/2022] Open
Abstract
Background Although Alzheimer’s disease (AD) is associated with alterations of the central nervous system, this disease has an echo in blood that might represent a valuable source of biomarkers for improved diagnosis, prognosis and for monitoring drug response. Methods We performed a targeted transcriptomics study on 38 mild Alzheimer’s disease (AD) patients and 38 matched controls for evaluating the expression levels of 136 inflammation and 84 redox genes in whole blood. Patients were diagnosed as mild AD based on altered levels of total TAU, phospho-TAU and Abeta(1–42) in cerebrospinal fluid, and Abeta(1–40), Abeta(1–42) and total TAU levels in plasma. Whenever possible, blood and brain comparisons were made using public datasets. Results We found 48 inflammation and 34 redox genes differentially expressed in the blood of AD patients vs controls (FC >1.5, p < 0.01), out of which 22 pro-inflammatory and 12 redox genes exhibited FC >2 and p < 0.001. Receiver operating characteristic (ROC) analysis identified nine inflammation and seven redox genes that discriminated between AD patients and controls (area under the curve >0.9). Correlations of the dysregulated inflammation and redox transcripts indicated that RELA may regulate several redox genes including DUOX1 and GSR. Based on the gene expression profile, we have found that the master regulators of inflammation and redox homeostasis, NFκB and NRF2, were significantly disturbed in the blood of AD patients, as well as several zinc finger and helix-loop-helix transcription factors. Conclusion The selected inflammation and redox genes might be useful biomarkers for monitoring anti-inflammatory therapy in mild AD.
Collapse
Affiliation(s)
- Elena Milanesi
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Maria Dobre
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | | | - Ana I Rojo
- Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain.,Neuroscience Section, Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, 28046, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain
| | - José Jiménez-Villegas
- Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain
| | - Estibaliz Capetillo-Zarate
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Carlos Matute
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastons Cognitius, Hospital Universitari Santa Maria-IRB Leida, Lleida, 25198, Spain
| | - Gina Manda
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania
| | - Antonio Cuadrado
- "Victor Babes" National Institute of Pathology, Bucharest, 050096, Romania.,Department of Endocrine Physiology and Nervous System, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, 28029, Spain.,Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, 28049, Spain.,Neuroscience Section, Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, 28046, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, 28031, Spain
| |
Collapse
|
17
|
Romero N, Favoreel HW. Pseudorabies Virus Infection Triggers NF-κB Activation via the DNA Damage Response but Actively Inhibits NF-κB-Dependent Gene Expression. J Virol 2021; 95:e0166621. [PMID: 34613805 PMCID: PMC8610585 DOI: 10.1128/jvi.01666-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022] Open
Abstract
The nuclear factor kappa B (NF-κB) pathway is known to integrate signaling associated with very diverse intra- and extracellular stressors, including virus infections, and triggers a powerful (proinflammatory) response through the expression of NF-κB-regulated genes. Typically, the NF-κB pathway collects and transduces threatening signals at the cell surface or in the cytoplasm leading to nuclear import of activated NF-κB transcription factors. In the current work, we demonstrate that the swine alphaherpesvirus pseudorabies virus (PRV) induces a peculiar mode of NF-κB activation known as "inside-out" NF-κB activation. We show that PRV triggers the DNA damage response (DDR) and that this DDR response drives NF-κB activation since inhibition of the nuclear ataxia telangiectasia-mutated (ATM) kinase, a chief controller of DDR, abolished PRV-induced NF-κB activation. Initiation of the DDR-NF-κB signaling axis requires viral protein synthesis but occurs before active viral genome replication. In addition, the initiation of the DDR-NF-κB signaling axis is followed by a virus-induced complete shutoff of NF-κB-dependent gene expression that depends on viral DNA replication. In summary, the results presented in this study reveal that PRV infection triggers a noncanonical DDR-NF-κB activation signaling axis and that the virus actively inhibits the (potentially antiviral) consequences of this pathway, by inhibiting NF-κB-dependent gene expression. IMPORTANCE The NF-κB signaling pathway plays a critical role in coordination of innate immune responses that are of vital importance in the control of infections. The current report generates new insights into the interaction of the alphaherpesvirus pseudorabies virus (PRV) with the NF-κB pathway, as they reveal that (i) PRV infection leads to NF-κB activation via a peculiar "inside-out" nucleus-to-cytoplasm signal that is triggered via the DNA damage response (DDR), (ii) the DDR-NF-κB signaling axis requires expression of viral proteins but is initiated before active PRV replication, and (iii) late viral factor(s) allow PRV to actively and efficiently inhibit NF-κB-dependent (proinflammatory) gene expression. These data suggest that activation of the DDR-NF-κB during PRV infection is host driven and that its potential antiviral consequences are actively inhibited by the virus.
Collapse
Affiliation(s)
- Nicolás Romero
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Herman W. Favoreel
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
18
|
Chen W, Lu W, Wolynes PG, Komives E. Single-molecule conformational dynamics of a transcription factor reveals a continuum of binding modes controlling association and dissociation. Nucleic Acids Res 2021; 49:11211-11223. [PMID: 34614173 PMCID: PMC8565325 DOI: 10.1093/nar/gkab874] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Binding and unbinding of transcription factors to DNA are kinetically controlled to regulate the transcriptional outcome. Control of the release of the transcription factor NF-κB from DNA is achieved through accelerated dissociation by the inhibitor protein IκBα. Using single-molecule FRET, we observed a continuum of conformations of NF-κB in free and DNA-bound states interconverting on the subseconds to minutes timescale, comparable to in vivo binding on the seconds timescale, suggesting that structural dynamics directly control binding kinetics. Much of the DNA-bound NF-κB is partially bound, allowing IκBα invasion to facilitate DNA dissociation. IκBα induces a locked conformation where the DNA-binding domains of NF-κB are too far apart to bind DNA, whereas a loss-of-function IκBα mutant retains the NF-κB conformational ensemble. Overall, our results suggest a novel mechanism with a continuum of binding modes for controlling association and dissociation of transcription factors.
Collapse
Affiliation(s)
- Wei Chen
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, USA
| | - Wei Lu
- Center for Theoretical Biological Physics, Departments of Chemistry, Physics, and Biosciences, Rice University, Houston, Texas 77005, USA
| | - Peter G Wolynes
- Center for Theoretical Biological Physics, Departments of Chemistry, Physics, and Biosciences, Rice University, Houston, Texas 77005, USA
| | - Elizabeth A Komives
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, USA
| |
Collapse
|
19
|
Yang H, Wang Y, Jin S, Pang Q, Shan A, Feng X. Dietary resveratrol alleviated lipopolysaccharide-induced ileitis through Nrf2 and NF-κB signalling pathways in ducks (Anas platyrhynchos). J Anim Physiol Anim Nutr (Berl) 2021; 106:1306-1320. [PMID: 34729831 DOI: 10.1111/jpn.13657] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
Gram-negative bacteria contamination of feed can occur at all the stage of feed production, storage, transportation and utilization. Lipopolysaccharide (LPS) is a major toxic metabolite of Gram-negative bacteria. The aim of this study was to explore the effect of dietary resveratrol on the duck ileitis caused by LPS and its optimum addition level in diet. The results showed that LPS-induced duck ileitis with the destruction of intestinal structure, oxidative stress, mitochondrial dysfunction, inflammatory response and permeability alteration. Dietary resveratrol alleviated LPS-induced intestinal dysfunction and the increase of intestinal permeability by linearly increasing mRNA levels of tight junction protein genes (Claudin-1, Occludin-1, ZO-1) (p < 0.05) and protein expression of Claudin-1 (p < 0.01). In addition, dietary resveratrol improved the antioxidant capacity of duck ileum by reducing the production of MDA and increasing the activity of T-SOD (p < 0.01) and CAT. Lipopolysaccharide increased Keap1 at mRNA and protein level (p < 0.01) and decreased the protein level of Nrf2 (p < 0.05). Dietary resveratrol significantly downregulated expression of Keap1 and upregulated expression of Nrf2 in duck (p < 0.05). Dietary resveratrol suppressed the TLR4/NF-κB signalling pathway and the expression of its downstream genes including IKK, TXNIP, NLRP3, Caspase-1, IL-6 and IL-18. Meanwhile, the levels of inflammatory cytokines (IL-6, IL-18 and TNF-α) showed a linearly decrease (p < 0.01) with increasing dietary resveratrol level. These results demonstrated that resveratrol alleviated the LPS-induced acute ileitis of duck through Nrf2 and NF-κB signalling pathways, and the dietary resveratrol of 500 mg/kg is more efficiently.
Collapse
Affiliation(s)
- Hao Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| | - Yingjie Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| | - Sanjun Jin
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| | - Qian Pang
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| | - Xingjun Feng
- Institute of Animal Nutrition, Northeast Agricultural University, Xiangfang District, Harbin, China
| |
Collapse
|
20
|
Meier-Soelch J, Mayr-Buro C, Juli J, Leib L, Linne U, Dreute J, Papantonis A, Schmitz ML, Kracht M. Monitoring the Levels of Cellular NF-κB Activation States. Cancers (Basel) 2021; 13:5351. [PMID: 34771516 PMCID: PMC8582385 DOI: 10.3390/cancers13215351] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
The NF-κB signaling system plays an important regulatory role in the control of many biological processes. The activities of NF-κB signaling networks and the expression of their target genes are frequently elevated in pathophysiological situations including inflammation, infection, and cancer. In these conditions, the outcome of NF-κB activity can vary according to (i) differential activation states, (ii) the pattern of genomic recruitment of the NF-κB subunits, and (iii) cellular heterogeneity. Additionally, the cytosolic NF-κB activation steps leading to the liberation of DNA-binding dimers need to be distinguished from the less understood nuclear pathways that are ultimately responsible for NF-κB target gene specificity. This raises the need to more precisely determine the NF-κB activation status not only for the purpose of basic research, but also in (future) clinical applications. Here we review a compendium of different methods that have been developed to assess the NF-κB activation status in vitro and in vivo. We also discuss recent advances that allow the assessment of several NF-κB features simultaneously at the single cell level.
Collapse
Affiliation(s)
- Johanna Meier-Soelch
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Christin Mayr-Buro
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Jana Juli
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Lisa Leib
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| | - Uwe Linne
- Mass Spectrometry Facility of the Department of Chemistry, Philipps University, 35032 Marburg, Germany;
| | - Jan Dreute
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - M. Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University, 35392 Giessen, Germany;
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, 35392 Giessen, Germany; (J.M.-S.); (C.M.-B.); (J.J.); (L.L.)
| |
Collapse
|
21
|
Postler TS, Peng V, Bhatt DM, Ghosh S. Metformin selectively dampens the acute inflammatory response through an AMPK-dependent mechanism. Sci Rep 2021; 11:18721. [PMID: 34548527 PMCID: PMC8455559 DOI: 10.1038/s41598-021-97441-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/25/2021] [Indexed: 12/30/2022] Open
Abstract
Metformin is a first-line drug in the treatment of type-2 diabetes mellitus (T2DM). In addition to its antigluconeogenic and insulin-sensitizing properties, metformin has emerged as a potent inhibitor of the chronic inflammatory response of macrophages. In particular, metformin treatment has been shown to reduce expression of interleukin (IL-) 1β during long-term exposure to the pro-inflammatory stimulus lipopolysaccharide (LPS) through a reduction in reactive oxygen species (ROS), which decreases the levels of the hypoxia-inducible factor (HIF) 1-α, and through enhanced expression of IL-10. However, the effect of metformin on the acute inflammatory response, before significant levels of ROS accumulate in the cell, has not been explored. Here, we show that metformin alters the acute inflammatory response through its activation of AMP-activated protein kinase (AMPK), but independently of HIF1-α and IL-10, in primary macrophages and two macrophage-like cell lines. Thus, metformin changes the acute and the chronic inflammatory response through fundamentally distinct mechanisms. Furthermore, RNA-seq analysis reveals that metformin pretreatment affects the levels of a large yet selective subset of inflammatory genes, dampening the response to short-term LPS exposure and affecting a wide range of pathways and biological functions. Taken together, these findings reveal an unexpected complexity in the anti-inflammatory properties of this widely used drug.
Collapse
Affiliation(s)
- Thomas S Postler
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Vincent Peng
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dev M Bhatt
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Amgen Research Oncology and Inflammation, South San Francisco, CA, 94080, USA
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Shamekhi S, Abdolalizadeh J, Ostadrahimi A, Mohammadi SA, Barzegari A, Lotfi H, Bonabi E, Zarghami N. Apoptotic Effect of Saccharomyces cerevisiae on Human Colon Cancer SW480 Cells by Regulation of Akt/NF-ĸB Signaling Pathway. Probiotics Antimicrob Proteins 2021; 12:311-319. [PMID: 30788662 DOI: 10.1007/s12602-019-09528-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance is one of the major problems, which causes recurrence of cancers. Therefore, complementary treatments are needed to improve the impacts of chemotherapy agents. The effect of probiotics as cancer-preventing agents through involvement in the activation of apoptotic pathways has been established. The present study sought to investigate how the heat-killed form of Saccharomyces cerevisiae (as a probiotic) could affect the Akt/NF-kB-induced apoptosis in colon cancer cells, the SW480 cell line. The cytotoxic effects of heat-killed yeast (HKY) and 5-fluorouracil (5-FU, as a positive control drug) were assayed using the MTT method. Morphological changes followed by apoptosis were examined using DAPI staining. The transcription and translation level of apoptosis genes were explored with qRT-PCR and western blotting. The data were analyzed using GraphPad Prism V6.0 Software. The results showed that HKY could induce apoptosis in colon cancer cell line through downregulation of p-Akt1, Rel A, Bcl-XL, pro-caspase 3, and pro-caspase 9 expressions, and upregulation of BAX, cleaved caspase-3, and cleaved caspase-9. Besides, Akt protein expression was not affected. It is noticeable that HKY had a better modulating effect on BAX expression compared with 5-FU. It was able to modulate Akt/NF-kB signaling pathway followed by the apoptotic cascade.
Collapse
Affiliation(s)
- Sara Shamekhi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Abolghasem Mohammadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Plant Breeding and Biotechnology, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esat Bonabi
- Department of Medical Microbiology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - Nosratollah Zarghami
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Deciphering the mechanisms of CC-122 resistance in DLBCL via a genome-wide CRISPR screen. Blood Adv 2021; 5:2027-2039. [PMID: 33847741 DOI: 10.1182/bloodadvances.2020003431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
CC-122 is a next-generation cereblon E3 ligase-modulating agent that has demonstrated promising clinical efficacy in patients with relapsed or refractory diffuse large B-cell lymphoma (R/R DLBCL). Mechanistically, CC-122 induces the degradation of IKZF1/3, leading to T-cell activation and robust cell-autonomous killing in DLBCL. We report a genome-wide CRISPR/Cas9 screening for CC-122 in a DLBCL cell line SU-DHL-4 with follow-up mechanistic characterization in 6 DLBCL cell lines to identify genes regulating the response to CC-122. Top-ranked CC-122 resistance genes encode, not only well-defined members or regulators of the CUL4/DDB1/RBX1/CRBN E3 ubiquitin ligase complex, but also key components of signaling and transcriptional networks that have not been shown to modulate the response to cereblon modulators. Ablation of CYLD, NFKBIA, TRAF2, or TRAF3 induces hyperactivation of the canonical and/or noncanonical NF-κB pathways and subsequently diminishes CC-122-induced apoptosis in 5 of 6 DLBCL cell lines. Depletion of KCTD5, the substrate adaptor of the CUL3/RBX1/KCTD5 ubiquitin ligase complex, promotes the stabilization of its cognate substrate, GNG5, resulting in CC-122 resistance in HT, SU-DHL-4, and WSU-DLCL2. Furthermore, knockout of AMBRA1 renders resistance to CC-122 in SU-DHL-4 and U-2932, whereas knockout of RFX7 leads to resistance specifically in SU-DHL-4. The ubiquitous and cell line-specific mechanisms of CC-122 resistance in DLBCL cell lines revealed in this work pinpoint genetic alternations that are potentially associated with clinical resistance in patients and facilitate the development of biomarker strategies for patient stratification, which may improve clinical outcomes of patients with R/R DLBCL.
Collapse
|
24
|
Mirando AC, Lima e Silva R, Chu Z, Campochiaro PA, Pandey NB, Popel AS. Suppression of Ocular Vascular Inflammation through Peptide-Mediated Activation of Angiopoietin-Tie2 Signaling. Int J Mol Sci 2020; 21:ijms21145142. [PMID: 32708100 PMCID: PMC7404316 DOI: 10.3390/ijms21145142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
Persistent inflammation is a complication associated with many ocular diseases. Changes in ocular vessels can amplify disease responses and contribute to vision loss by influencing the delivery of leukocytes to the eye, vascular leakage, and perfusion. Here, we report the anti-inflammatory activity for AXT107, a non-RGD, 20-mer αvβ3 and α5β1 integrin-binding peptide that blocks vascular endothelial growth factor (VEGF)-signaling and activates tyrosine kinase with immunoglobulin and EGF-like domains 2 (Tie2) using the normally inhibitory ligand angiopoietin 2 (Ang2). Tumor necrosis factor α (TNFα), a central inflammation mediator, induces Ang2 release from endothelial cells to enhance its stimulation of inflammation and vascular leakage. AXT107 resolves TNFα-induced vascular inflammation in endothelial cells by converting the endogenously released Ang2 into an agonist of Tie2 signaling, thereby disrupting both the synergism between TNFα and Ang2 while also preventing inhibitor of nuclear factor-κB α (IκBα) degradation directly through Tie2 signaling. This recovery of IκBα prevents nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear localization, thereby blocking NF-κB-induced inflammatory responses, including the production of VCAM-1 and ICAM-1, leukostasis, and vascular leakage in cell and mouse models. AXT107 also decreased the levels of pro-inflammatory TNF receptor 1 (TNFR1) without affecting levels of the more protective TNFR2. These data suggest that AXT107 may provide multiple benefits in the treatment of retinal/choroidal and other vascular diseases by suppressing inflammation and promoting vascular stabilization.
Collapse
MESH Headings
- Angiopoietin-1/metabolism
- Angiopoietin-2/metabolism
- Animals
- Capillary Permeability/drug effects
- Choroid Diseases/drug therapy
- Collagen Type IV/pharmacology
- Collagen Type IV/therapeutic use
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Human Umbilical Vein Endothelial Cells
- Humans
- I-kappa B Kinase/metabolism
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Leukostasis/drug therapy
- Leukostasis/metabolism
- Mice
- Mice, Inbred C57BL
- Peptide Fragments/pharmacology
- Peptide Fragments/therapeutic use
- Receptor, TIE-2/agonists
- Receptor, TIE-2/metabolism
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Retinal Diseases/drug therapy
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/pharmacology
- Vascular Cell Adhesion Molecule-1/metabolism
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Adam C. Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
| | - Raquel Lima e Silva
- Department of Ophthalmology and the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.L.e.S.); (P.A.C.)
| | - Zenny Chu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
| | - Peter A. Campochiaro
- Department of Ophthalmology and the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (R.L.e.S.); (P.A.C.)
| | - Niranjan B. Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
- AsclepiX Therapeutics, Inc., Baltimore, MD 21211, USA
- Correspondence: (N.B.P.); (A.S.P.); Tel.: +908-328-2019 (N.B.P.); +410-955-6419 (A.S.P.)
| | - Aleksander S. Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (A.C.M.); (Z.C.)
- Correspondence: (N.B.P.); (A.S.P.); Tel.: +908-328-2019 (N.B.P.); +410-955-6419 (A.S.P.)
| |
Collapse
|
25
|
Recent insights of T cell receptor-mediated signaling pathways for T cell activation and development. Exp Mol Med 2020; 52:750-761. [PMID: 32439954 PMCID: PMC7272404 DOI: 10.1038/s12276-020-0435-8] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022] Open
Abstract
T cell activation requires extracellular stimulatory signals that are mainly mediated by T cell receptor (TCR) complexes. The TCR recognizes antigens on major histocompatibility complex molecules with the cooperation of CD4 or CD8 coreceptors. After recognition, TCR-induced signaling cascades that propagate signals via various molecules and second messengers are induced. Consequently, many features of T cell-mediated immune responses are determined by these intracellular signaling cascades. Furthermore, differences in the magnitude of TCR signaling direct T cells toward distinct effector linages. Therefore, stringent regulation of T cell activation is crucial for T cell homeostasis and proper immune responses. Dysregulation of TCR signaling can result in anergy or autoimmunity. In this review, we summarize current knowledge on the pathways that govern how the TCR complex transmits signals into cells and the roles of effector molecules that are involved in these pathways.
Collapse
|
26
|
Lu J, Lu L, Yu Y, Baranowski J, Claud EC. Maternal administration of probiotics promotes brain development and protects offspring's brain from postnatal inflammatory insults in C57/BL6J mice. Sci Rep 2020; 10:8178. [PMID: 32424168 PMCID: PMC7235088 DOI: 10.1038/s41598-020-65180-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Neonatal morbidities are associated with long term neurological deficits in life and have also been associated with dysbiosis. We tested whether optimizing the neonate's microbiome through maternal probiotic supplementation can improve offspring's neurodevelopmental outcomes. Maternal LB supplementation, carried out by giving Lactobacillus acidophilus and Bifidobacterium infantis (LB) to pregnant C57/BL6J mice daily from E16 to weaning, significantly suppressed postnatal peripheral proinflammatory insult-induced systemic inflammation and normalized compromised blood-brain barrier permeability and tight junction protein expression in the offspring at pre-weaned age. Maternal LB exposure also regulated markers associated with leukocyte transendothelial migration, extracellular matrix injury and neuroinflammation. The suppressed neuroinflammation by maternal LB supplementation was associated with reduced astrocyte/microglia activation and downregulation of the transcriptional regulators CEBPD and IκBα. Furthermore, maternal LB supplementation promoted neuronal and oligodendrocyte progenitor cell development. Our study demonstrates the efficacy of maternal LB supplementation in modulating systemic and central nervous system inflammation as well as promoting neural/oligodendrocyte progenitor development in the offspring. This evidence suggests that maternal probiotic supplementation may be a safe and effective strategy to improve neurological outcomes in the offspring.
Collapse
Affiliation(s)
- Jing Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Lei Lu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Yueyue Yu
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA
| | - Jillian Baranowski
- The University of Chicago, Pritzker School of Medicine, Chicago, IL, 60637, USA
| | - Erika C Claud
- The University of Chicago, Pritzker School of Medicine, Department of Pediatrics, Chicago, IL, 60637, USA.
| |
Collapse
|
27
|
Romero N, Van Waesberghe C, Favoreel HW. Pseudorabies Virus Infection of Epithelial Cells Leads to Persistent but Aberrant Activation of the NF-κB Pathway, Inhibiting Hallmark NF-κB-Induced Proinflammatory Gene Expression. J Virol 2020; 94:e00196-20. [PMID: 32132236 PMCID: PMC7199412 DOI: 10.1128/jvi.00196-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/27/2020] [Indexed: 02/03/2023] Open
Abstract
The nuclear factor kappa B (NF-κB) is a potent transcription factor, activation of which typically results in robust proinflammatory signaling and triggering of fast negative feedback modulators to avoid excessive inflammatory responses. Here, we report that infection of epithelial cells, including primary porcine respiratory epithelial cells, with the porcine alphaherpesvirus pseudorabies virus (PRV) results in the gradual and persistent activation of NF-κB, illustrated by proteasome-dependent degradation of the inhibitory NF-κB regulator IκB and nuclear translocation and phosphorylation of the NF-κB subunit p65. PRV-induced persistent activation of NF-κB does not result in expression of negative feedback loop genes, like the gene for IκBα or A20, and does not trigger expression of prototypical proinflammatory genes, like the gene for tumor necrosis factor alpha (TNF-α) or interleukin-6 (IL-6). In addition, PRV infection inhibits TNF-α-induced canonical NF-κB activation. Hence, PRV infection triggers persistent NF-κB activation in an unorthodox way and dramatically modulates the NF-κB signaling axis, preventing typical proinflammatory gene expression and the responsiveness of cells to canonical NF-κB signaling, which may aid the virus in modulating early proinflammatory responses in the infected host.IMPORTANCE The NF-κB transcription factor is activated via different key inflammatory pathways and typically results in the fast expression of several proinflammatory genes as well as negative feedback loop genes to prevent excessive inflammation. In the current report, we describe that infection of cells with the porcine alphaherpesvirus pseudorabies virus (PRV) triggers a gradual and persistent aberrant activation of NF-κB, which does not result in expression of hallmark proinflammatory or negative feedback loop genes. In addition, although PRV-induced NF-κB activation shares some mechanistic features with canonical NF-κB activation, it also shows remarkable differences; e.g., it is largely independent of the canonical IκB kinase (IKK) and even renders infected cells resistant to canonical NF-κB activation by the inflammatory cytokine TNF-α. Aberrant PRV-induced NF-κB activation may therefore paradoxically serve as a viral immune evasion strategy and may represent an important tool to unravel currently unknown mechanisms and consequences of NF-κB activation.
Collapse
Affiliation(s)
- Nicolás Romero
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Cliff Van Waesberghe
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Herman W Favoreel
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
28
|
Arantes MR, Peijnenburg A, Hendriksen PJM, Stoopen G, Almeida TS, Souza TM, Farias DF, Carvalho AFU, Rocha TM, Leal LKAM, Vasconcelos IM, Oliveira JTA. In vitro toxicological characterisation of the antifungal compound soybean toxin (SBTX). Toxicol In Vitro 2020; 65:104824. [PMID: 32165152 DOI: 10.1016/j.tiv.2020.104824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/13/2020] [Accepted: 03/08/2020] [Indexed: 12/18/2022]
Abstract
Soybean toxin (SBTX) is a protein isolated from soybean seeds and composed of two polypeptide subunits (17 and 27 kDa). SBTX has in vitro activity against phytopathogenic fungi such as Cercospora sojina, Aspergillus niger, and Penicillium herguei, and yeasts like Candida albicans, C. parapsilosis, Kluyveromyces marxiannus, and Pichia membranifaciens. The present study aimed to analyze in vitro whether SBTX causes any side effects on non-target bacterial and mammalian cells that could impede its potential use as a novel antifungal agent. SBTX at 100 μg/mL and 200 μg/mL did not hinder the growth of the bacteria Salmonella enterica (subspecies enterica serovar choleraesuis), Bacillus subtilis (subspecies spizizenii) and Staphylococcus aureus. Moreover, SBTX at concentrations up to 500 μg/mL did not significantly affect the viability of erythrocytes, neutrophils, and human intestinal Caco-2 cells. To study whether SBTX could induce relevant alterations in gene expression, in vitro DNA microarray experiments were conducted in which differentiated Caco-2 cells were exposed for 24 h to 100 μg/mL or 200 μg/mL SBTX. SBTX up-regulated genes involved in cell cycle and immune response pathways, but down-regulated genes that play a role in cholesterol biosynthesis and platelet degranulation pathways. Thus, although SBTX did not affect bacteria, nor induced cytotoxity in mammalian cells, it affected some biological pathways in the human Caco-2 cell line that warrants further investigation.
Collapse
Affiliation(s)
- Mariana Reis Arantes
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil
| | - Ad Peijnenburg
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Peter J M Hendriksen
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Geert Stoopen
- WFSR, Wageningen University and Research Centre, P.O. Box 230, 6700 AE Wageningen, the Netherlands.
| | - Thiago Silva Almeida
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil
| | - Terezinha Maria Souza
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229, ER, the Netherlands.
| | - Davi Felipe Farias
- Department of Molecular Biology, Federal University of Paraíba, 58051-900 Joao Pessoa, PB, Brazil.
| | | | | | | | - Ilka Maria Vasconcelos
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil.
| | - Jose Tadeu Abreu Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Ceara, 60020-181 Fortaleza, CE, Brazil.
| |
Collapse
|
29
|
Regulation of the linear ubiquitination of STAT1 controls antiviral interferon signaling. Nat Commun 2020; 11:1146. [PMID: 32123171 PMCID: PMC7052135 DOI: 10.1038/s41467-020-14948-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Linear ubiquitination is a critical regulator of inflammatory signaling pathways. However, linearly ubiquitinated substrates and the biological significance of linear ubiquitination is incompletely understood. Here, we show that STAT1 has linear ubiquitination at Lys511 and Lys652 residues in intact cells, which inhibits STAT1 binding to the type-I interferon receptor IFNAR2, thereby restricting STAT1 activation and resulting in type-I interferon signaling homeostasis. Linear ubiquitination of STAT1 is removed rapidly by OTULIN upon type-I interferon stimulation, which facilitates activation of interferon-STAT1 signaling. Furthermore, viruses induce HOIP expression through the NF-κB pathway, which in turn increases linear ubiquitination of STAT1 and thereby inhibits interferon antiviral response. Consequently, HOIL-1L heterozygous mice have active STAT1 signaling and enhanced responses to type-I interferons. These findings demonstrate a linear ubiquitination-mediated switch between homeostasis and activation of type-I interferon signaling, and suggest potential strategies for clinical antiviral therapy.
Collapse
|
30
|
Watson CJF, Maguire ARR, Rouillard MM, Crozier RWE, Yousef M, Bruton KM, Fajardo VA, MacNeil AJ. TAK1 signaling activity links the mast cell cytokine response and degranulation in allergic inflammation. J Leukoc Biol 2020; 107:649-661. [PMID: 32108376 DOI: 10.1002/jlb.2a0220-401rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 12/23/2022] Open
Abstract
Mast cells drive the inappropriate immune response characteristic of allergic inflammatory disorders via release of pro-inflammatory mediators in response to environmental cues detected by the IgE-FcεRI complex. The role of TGF-β-activated kinase 1 (TAK1), a participant in related signaling in other contexts, remains unknown in allergy. We detect novel activation of TAK1 at Ser412 in response to IgE-mediated activation under SCF-c-kit potentiation in a mast cell-driven response characteristic of allergic inflammation, which is potently blocked by TAK1 inhibitor 5Z-7-oxozeaenol (OZ). We, therefore, interrogated the role of TAK1 in a series of mast cell-mediated responses using IgE-sensitized murine bone marrow-derived mast cells, stimulated with allergen under several TAK1 inhibition strategies. TAK1 inhibition by OZ resulted in significant impairment in the phosphorylation of MAPKs p38, ERK, and JNK; and mediation of the NF-κB pathway via IκBα. Impaired gene expression and near abrogation in release of pro-inflammatory cytokines TNF, IL-6, IL-13, and chemokines CCL1, and CCL2 was detected. Finally, a significant inhibition of mast cell degranulation, accompanied by an impairment in calcium mobilization, was observed in TAK1-inhibited cells. These results suggest that TAK1 acts as a signaling node, not only linking the MAPK and NF-κB pathways in driving the late-phase response, but also initiation of the degranulation mechanism of the mast cell early-phase response following allergen recognition and may warrant consideration in future therapeutic development.
Collapse
Affiliation(s)
- Colton J F Watson
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Aindriu R R Maguire
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Melissa M Rouillard
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Robert W E Crozier
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Michael Yousef
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Kelly M Bruton
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| |
Collapse
|
31
|
Pyrocatechol, a component of coffee, suppresses LPS-induced inflammatory responses by inhibiting NF-κB and activating Nrf2. Sci Rep 2020; 10:2584. [PMID: 32054966 PMCID: PMC7018815 DOI: 10.1038/s41598-020-59380-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022] Open
Abstract
Coffee is a complex mixture of many bioactive compounds possessing anti-inflammatory properties. However, the mechanisms by which coffee exerts anti-inflammatory effects remains unclear and the active ingredients have not yet been identified. In this study, we found that coffee extract at more than 2.5%(v/v) significantly inhibited LPS-induced inflammatory responses in RAW264.7 cells and that anti-inflammatory activity of coffee required the roasting process. Interestingly, we identified pyrocatechol, a degradation product derived from chlorogenic acid during roasting, as the active ingredient exhibiting anti-inflammatory activity in coffee. HPLC analysis showed that 124 μM pyrocatechol was included in 100% (v/v) roasted coffee. A treatment with 5%(v/v) coffee extract and more than 2.5 μM pyrocatechol inhibited the LPS-induced activation of NF-κB and also significantly activated Nrf2, which acts as a negative regulator in LPS-induced inflammation. Furthermore, intake of 60% (v/v) coffee extract and 74.4 μM pyrocatechol, which is the concentration equal to contained in 60% (v/v) coffee, markedly inhibited the LPS-induced inflammatory responses in mice. Collectively, these results demonstrated that pyrocatechol, which was formed by the roasting of coffee green beans, is one of the ingredients contributing to the anti-inflammatory activity of coffee.
Collapse
|
32
|
Chronic activation of endothelial MAPK disrupts hematopoiesis via NFKB dependent inflammatory stress reversible by SCGF. Nat Commun 2020; 11:666. [PMID: 32015345 PMCID: PMC6997369 DOI: 10.1038/s41467-020-14478-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 01/13/2020] [Indexed: 02/08/2023] Open
Abstract
Inflammatory signals arising from the microenvironment have emerged as critical regulators of hematopoietic stem cell (HSC) function during diverse processes including embryonic development, infectious diseases, and myelosuppressive injuries caused by irradiation and chemotherapy. However, the contributions of cellular subsets within the microenvironment that elicit niche-driven inflammation remain poorly understood. Here, we identify endothelial cells as a crucial component in driving bone marrow (BM) inflammation and HSC dysfunction observed following myelosuppression. We demonstrate that sustained activation of endothelial MAPK causes NF-κB-dependent inflammatory stress response within the BM, leading to significant HSC dysfunction including loss of engraftment ability and a myeloid-biased output. These phenotypes are resolved upon inhibition of endothelial NF-κB signaling. We identify SCGF as a niche-derived factor that suppresses BM inflammation and enhances hematopoietic recovery following myelosuppression. Our findings demonstrate that chronic endothelial inflammation adversely impacts niche activity and HSC function which is reversible upon suppression of inflammation.
Collapse
|
33
|
Clusterin Silencing in Prostate Cancer Induces Matrix Metalloproteinases by an NF- κB-Dependent Mechanism. JOURNAL OF ONCOLOGY 2019; 2019:4081624. [PMID: 31885575 PMCID: PMC6925831 DOI: 10.1155/2019/4081624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/31/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
Abstract
Clusterin (CLU) is a stress-activated glycoprotein, whose expression is altered both in inflammation and cancer. Previously, we showed that abrogation of CLU expression in cancer-prone mice (TRAMP) results in the enhancement of tumor spreading and homing, concomitant with an enhanced expression of NF-κB. In the present paper, we carried out an extensive experimental work by utilizing microarray gene expression data, as well as in vitro and in vivo models of prostate cancer (PCa). Our results demonstrated that (i) CLU expression is significantly downregulated in human PCa and inversely correlates with the expression of p65 in metastases; (ii) CLU overexpression in PCa cells reduces the Ser536 phosphorylation of p65, inhibits NF-κB nuclear translocation, and reduces the transcription of matrix metalloproteinase-9 and metalloproteinase-2 (MMP-9 and MMP-2). Conversely, CLU silencing promotes NF-κB activation and transcriptional upregulation of MMP-9; and (iii) expression and activity of MMP-2 and MMP-9 are increased in CLU−/− mice (CLUKO) and in TRAMP/CLUKO mice in comparison to their relative Clu+/+ littermates. Taken together, our data support the hypothesis that CLU downregulation, an early and relevant event in PCa onset, may inhibit NF-κB activation and limit the execution of a transcriptional program that favor the disease progression towards a metastatic stage.
Collapse
|
34
|
Zhang Y, Xie H, Tang W, Zeng X, Lin Y, Xu L, Xiao L, Xu J, Wu Z, Yuan D. Trichostatin A, a Histone Deacetylase Inhibitor, Alleviates Eosinophilic Meningitis Induced by Angiostrongylus cantonensis Infection in Mice. Front Microbiol 2019; 10:2280. [PMID: 31636619 PMCID: PMC6787401 DOI: 10.3389/fmicb.2019.02280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylase inhibitor (HDACi) has been used in the treatment of neurodegenerative or autoimmune diseases. Angiostrongyliasis cantonensis caused by Angiostrongylus cantonensis infection is an emerging zoonosis of human eosinophilic meningitis or meningoencephalitis. Progressive neuronal apoptosis is the pathological basis of behavioral dysfunctions in angiostrongyliasis cantonensis. Neurological defects after anthelmintic treatment for angiostrongyliasis cantonensis are still common. In this study, we examined the effects of trichostatin A (TSA), a HDACi, on eosinophilic meningitis induced by A. cantonensis in mice. Intragastric administration of TSA significantly ameliorated brain injury and decreased cognitive impairments in mice at 15 days post-infection. TSA administration effectively reduced the inflammatory factor levels of iNOS, TNF-α, IL-5, IL-6, and IL-13 in infected mice. TSA treatment counteracted apoptosis with reduced expression levels of cleaved caspase-3, -4, -6, and RIP3 in A. cantonensis infected mice. In addition, TSA administration reduced total HDAC activity and increased the acetylation of histone H3 and H4 in the brain tissue of infected mice. The underlying mechanism of TSA on eosinophilic meningitis might be associated with decreased NF-κB p65 nuclear accumulation by inhibiting IκB phosphorylation. Furthermore, a co-expressive network of NF-κB p65 with 22 other genes was constructed according to our previous transcriptomic data in infected mice. We identified the correlations in the gene expression of NF-κB p65 with Lrp10, Il12rb1, Nfkbia, Ube2n, and Ube2d1 in infected mice after TSA administration. Thus, TSA has a protective effect on the progression of eosinophilic meningitis induced by A. cantonensis in mice.
Collapse
Affiliation(s)
- Yanhua Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Hui Xie
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Wenyan Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingda Zeng
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Yu Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lian Xu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Lihua Xiao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jun Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Tropical Diseases Control (SYSU), Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-Vectors Control, Guangzhou, China
| | - Dongjuan Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
35
|
Zhang H, Wu ZM, Yang YP, Shaukat A, Yang J, Guo YF, Zhang T, Zhu XY, Qiu JX, Deng GZ, Shi DM. Catalpol ameliorates LPS-induced endometritis by inhibiting inflammation and TLR4/NF-κB signaling. J Zhejiang Univ Sci B 2019; 20:816-827. [PMID: 31489801 PMCID: PMC6751487 DOI: 10.1631/jzus.b1900071] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Catalpol is the main active ingredient of an extract from Radix rehmanniae, which in a previous study showed a protective effect against various types of tissue injury. However, a protective effect of catalpol on uterine inflammation has not been reported. In this study, to investigate the protective mechanism of catalpol on lipopolysaccharide (LPS)-induced bovine endometrial epithelial cells (bEECs) and mouse endometritis, in vitro and in vivo inflammation models were established. The Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling pathway and its downstream inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qRT-PCR), western blot (WB), and immunofluorescence techniques. The results from ELISA and qRT-PCR showed that catalpol dose-dependently reduced the expression of pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α), interleukin (IL)-1β, and IL-6, and chemokines such as C-X-C motif chemokine ligand 8 (CXCL8) and CXCL5, both in bEECs and in uterine tissue. From the experimental results of WB, qRT-PCR, and immunofluorescence, the expression of TLR4 and the phosphorylation of NF-κB p65 were markedly inhibited by catalpol compared with the LPS group. The inflammatory damage to the mouse uterus caused by LPS was greatly reduced and was accompanied by a decline in myeloperoxidase (MPO) activity. The results of this study suggest that catalpol can exert an anti-inflammatory impact on LPS-induced bEECs and mouse endometritis by inhibiting inflammation and activation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Department of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China
| | - Zhi-min Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ya-ping Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ying-fang Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xin-ying Zhu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jin-xia Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Gan-zhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- †E-mail:
| | - Dong-mei Shi
- Department of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China
- †E-mail:
| |
Collapse
|
36
|
LaCanna R, Liccardo D, Zhang P, Tragesser L, Wang Y, Cao T, Chapman HA, Morrisey EE, Shen H, Koch WJ, Kosmider B, Wolfson MR, Tian Y. Yap/Taz regulate alveolar regeneration and resolution of lung inflammation. J Clin Invest 2019; 129:2107-2122. [PMID: 30985294 DOI: 10.1172/jci125014] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/05/2019] [Indexed: 12/12/2022] Open
Abstract
Alveolar epithelium plays a pivotal role in protecting the lungs from inhaled infectious agents. Therefore, the regenerative capacity of the alveolar epithelium is critical for recovery from these insults in order to rebuild the epithelial barrier and restore pulmonary functions. Here, we show that sublethal infection of mice with Streptococcus pneumoniae, the most common pathogen of community-acquired pneumonia, led to exclusive damage in lung alveoli, followed by alveolar epithelial regeneration and resolution of lung inflammation. We show that surfactant protein C-expressing (SPC-expressing) alveolar epithelial type II cells (AECIIs) underwent proliferation and differentiation after infection, which contributed to the newly formed alveolar epithelium. This increase in AECII activities was correlated with increased nuclear expression of Yap and Taz, the mediators of the Hippo pathway. Mice that lacked Yap/Taz in AECIIs exhibited prolonged inflammatory responses in the lung and were delayed in alveolar epithelial regeneration during bacterial pneumonia. This impaired alveolar epithelial regeneration was paralleled by a failure to upregulate IκBa, the molecule that terminates NF-κB-mediated inflammatory responses. These results demonstrate that signals governing resolution of lung inflammation were altered in Yap/Taz mutant mice, which prevented the development of a proper regenerative niche, delaying repair and regeneration of alveolar epithelium during bacterial pneumonia.
Collapse
Affiliation(s)
- Ryan LaCanna
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniela Liccardo
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Peggy Zhang
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Tragesser
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yan Wang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tongtong Cao
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Harold A Chapman
- Department of Medicine, Cardiovascular Research Institute, UCSF, San Francisco, California, USA
| | - Edward E Morrisey
- Department of Medicine, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hao Shen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Walter J Koch
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Beata Kosmider
- Department of Physiology, Department of Thoracic Medicine and Surgery, Center for Inflammation, Translational and Clinical Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Marla R Wolfson
- Department of Physiology, Department of Thoracic Medicine and Surgery, Center for Inflammation, Translational and Clinical Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ying Tian
- Department of Pharmacology, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
37
|
Kitamura K, Isoda K, Akita K, Miyosawa K, Kadoguchi T, Shimada K, Daida H. Lack of IκBNS promotes cholate-containing high-fat diet-induced inflammation and atherogenesis in low-density lipoprotein (LDL) receptor-deficient mice. IJC HEART & VASCULATURE 2019; 23:100344. [PMID: 30976650 PMCID: PMC6439265 DOI: 10.1016/j.ijcha.2019.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 11/21/2022]
Abstract
Background IκBNS, a nuclear IκB protein, regulates a subset of Toll-like receptor (TLR) dependent genes. A cholate-containing high-fat diet (HFD(CA(+))) induces TLR4 mediated early inflammatory response. The present study aims to clarify that the lack of IκBNS promotes atherogenesis in low-density lipoprotein receptor-deficient (LDLr−/−) mice fed HFD(CA(+)) compared with those fed a cholate-free HFD (HFD(CA(−))). Methods and results Mice that lacked IκBNS (IκBNS−/−) were crossed with LDLr−/− mice and formation of atherosclerotic lesions was analyzed after 6-week consumption of HFD(CA(+)) or HFD(CA(−)). IκBNS−/−/LDLr−/− mice fed HFD(CA(+)) (IκBNS−/−/LDLr−/−(CA(+))) showed a 3.5-fold increase of atherosclerotic lesion size in the aorta compared with LDLr−/−(CA(+)) mice (p < 0.01), whereas there was no difference between LDLr−/−(CA(−)) and IκBNS−/−/LDLr−/−(CA(−)) mice. Immunohistochemical analysis of the aortic root revealed that HFD(CA(+)) significantly increased Mac-3 (macrophage)-positive area by 1.5-fold (p < 0.01) and TLR4, interleukin-6 (IL-6) expression by 1.7-fold (p < 0.05) and 1.5-fold (p < 0.05), respectively, in IκBNS−/−/LDLr−/−(CA(+)) compared with LDLr-/-−−(CA(+)) mice. Furthermore, active STAT3 (pSTAT3)-positive cells were significantly increased by 1.7-fold in the lesions of IκBNS−/−/LDLr−/−(CA(+)) compared with LDLr−/−(CA(+)) mice (p < 0.01). These findings suggest that IκBNS deficiency and HFD(CA(+)) promote atherogenesis in LDLr−/− mice via TLR4/IL-6/STAT3 pathway. Finally, we showed that the monocytes from peripheral blood of IκBNS−/−/LDLr−/−(CA(+)) mice were found to contain the highest proportion of Ly6Chi monocytes among the four groups, suggesting that lack of IκBNS enhanced inflammation in response to HFD(CA(+)) feeding. Conclusions The present study is the first to demonstrate that the activation of innate immune system using HFD(CA(+)) induced significant inflammation and atherogenesis in IκBNS−/−/LDLr−/− compared with LDLr−/− mice.
Collapse
Affiliation(s)
| | - Kikuo Isoda
- Corresponding author at: Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | | | | | | | |
Collapse
|
38
|
Kim Y, Lee J, Lee D, Othmer HG. Synergistic Effects of Bortezomib-OV Therapy and Anti-Invasive Strategies in Glioblastoma: A Mathematical Model. Cancers (Basel) 2019; 11:E215. [PMID: 30781871 PMCID: PMC6406513 DOI: 10.3390/cancers11020215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/18/2022] Open
Abstract
It is well-known that the tumor microenvironment (TME) plays an important role in the regulation of tumor growth and the efficacy of anti-tumor therapies. Recent studies have demonstrated the potential of combination therapies, using oncolytic viruses (OVs) in conjunction with proteosome inhibitors for the treatment of glioblastoma, but the role of the TME in such therapies has not been studied. In this paper, we develop a mathematical model for combination therapies based on the proteosome inhibitor bortezomib and the oncolytic herpes simplex virus (oHSV), with the goal of understanding their roles in bortezomib-induced endoplasmic reticulum (ER) stress, and how the balance between apoptosis and necroptosis is affected by the treatment protocol. We show that the TME plays a significant role in anti-tumor efficacy in OV combination therapy, and illustrate the effect of different spatial patterns of OV injection. The results illustrate a possible phenotypic switch within tumor populations in a given microenvironment, and suggest new anti-invasion therapies.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Korea.
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
39
|
Nelson RH, Nelson DE. Signal Distortion: How Intracellular Pathogens Alter Host Cell Fate by Modulating NF-κB Dynamics. Front Immunol 2018; 9:2962. [PMID: 30619320 PMCID: PMC6302744 DOI: 10.3389/fimmu.2018.02962] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 12/03/2018] [Indexed: 01/17/2023] Open
Abstract
By uncovering complex dynamics in the expression or localization of transcriptional regulators in single cells that were otherwise hidden at the population level, live cell imaging has transformed our understanding of how cells sense and orchestrate appropriate responses to changes in their internal state or extracellular environment. This has proved particularly true for the nuclear factor-kappaB (NF-κB) family of transcription factors, key regulators of the inflammatory response and innate immune function, which are capable of encoding information about the mode and intensity of stimuli in the dynamics of NF-κB nuclear accumulation and loss. While live cell imaging continues to serve as a useful tool in ongoing efforts to characterize the feedbacks that shape these dynamics and to connect dynamics to downstream gene expression, it is also proving invaluable for recent studies that seek to determine how intracellular pathogens subvert NF-κB signaling to survive and replicate within host cells by providing quantitative information about the pathogen and changes in NF-κB activity during different stages of an infection. Here, we provide a brief overview of NF-κB signaling in innate immune cells and review recent literature that uses live imaging to investigate the mechanisms by which bacterial and yeast pathogens modulate NF-κB in a variety of different host cell types to evade destruction or maintain the viability of an intracellular growth niche.
Collapse
Affiliation(s)
- Rachel H Nelson
- Cellular Generation and Phenotyping Core Facility, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - David E Nelson
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
40
|
Krajewski D, Kaczenski E, Rovatti J, Polukort S, Thompson C, Dollard C, Ser-Dolansky J, Schneider SS, Kinney SRM, Mathias CB. Epigenetic Regulation via Altered Histone Acetylation Results in Suppression of Mast Cell Function and Mast Cell-Mediated Food Allergic Responses. Front Immunol 2018; 9:2414. [PMID: 30405614 PMCID: PMC6206211 DOI: 10.3389/fimmu.2018.02414] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/28/2018] [Indexed: 01/08/2023] Open
Abstract
Mast cells are highly versatile cells that perform a variety of functions depending on the immune trigger, context of activation, and cytokine stimulus. Antigen-mediated mast cell responses are regulated by transcriptional processes that result in the induction of numerous genes contributing to mast cell function. Recently, we also showed that exposure to dietary agents with known epigenetic actions such as curcumin can suppress mast cell-mediated food allergy, suggesting that mast cell responses in vivo may be epigenetically regulated. To further assess the effects of epigenetic modifications on mast cell function, we examined the behavior of bone marrow-derived mast cells (BMMCs) in response to trichostatin A (TSA) treatment, a well-studied histone deacetylase inhibitor. IgE-mediated BMMC activation resulted in enhanced expression and secretion of IL-4, IL-6, TNF-α, and IL-13. In contrast, pretreatment with TSA resulted in altered cytokine secretion. This was accompanied by decreased expression of FcεRI and mast cell degranulation. Interestingly, exposure to non-IgE stimuli such as IL-33, was also affected by TSA treatment. Furthermore, continuous TSA exposure contributed to mast cell apoptosis and a decrease in survival. Further examination revealed an increase in I-κBα and a decrease in phospho-relA levels in TSA-treated BMMCs, suggesting that TSA alters transcriptional processes, resulting in enhancement of I-κBα transcription and decreased NF-κB activation. Lastly, treatment of wild-type mice with TSA in a model of ovalbumin-induced food allergy resulted in a significant attenuation in the development of food allergy symptoms including decreases in allergic diarrhea and mast cell activation. These data therefore suggest that the epigenetic regulation of mast cell activation during immune responses may occur via altered histone acetylation, and that exposure to dietary substances may induce epigenetic modifications that modulate mast cell function.
Collapse
Affiliation(s)
- Dylan Krajewski
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Edwin Kaczenski
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Jeffrey Rovatti
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Stephanie Polukort
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Chelsea Thompson
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Catherine Dollard
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States.,Northampton High School, Northampton, MA, United States
| | - Jennifer Ser-Dolansky
- Baystate Medical Center, Pioneer Valley Life Sciences Institute, Springfield, MA, United States
| | - Sallie S Schneider
- Baystate Medical Center, Pioneer Valley Life Sciences Institute, Springfield, MA, United States
| | - Shannon R M Kinney
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Clinton B Mathias
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| |
Collapse
|
41
|
Activation of NF-κB driven inflammatory programs in mesenchymal elements attenuates hematopoiesis in low-risk myelodysplastic syndromes. Leukemia 2018; 33:536-541. [PMID: 30315231 PMCID: PMC6365382 DOI: 10.1038/s41375-018-0267-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/23/2018] [Accepted: 08/28/2018] [Indexed: 01/19/2023]
Abstract
Activation of NF-κB signaling in mesenchymal cells is common in LR-MDS. Activation of NF-κB in mesenchymal cells leads to transcriptional overexpression of inflammatory factors including negative regulators of hematopoiesis. Activation of NF-κB attenuates HSPC numbers and function ex vivo.
Collapse
|
42
|
Li XH, Gaynor RB. Regulation of NF-kappaB by the HTLV-1 Tax protein. Gene Expr 2018; 7:233-45. [PMID: 10440224 PMCID: PMC6174672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
The Tax protein encoded by the human T-cell leukemia virus type 1 (HTLV-1) activates viral gene expression via the ATF/CREB pathway. Tax also induces a variety of cellular genes through activation of the transcription factor NF-kappaB. The ability of Tax to activate the NF-kappaB pathway plays an essential role in HTLV-1-induced cellular transformation. This review briefly summarizes the remarkable discoveries of the past several years that have greatly advanced our knowledge on signal-mediated activation of the NF-kappaB pathway. It highlights our current understanding of how viral agents like Tax modulate cellular signaling machinery to activate the NF-kappaB pathway.
Collapse
Affiliation(s)
- Xiao Hua Li
- Division of Hematology-Oncology, Department of Medicine, Harold Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235-8594
| | - Richard B. Gaynor
- Division of Hematology-Oncology, Department of Medicine, Harold Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235-8594
- Address correspondence to Richard B. Gaynor, Division of Hematology-Oncology, Department of Medicine, U.T. Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75235-8594. Tel: (214) 648-7570; Fax: (214) 648-8862; E-mail:
| |
Collapse
|
43
|
Chekalina N, Burmak Y, Petrov Y, Borisova Z, Manusha Y, Kazakov Y, Kaidashev I. Quercetin reduces the transcriptional activity of NF-kB in stable coronary artery disease. Indian Heart J 2018; 70:593-597. [PMID: 30392493 PMCID: PMC6204471 DOI: 10.1016/j.ihj.2018.04.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 03/08/2018] [Accepted: 04/10/2018] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE The aim of this study was to determine the effect of quercetin on the indicators of chronic systemic inflammation (CSI) in stable coronary artery disease (CAD). METHODS This study included 85 patients with CAD, stable angina pectoris, functional class (FC) II, and heart failure (НF) 0-І. Each patient was prescribed beta-blockers, statins, and aspirin. In addition, a total of 30 patients, forming the study group received quercetin at a daily dose of 120mg for two months, while the remaining 55 patients made up the control group. The levels of cytokines, such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) in serum and the expression of the inhibitor of kappa B α (IkBα) gene in blood mononuclear cells, were determined. RESULTS The increased levels of IL-1β and TNF-α, as well as a moderate increase in IL-10 levels, were detected in the serum of patients with CAD. The expression of the IkBα gene (2-δСt) did not differ significantly between the groups. Under the influence of quercetin, levels of IL-1β and TNF-α were reduced and IL-10 levels tended to decrease. In contrast, the serum levels of these cytokines did not change significantly in the control group. The administration of quercetin decreased the expression of the IkBα gene (0.0092±0.0033 against 0.0261±0.0166, р=0.003; 2-δδСt, 2.82±1.39 times) in contrast to the control group. CONCLUSION Quercetin showed anti-inflammatory properties in patients with CAD, indicating a decrease in transcriptional activity of the nuclear factor of transcription kappa B (NF-kB).
Collapse
Affiliation(s)
- Nataliya Chekalina
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine.
| | - Yurii Burmak
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| | - Yeugen Petrov
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| | - Zinaida Borisova
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| | - Yulija Manusha
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| | - Yurii Kazakov
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| | - Igor Kaidashev
- Higher State Educational Establishment of Ukraine "Ukrainian Medical Stomatological Academy", Str. Shevchenka, 23, 36000, Poltava, Ukraine
| |
Collapse
|
44
|
Humphries JE, Deneckere LE. Characterization of a Toll-like receptor (TLR) signaling pathway in Biomphalaria glabrata and its potential regulation by NF-kappaB. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 86:118-129. [PMID: 29746981 DOI: 10.1016/j.dci.2018.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 05/16/2023]
|
45
|
Baig MS, Roy A, Saqib U, Rajpoot S, Srivastava M, Naim A, Liu D, Saluja R, Faisal SM, Pan Q, Turkowski K, Darwhekar GN, Savai R. Repurposing Thioridazine (TDZ) as an anti-inflammatory agent. Sci Rep 2018; 8:12471. [PMID: 30127400 PMCID: PMC6102213 DOI: 10.1038/s41598-018-30763-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 08/03/2018] [Indexed: 01/11/2023] Open
Abstract
Nuclear factor-kB (NF-kB) is a crucial transcription factor in the signal transduction cascade of the inflammatory signaling. Activation of NF-κB depends on the phosphorylation of IκBα by IκB kinase (IKKβ) followed by subsequent ubiquitination and degradation. This leads to the nuclear translocation of the p50- p65 subunits of NF-κB, and further triggers pro-inflammatory cytokine gene expression. Thus, in the need of a more effective therapy for the treatment of inflammatory diseases, specific inhibition of IKKβ represents a rational alternative strategy to the current therapies. A computer-aided drug identification protocol was followed to identify novel IKKβ inhibitors from a database of over 1500 Food and Drug Administration (FDA) drugs. The best scoring compounds were compared with the already known high-potency IKKβ inhibitors for their ability to bind and inhibit IKKβ by evaluating their docking energy. Finally, Thioridazinehydrochloride (TDZ), a potent antipsychotic drug against Schizophrenia was selected and its efficiency in inhibiting IκBα protein degradation and NF-κB activation was experimentally validated. Our study has demonstrated that TDZ blocks IκBα protein degradation and subsequent NF-κB activation to inhibit inflammation. Thus, it is a potential repurposed drug against inflammation.
Collapse
Affiliation(s)
- Mirza S Baig
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, 453552, India.
| | - Anjali Roy
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, 453552, India
| | - Uzma Saqib
- Discipline of Chemistry, Indian Institute of Technology Indore (IITI), Simrol, 453552, India
| | - Sajjan Rajpoot
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, 453552, India
| | - Mansi Srivastava
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, 453552, India
| | - Adnan Naim
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, 453552, India
| | - Dongfang Liu
- Centre for Inflammation & Epigenetics, Houston Methodist Research Institute, Houston, Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Bhopal, 462020, India
| | - Syed M Faisal
- National Institute of Animal Biotechnology (NIAB), Hyderabad, 500032, India
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, 3015 CE, Rotterdam, The Netherlands
| | - Kati Turkowski
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, 61231, Bad Nauheim, Germany
| | - Gajanan N Darwhekar
- Acropolis Institute of Pharmaceutical Education and Research (AIPER), Indore, 453771, India
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, 61231, Bad Nauheim, Germany.
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), member of the DZL, Justus Liebig University, Giessen, 35392, Germany.
| |
Collapse
|
46
|
Unger A, Finkernagel F, Hoffmann N, Neuhaus F, Joos B, Nist A, Stiewe T, Visekruna A, Wagner U, Reinartz S, Müller-Brüsselbach S, Müller R, Adhikary T. Chromatin Binding of c-REL and p65 Is Not Limiting for Macrophage IL12B Transcription During Immediate Suppression by Ovarian Carcinoma Ascites. Front Immunol 2018; 9:1425. [PMID: 29997615 PMCID: PMC6030372 DOI: 10.3389/fimmu.2018.01425] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/08/2018] [Indexed: 12/14/2022] Open
Abstract
Tumors frequently exploit homeostatic mechanisms that suppress expression of IL-12, a central mediator of inflammatory and anti-tumor responses. The p40 subunit of the IL-12 heterodimer, encoded by IL12B, is limiting for these functions. Ovarian carcinoma patients frequently produce ascites which exerts immunosuppression by means of soluble factors. The NFκB pathway is necessary for transcription of IL12B, which is not expressed in macrophages freshly isolated from ascites. This raises the possibility that ascites prevents IL12B expression by perturbing NFκB binding to chromatin. Here, we show that ascites-mediated suppression of IL12B induction by LPS plus IFNγ in primary human macrophages is rapid, and that suppression can be reversible after ascites withdrawal. Nuclear translocation of the NFκB transcription factors c-REL and p65 was strongly reduced by ascites. Surprisingly, however, their binding to the IL12B locus and to CXCL10, a second NFκB target gene, was unaltered, and the induction of CXCL10 transcription was not suppressed by ascites. These findings indicate that, despite its reduced nuclear translocation, NFκB function is not generally impaired by ascites, suggesting that ascites-borne signals target additional pathways to suppress IL12B induction. Consistent with these data, IL-10, a clinically relevant constituent of ascites and negative regulator of NFκB translocation, only partially recapitulated IL12B suppression by ascites. Finally, restoration of a defective IL-12 response by appropriate culture conditions was observed only in macrophages from a subset of donors, which may have important implications for the understanding of patient-specific immune responses.
Collapse
Affiliation(s)
- Annika Unger
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Florian Finkernagel
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Nathalie Hoffmann
- Experimental Tumor Research Group, Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Felix Neuhaus
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Barbara Joos
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute for Medical Microbiology and Hygiene, Biomedical Research Center (BMFZ), Philipps University of Marburg, Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Philipps University of Marburg, Marburg, Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, ZTI, Philipps University of Marburg, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Rolf Müller
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| | - Till Adhikary
- Institute for Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunobiology (ZTI), Philipps University of Marburg, Marburg, Germany
| |
Collapse
|
47
|
Song JH, Kang HB, Park SH, Jeong JH, Park J, You Y, Lee YH, Lee J, Kim E, Choi KC, Jun W. Extracts of Porphyra tenera (Nori Seaweed) Activate the Immune Response in Mouse RAW264.7 Macrophages via NF-κB Signaling. J Med Food 2018; 20:1152-1159. [PMID: 29243967 DOI: 10.1089/jmf.2017.4014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Porphyra tenera, also known as nori, is a red algal species of seaweed. It is cultivated in Asia for culinary purposes. We report that P. tenera extract (PTE) enhances the immune response in mouse macrophages. We found that P. tenera extract regulates the NF-κB IκB kinase (IKK) signaling pathway, and we assessed the expression and translocation of p65, a subunit of NF-κB, in RAW264.7 mouse macrophage cells after treatment with PTE. We also investigated the effects of 10% ethanol PTE (PTE10) in RAW264.7 cells. The production of IL-10, IL-6, TNF-α, and IFN-γ was induced by PTE treatment of the macrophages, and PTE also enhanced p-IκB and p-AKT. PTE10 showed no cytotoxicity at 10-20 μg/mL in RAW264.7 cells. PTE10, in fact, increased cell viability at 24 h, stimulated macrophage cells, and induced the phosphorylation of Akt. Akt stimulates IKK activity through the phosphorylation of IKKα and enhances immune activity through the activation of NF-κB. In this study, NF-κB activation was induced by increasing p-NF-κB and p-IKK. A subunit of NF-κB, p65, was located in the nucleus and increased the expression of various cytokines. PTE thus enhanced the immune response through IκB-α immunostimulation signaling in RAW264.7 cells. PTE10 has potential therefore for development of future treatments requiring immune system stimulation.
Collapse
Affiliation(s)
- Ji-Hye Song
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Hee-Bum Kang
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Seung-Ho Park
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Ji-Hoon Jeong
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Jeongjin Park
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| | - Yanghee You
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| | - Yoo-Hyun Lee
- 5 Department of Food Science and Nutrition, The University of Suwon , Suwon, Korea
| | - Jeongmin Lee
- 6 Department of Medical Nutrition, Kyung Hee University , Yongin, Korea
| | - Eungpil Kim
- 7 Marine Biotechnology Research Center , Jeonnam Bioindustry Foundation, Wando, Korea
| | - Kyung-Chul Choi
- 1 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul, South Korea .,2 Department of Pharmacology, University of Ulsan College of Medicine , Seoul, Korea
| | - Woojin Jun
- 3 Division of Food and Nutrition, Chonnam National University , Gwangju, Korea.,4 Research Institute for Human Ecology, Chonnam National University , Gwangju, Korea
| |
Collapse
|
48
|
Jönsson S, Lundberg AK, Chung RWS, Jonasson L. Glucocorticoid sensitivity and inflammatory status of peripheral blood mononuclear cells in patients with coronary artery disease. Ann Med 2018; 50:260-268. [PMID: 29473427 DOI: 10.1080/07853890.2018.1445278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Mechanisms behind sustained inflammation in patients with coronary artery disease (CAD) are not clarified but hypothalamus-pituitary-adrenal (HPA) axis dysfunction may have a role. Here, we investigated whether inflammatory status of peripheral blood mononuclear cells (PBMCs) was associated with altered glucocorticoid sensitivity in CAD patients. METHODS In 55 CAD patients and 30 controls, mRNA levels of GR-α, GR-β, NF-κB, IκBα, MMP-9 and TIMP-1 were measured in PBMCs. Suppressive effects of dexamethasone on GR-α, GR-β, NF-κB, IκBα, MMP-9 and TIMP-1 mRNA levels were assessed in PBMCs ex vivo. Salivary cortisol was repeatedly measured over 3 days. RESULTS GR-α mRNA levels were higher in CAD patients than in controls, 0.50 (0.38-0.59) versus 0.26 (0.18-0.37), p < .001, while GR-β mRNA levels were equally low in both groups. GR-α mRNA expression was associated with inflammatory gene expression and, also, with flatter diurnal cortisol rhythm. In both patients and controls, dexamethasone suppressed gene expression of NF-κB, IκBα, MMP-9 and TIMP-1 (p < .001). Dexamethasone also reduced GR-α mRNA levels (p < .001), while LPS increased it (p < .001). CONCLUSIONS PBMCs from CAD patients displayed an inflammatory gene expression profile. This was not explained by reduced glucocorticoid sensitivity. Instead, inflammation was associated with increased expression of GR-α mRNA, suggesting a hypocortisolemic state. Key messages • Peripheral blood mononuclear cells from patients with coronary artery disease (CAD) display an inflammatory gene expression profile. • This inflammatory state cannot be explained by reduced glucocorticoid sensitivity in CAD patients. • Instead, the inflammatory gene expression profile is associated with upregulated levels of glucocorticoid receptor-α mRNA, suggesting a hypocortisolemic state.
Collapse
Affiliation(s)
- Simon Jönsson
- a Department of Medical and Health Sciences, Division of Cardiovascular Medicine , Linköping University , Linköping , Sweden
| | - Anna K Lundberg
- a Department of Medical and Health Sciences, Division of Cardiovascular Medicine , Linköping University , Linköping , Sweden
| | - Rosanna W S Chung
- a Department of Medical and Health Sciences, Division of Cardiovascular Medicine , Linköping University , Linköping , Sweden
| | - Lena Jonasson
- a Department of Medical and Health Sciences, Division of Cardiovascular Medicine , Linköping University , Linköping , Sweden
| |
Collapse
|
49
|
Lee DK, Kim JH, Kim J, Choi S, Park M, Park W, Kim S, Lee KS, Kim T, Jung J, Choi YK, Ha KS, Won MH, Billiar TR, Kwon YG, Kim YM. REDD-1 aggravates endotoxin-induced inflammation via atypical NF-κB activation. FASEB J 2018; 32:4585-4599. [PMID: 29547704 DOI: 10.1096/fj.201701436r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Regulated in development and DNA damage responses 1 (REDD-1), an inhibitor of mammalian target of rapamycin (mTOR), is induced by various cell stressors, including LPS, a major player in the pathogenesis of endotoxemic shock. However, the pathologic role of REDD-1 in endotoxemia is largely unknown. We found that LPS increased REDD-1 expression, nuclear transcription factor-κB (NF-κB) activation, and inflammation and that these responses were suppressed by REDD-1 knockdown and in REDD-1+/- macrophages. REDD-1 overexpression stimulated NF-κB-dependent inflammation without additional LPS stimulation. REDD-1-induced NF-κB activation was independent of 2 classic IKK-dependent NF-κB pathways and the mTOR signaling pathway; however, REDD-1, particularly its C-terminal region (178-229), interacted with and sequestered IκBα, to elicit atypical NF-κB activation during the delayed and persistent phases of inflammation after stimulation. Moreover, REDD-1 knockdown mitigated vascular inflammation and permeability in endotoxemic mice, resulting in decreases in immune cell infiltration, systemic inflammation, caspase-3 activation, apoptosis, and consequent mortality. We further confirmed the inflammatory and cytotoxic effects of REDD-1 in endotoxemic REDD-1+/- mice. Our data support the likelihood that REDD-1 exacerbates endotoxemic inflammation via atypical NF-κB activation by sequestering IκBα.-Lee, D.-K., Kim, J.-H., Kim, J., Choi, S., Park, M., Park, W., Kim, S., Lee, K.-S., Kim, T., Jung, J., Choi, Y. K., Ha, K.-S., Won, M.-H., Billiar, T. R., Kwon, Y.-G., Kim, Y.-M. REDD-1 aggravates endotoxin-induced inflammation via atypical NF-κB activation.
Collapse
Affiliation(s)
- Dong-Keon Lee
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ji-Hee Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joohwan Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Seunghwan Choi
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - MinSik Park
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Wonjin Park
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Suji Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Kyu-Sun Lee
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Taesam Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jiwon Jung
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Yoon Kyung Choi
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Seoul, South Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
50
|
Canton J, Fehr AR, Fernandez-Delgado R, Gutierrez-Alvarez FJ, Sanchez-Aparicio MT, García-Sastre A, Perlman S, Enjuanes L, Sola I. MERS-CoV 4b protein interferes with the NF-κB-dependent innate immune response during infection. PLoS Pathog 2018; 14:e1006838. [PMID: 29370303 PMCID: PMC5800688 DOI: 10.1371/journal.ppat.1006838] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 02/06/2018] [Accepted: 12/21/2017] [Indexed: 12/28/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a novel human coronavirus that emerged in 2012, causing severe pneumonia and acute respiratory distress syndrome (ARDS), with a case fatality rate of ~36%. When expressed in isolation, CoV accessory proteins have been shown to interfere with innate antiviral signaling pathways. However, there is limited information on the specific contribution of MERS-CoV accessory protein 4b to the repression of the innate antiviral response in the context of infection. We found that MERS-CoV 4b was required to prevent a robust NF-κB dependent response during infection. In wild-type virus infected cells, 4b localized to the nucleus, while NF-κB was retained in the cytoplasm. In contrast, in the absence of 4b or in the presence of cytoplasmic 4b mutants lacking a nuclear localization signal (NLS), NF-κB was translocated to the nucleus leading to the expression of pro-inflammatory cytokines. This indicates that NF-κB repression required the nuclear import of 4b mediated by a specific NLS. Interestingly, we also found that both in isolation and during infection, 4b interacted with α-karyopherin proteins in an NLS-dependent manner. In particular, 4b had a strong preference for binding karyopherin-α4 (KPNA4), which is known to translocate the NF-κB protein complex into the nucleus. Binding of 4b to KPNA4 during infection inhibited its interaction with NF-κB-p65 subunit. Thereby we propose a model where 4b outcompetes NF-κB for KPNA4 binding and translocation into the nucleus as a mechanism of interference with the NF-κB-mediated innate immune response.
Collapse
Affiliation(s)
- Javier Canton
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Anthony R. Fehr
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
| | - Raúl Fernandez-Delgado
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | | | - Maria T. Sanchez-Aparicio
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Global Health and Emerging Pathogens Institute. Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Global Health and Emerging Pathogens Institute. Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States of America
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Isabel Sola
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|