151
|
Anti-inflammatory effects of secondary metabolites isolated from the marine-derived fungal strain Penicillium sp. SF-5629. Arch Pharm Res 2017; 40:328-337. [PMID: 28074397 DOI: 10.1007/s12272-017-0890-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 01/05/2017] [Indexed: 01/01/2023]
Abstract
After the chemical investigation of the ethyl acetate extract of the marine-derived fungal strain Penicillium sp. SF-5629, the isolation and structural elucidation of eight secondary metabolites, including (3R,4S)-6,8-dihydroxy-3,4,7-trimethylisocoumarin (1), (3S,4S)-sclerotinin A (2), penicitrinone A (3), citrinin H1 (4), emodin (5), ω-hydroxyemodin (6), 8-hydroxy-6-methyl-9-oxo-9H-xanthene-1-carboxylate (7), and 3,8-dihydroxy-6-methyl-9-oxo-9H-xanthene-1-carboxylate (8) were carried out. Evaluation of the anti-inflammatory activity of these metabolites showed that 4 inhibited nitric oxide and prostaglandin E2 production in lipopolysaccharide-stimulated BV2 microglia, with IC50 values of 8.1 ± 1.9 and 8.0 ± 2.8 μM, respectively. The inhibitory function of 4 was confirmed based on decreases in inducible nitric oxide synthesis and cyclooxygenase-2 gene expression. In addition, 4 was found to suppress the phosphorylation of inhibitor kappa B-α, interrupt the nuclear translocation of nuclear factor kappa B, and decrease the activation of p38 mitogen-activated protein kinase.
Collapse
|
152
|
Cho YC, Bach TT, Kim BR, Vuong HL, Cho S. Spilanthes acmella inhibits inflammatory responses via inhibition of NF-κB and MAPK signaling pathways in RAW 264.7 macrophages. Mol Med Rep 2017; 16:339-346. [DOI: 10.3892/mmr.2017.6555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/23/2017] [Indexed: 11/05/2022] Open
|
153
|
The matrix protein of rabies virus binds to RelAp43 to modulate NF-κB-dependent gene expression related to innate immunity. Sci Rep 2016; 6:39420. [PMID: 28000711 PMCID: PMC5175135 DOI: 10.1038/srep39420] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/22/2016] [Indexed: 02/08/2023] Open
Abstract
The matrix (M) protein of wild isolates of rabies virus such as Tha (M-Tha) was previously shown to be able to interact with RelAp43, a protein of the NF-κB family, and to efficiently suppress NF-κB-dependent reporter gene expression, in contrast with the vaccine strain SAD. Here, we analyze the mechanisms involved in RelAp43-M protein interaction. We demonstrate that the central part of M-Tha, and the specific C-terminal region of RelAp43 are required for this interaction. Four differences in the corresponding amino acid sequences of the M-Tha and M-SAD are shown to be crucial for RelAp43 interaction and subsequent modulation of innate immune response. Furthermore, the capacity of M-Tha to interact with RelAp43 was shown to be crucial for the control of the expression of four genes (IFN, TNF, IL8 and CXCL2) during viral infection. These findings reveal that RelAp43 is a potent regulator of transcription of genes involved in innate immune response during rabies virus infection and that the M protein of wild isolates of rabies virus is a viral immune-modulatory factor playing an important role in this RelAp43-mediated host innate immunity response in contrast to M protein of vaccine strains, which have lost this property.
Collapse
|
154
|
Keegan PM, Anbazhakan S, Kang B, Pace BS, Platt MO. Biomechanical and biochemical regulation of cathepsin K expression in endothelial cells converge at AP-1 and NF-κB. Biol Chem 2016; 397:459-68. [PMID: 26760306 DOI: 10.1515/hsz-2015-0244] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/04/2016] [Indexed: 11/15/2022]
Abstract
Cathepsins K and V are powerful elastases elevated in endothelial cells by tumor necrosis factor-α (TNFα) stimulation and disturbed blood flow both of which contribute to inflammation-mediated arterial remodeling. However, mechanisms behind endothelial cell integration of biochemical and biomechanical cues to regulate cathepsin production are not known. To distinguish these mechanisms, human aortic endothelial cells (HAECs) were stimulated with TNFα and exposed to pro-remodeling or vasoprotective shear stress profiles. TNFα upregulated cathepsin K via JNK/c-jun activation, but vasoprotective shear stress inhibited TNFα-stimulated cathepsin K expression. JNK/c-jun were still phosphorylated, but cathepsin K mRNA levels were significantly reduced to almost null indicating separate biomechanical regulation of cathepsin K by shear stress separate from biochemical stimulation. Treatment with Bay 11-7082, an inhibitor of IκBα phosphorylation, was sufficient to block induction of cathepsin K by both pro-remodeling shear stress and TNFα, implicating NF-κB as the biomechanical regulator, and its protein levels were reduced in HAECs by vasoprotective shear stress. In conclusion, NF-κB and AP-1 activation were necessary to activate cathepsin K expression in endothelial cells, highlighting integration of biochemical and biomechanical stimuli to control cathepsins K and V, powerful elastases implicated for arterial remodeling due to chronic inflammation and disturbed blood flow.
Collapse
|
155
|
Honke N, Shaabani N, Zhang DE, Hardt C, Lang KS. Multiple functions of USP18. Cell Death Dis 2016; 7:e2444. [PMID: 27809302 PMCID: PMC5260889 DOI: 10.1038/cddis.2016.326] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/12/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022]
Abstract
Since the discovery of the ubiquitin system and the description of its important role in the degradation of proteins, many studies have shown the importance of ubiquitin-specific peptidases (USPs). One special member of this family is the USP18 protein (formerly UBP43). In the past two decades, several functions of USP18 have been discovered: this protein is not only an isopeptidase but also a potent inhibitor of interferon signaling. Therefore, USP18 functions as 'a' maestro of many biological pathways in various cell types. This review outlines multiple functions of USP18 in the regulation of various immunological processes, including pathogen control, cancer development, and autoimmune diseases.
Collapse
Affiliation(s)
- Nadine Honke
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Namir Shaabani
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany.,Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dong-Er Zhang
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Cornelia Hardt
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Karl S Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| |
Collapse
|
156
|
Yazdi S, Naumann M, Stein M. Double phosphorylation-induced structural changes in the signal-receiving domain of IκBα in complex with NF-κB. Proteins 2016; 85:17-29. [DOI: 10.1002/prot.25181] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/19/2016] [Accepted: 09/24/2016] [Indexed: 02/01/2023]
Affiliation(s)
- Samira Yazdi
- Max Planck Institute for Dynamics of Complex Technical Systems, Molecular Simulations and Design Group; Sandtorstrasse 1 39106 Magdeburg Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg; Leipziger Strasse 44 39120 Magdeburg Germany
| | - Matthias Stein
- Max Planck Institute for Dynamics of Complex Technical Systems, Molecular Simulations and Design Group; Sandtorstrasse 1 39106 Magdeburg Germany
| |
Collapse
|
157
|
Galdieri L, Gatla H, Vancurova I, Vancura A. Activation of AMP-activated Protein Kinase by Metformin Induces Protein Acetylation in Prostate and Ovarian Cancer Cells. J Biol Chem 2016; 291:25154-25166. [PMID: 27733682 DOI: 10.1074/jbc.m116.742247] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/29/2016] [Indexed: 12/13/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is an energy sensor and master regulator of metabolism. AMPK functions as a fuel gauge monitoring systemic and cellular energy status. Activation of AMPK occurs when the intracellular AMP/ATP ratio increases and leads to a metabolic switch from anabolism to catabolism. AMPK phosphorylates and inhibits acetyl-CoA carboxylase (ACC), which catalyzes carboxylation of acetyl-CoA to malonyl-CoA, the first and rate-limiting reaction in de novo synthesis of fatty acids. AMPK thus regulates homeostasis of acetyl-CoA, a key metabolite at the crossroads of metabolism, signaling, chromatin structure, and transcription. Nucleocytosolic concentration of acetyl-CoA affects histone acetylation and links metabolism and chromatin structure. Here we show that activation of AMPK with the widely used antidiabetic drug metformin or with the AMP mimetic 5-aminoimidazole-4-carboxamide ribonucleotide increases the inhibitory phosphorylation of ACC and decreases the conversion of acetyl-CoA to malonyl-CoA, leading to increased protein acetylation and altered gene expression in prostate and ovarian cancer cells. Direct inhibition of ACC with allosteric inhibitor 5-(tetradecyloxy)-2-furoic acid also increases acetylation of histones and non-histone proteins. Because AMPK activation requires liver kinase B1, metformin does not induce protein acetylation in liver kinase B1-deficient cells. Together, our data indicate that AMPK regulates the availability of nucleocytosolic acetyl-CoA for protein acetylation and that AMPK activators, such as metformin, have the capacity to increase protein acetylation and alter patterns of gene expression, further expanding the plethora of metformin's physiological effects.
Collapse
Affiliation(s)
- Luciano Galdieri
- From the Department of Biological Sciences, St. John's University, Queens, New York 11439
| | - Himavanth Gatla
- From the Department of Biological Sciences, St. John's University, Queens, New York 11439
| | - Ivana Vancurova
- From the Department of Biological Sciences, St. John's University, Queens, New York 11439
| | - Ales Vancura
- From the Department of Biological Sciences, St. John's University, Queens, New York 11439
| |
Collapse
|
158
|
Tang N, Ma J, Wang KS, Mi C, Lv Y, Piao LX, Xu GH, Li X, Lee JJ, Jin X. Dihydromyricetin suppresses TNF-α-induced NF-κB activation and target gene expression. Mol Cell Biochem 2016; 422:11-20. [PMID: 27686451 DOI: 10.1007/s11010-016-2799-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022]
Abstract
Nuclear factor-kappa B (NF-κB) has been reported to play a pivotal role in many physiological processes including inflammation, apoptosis, and angiogenesis. We discovered a potent natural NF-κB inhibitor, dihydromyricetin, from the traditional herb Ampelopsis grossedentata, which has a long history of use in food and medicine. In this study, we demonstrated the effect of dihydromyricetin on NF-κB activation in TNF-α-induced HeLa cells. Dihydromyricetin was found to markedly inhibit the phosphorylation and degradation of the inhibitor of NF-κB alpha (IκBα), and subsequent nuclear translocation of p65. Dihydromyricetin also has an impact on upstream signaling of IKK through the inhibition of expression of adaptor proteins, TNF receptor-associated factor 2 (TRAF2), and receptor-interacting protein 1 (RIP1). Furthermore, the current results reveal that dihydromyricetin led to the downregulation of target genes involved in inflammation, proliferation, as well as potentiation of TNF-α-induced apoptosis through suppressing the activation of NF-κB. In conclusion, our data indicate that dihydromyricetin may be a potentially useful therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Nina Tang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Juan Ma
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Ke Si Wang
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Chunliu Mi
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Ying Lv
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Lian Xun Piao
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Guang Hua Xu
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Xuezheng Li
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Jung Joon Lee
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China
| | - Xuejun Jin
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin Province, China.
| |
Collapse
|
159
|
Wostradowski T, Prajeeth CK, Gudi V, Kronenberg J, Witte S, Brieskorn M, Stangel M. In vitro evaluation of physiologically relevant concentrations of teriflunomide on activation and proliferation of primary rodent microglia. J Neuroinflammation 2016; 13:250. [PMID: 27658519 PMCID: PMC5034581 DOI: 10.1186/s12974-016-0715-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/08/2016] [Indexed: 11/24/2022] Open
Abstract
Background Teriflunomide, an inhibitor of dihydroorotate dehydrogenase, is thought to ameliorate multiple sclerosis by reducing activation-induced proliferation of lymphocytes, which is highly dependent on de novo pyrimidine synthesis. Nevertheless, its immunomodulatory effects on resident glial cells in the central nervous system are only poorly understood. Methods In this study, we employed physiologically relevant concentrations of teriflunomide and investigated its effects on survival, proliferation, activation, and function of primary rat microglia in vitro. Results We demonstrate that teriflunomide had no cytotoxic effect on microglia and had only a minor impact on microglial activation. In a concentration- and time-dependent manner, teriflunomide significantly downregulated surface expression of the co-stimulatory molecule CD86. Furthermore, in the highest concentration applied (5 μM), it slightly increased the expression of interleukin-10 in microglia in response to lipopolysaccharide. Treatment with low concentrations of teriflunomide (0.25–1 μM) did not have any impact on the activation or proliferation of microglia. At 5 μM concentration of teriflunomide, we observed a reduction of approximately 30 % in proliferation of microglia in mixed glial cell cultures. Conclusions Taken together, our in vitro findings suggest that at higher concentrations, teriflunomide potentially exerts its effects by reducing microglial proliferation and not by modulating the M1-/M2-like cell differentiation of primary rat microglia. Thus, teriflunomide has no major impact on the plasticity of microglia; however, the anti-proliferative and minimal anti-inflammatory effects might be clinically relevant for immune modulation in the treatment of neuroinflammatory CNS diseases such as multiple sclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0715-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tanja Wostradowski
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,Center for Systems Neuroscience, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Chittappen Kandiyil Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Jessica Kronenberg
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany.,Center for Systems Neuroscience, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sina Witte
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Marina Brieskorn
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Carl-Neuberg-Str.1, 30625, Hannover, Germany. .,Center for Systems Neuroscience, University of Veterinary Medicine Hannover, 30559, Hannover, Germany.
| |
Collapse
|
160
|
Kang K, Won M, Yuk JM, Park CY, Byun HS, Park KA, Lee SR, Kang YG, Shen HM, Lee IY, Hur GM. IinQ attenuates systemic inflammatory responses via selectively impairing the Myddosome complex formation upon TLR4 ligation. Biochem Pharmacol 2016; 121:52-66. [PMID: 27664853 DOI: 10.1016/j.bcp.2016.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/19/2016] [Indexed: 01/21/2023]
Abstract
A specific small-molecule inhibitor of the TLR4 signaling complex upstream of the IKK would likely provide therapeutic benefit for NF-κB-mediated inflammatory disease. We previously identified brazilin as a selective upstream IKK inhibitor targeting the Myddosome complex. In this study, using a cell-based ubiquitination assay for IRAK1 and a chemical library comprising a series of structural analogues of brazilin, a novel small molecule, 2-hydroxy-5,6-dihydroisoindolo[1,2-a]isoquinoline-3,8-dione (IinQ), was identified as a selective and potent inhibitor of IRAK1-dependent NF-κB activation upon TLR4 ligation. In RAW264.7 macrophages, IinQ drastically suppressed activation of upstream IKK signaling events including membrane-bound IRAK1 ubiquitination and IKK phosphorylation by the TLR4 ligand, resulting in reduced expression of proinflammatory mediators including IL-6, TNF-α, and nitric oxide. Interestingly, IinQ did not suppress NF-κB activation via the TLR3 ligand, DNA damaging agents, or a protein kinase C activator, indicating IinQ is specific for TLR4 signaling. Analysis of upstream signaling events further confirmed that IinQ disrupts the MyD88-IRAK1-TRAF6 complex formation induced by LPS treatment, without affecting TLR4 oligomerization. Moreover, intravenous administration of IinQ significantly reduced lethality and attenuated systemic inflammatory responses in an in vivo mouse model of endotoxin shock following LPS challenge. Thus, IinQ represents a novel class of brazilin analogues with improved potency and specificity toward disruption of Myddosome complex formation in TLR4 signaling, indicating that IinQ may be a promising therapeutic candidate for the treatment of systemic inflammatory diseases.
Collapse
Affiliation(s)
- Kidong Kang
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Minho Won
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Jae-Min Yuk
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Infection Biology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Chan-Yong Park
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea
| | - Hee Sun Byun
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Kyeong Ah Park
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - So-Ra Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea
| | - Young-Goo Kang
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ill Young Lee
- Eco-Friendly New Materials Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yusung-gu, Daejeon 34114, Republic of Korea.
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea; Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Daejeon 35015, Republic of Korea.
| |
Collapse
|
161
|
Rosiglitazone, a Peroxisome Proliferator-Activated Receptor (PPAR)-γ Agonist, Attenuates Inflammation Via NF-κB Inhibition in Lipopolysaccharide-Induced Peritonitis. Inflammation 2016; 38:2105-15. [PMID: 26047949 DOI: 10.1007/s10753-015-0193-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We assessed the anti-inflammatory effect of peroxisome proliferator-activated receptor (PPAR)-γ agonist, rosiglitazone, in a lipopolysaccharide (LPS)-induced peritonitis rat model. LPS was intraperitoneally injected into rats to establish peritonitis model. Male Sprague-Dawley (SD) rats were assigned to normal saline (the solvent of LPS), LPS, rosiglitazone plus LPS, and rosiglitazone alone. A simple peritoneal equilibrium test was performed with 20 ml 4.25 % peritoneal dialysis fluid. We measured the leukocyte count in dialysate and ultrafiltration volume. Peritoneal membrane histochemical staining was performed, and peritoneal thickness was assessed. CD40 and intercellular adhesion molecule-1 messenger RNA (ICAM-1 mRNA) levels in rat visceral peritoneum were detected by reverse transcription (RT)-PCR. IL-6 in rat peritoneal dialysis effluent was measured using enzyme-linked immunosorbent assay. The phosphorylation of NF-κB-p65 and IκBα was analyzed by Western blot. LPS administration resulted in increased peritoneal thickness and decreased ultrafiltration volume. Rosiglitazone pretreatment significantly decreased peritoneal thickness. In addition to CD40 and ICAM-1 mRNA expression, the IL-6, p-p65, and p-IκBα protein expressions were enhanced in LPS-administered animals. Rosiglitazone pretreatment significantly decreased ICAM-1 mRNA upregulation, secretion of IL-6 protein, and phosphorylation of NF-κB-p65 and IκBα without decreasing CD40 mRNA expression. Rosiglitazone has a protective effect in peritonitis, simultaneously decreasing NF-κB phosphorylation, suggesting that NF-κB signaling pathway mediated peritoneal inflammation induced by LPS. PPAR-γ might be considered a potential therapeutic target against peritonitis.
Collapse
|
162
|
Rithidech KN, Jangiam W, Tungjai M, Gordon C, Honikel L, Whorton EB. Induction of Chronic Inflammation and Altered Levels of DNA Hydroxymethylation in Somatic and Germinal Tissues of CBA/CaJ Mice Exposed to (48)Ti Ions. Front Oncol 2016; 6:155. [PMID: 27446801 PMCID: PMC4921787 DOI: 10.3389/fonc.2016.00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/10/2016] [Indexed: 11/17/2022] Open
Abstract
Although the lung is one of the target organs at risk for cancer induction from exposure to heavy ions found in space, information is insufficient on cellular/molecular responses linked to increased cancer risk. Knowledge of such events may aid in the development of new preventive measures. Furthermore, although it is known that germinal cells are sensitive to X- or γ-rays, there is little information on the effects of heavy ions on germinal cells. Our goal was to investigate in vivo effects of 1 GeV/n (48)Ti ions (one of the important heavy ions found in the space environment) on somatic (lung) and germinal (testis) tissues collected at various times after a whole body irradiation of CBA/CaJ mice (0, 0.1, 0.25, or 0.5 Gy, delivered at 1 cGy/min). We hypothesized that (48)Ti-ion-exposure induced damage in both tissues. Lung tissue was collected from each mouse from each treatment group at 1 week, 1 month, and 6 months postirradiation. For the testis, we collected samples at 6 months postirradiation. Hence, only late-occurring effects of (48)Ti ions in the testis were studied. There were five mice per treatment group at each harvest time. We investigated inflammatory responses after exposure to (48)Ti ions by measuring the levels of activated nuclear factor kappa B and selected pro-inflammatory cytokines in both tissues of the same mouse. These measurements were coupled with the quantitation of the levels of global 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC). Our data clearly showed the induction of chronic inflammation in both tissues of exposed mice. A dose-dependent reduction in global 5hmC was found in the lung at all time-points and in testes collected at 6 months postirradiation. In contrast, significant increases in global 5mC were found only in lung and testes collected at 6 months postirradiation from mice exposed to 0.5 Gy of 1 GeV/n (48)Ti ions. Overall, our data showed that (48)Ti ions may create health risks in both lung and testicular tissues.
Collapse
Affiliation(s)
| | - Witawat Jangiam
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- Department of Chemical Engineering, Faculty of Engineering, Burapha University, Chonburi, Thailand
| | - Montree Tungjai
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Center of Excellence for Molecular Imaging, Chiang Mai University, Chiang Mai, Thailand
| | - Chris Gordon
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Louise Honikel
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
163
|
He G, Li LI, Guan E, Chen J, Qin YI, Xie Y. Fentanyl inhibits the progression of human gastric carcinoma MGC-803 cells by modulating NF-κB-dependent gene expression in vivo. Oncol Lett 2016; 12:563-571. [PMID: 27347181 DOI: 10.3892/ol.2016.4619] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 03/24/2016] [Indexed: 12/13/2022] Open
Abstract
Fentanyl is widely used to treat acute and chronic pain. Previous in vitro studies by the present authors demonstrated that fentanyl inhibits the progression of the MGC-803 human gastric carcinoma cell line by affecting apoptosis-related genes, including nuclear factor-kappa B (NF-κB) and phosphatase and tensin homolog. In the present study, the effects of fentanyl on NF-κB-dependent gene expression were investigated in vivo. Nude mice were inoculated with an MGC-803 cell suspension, and mice that developed subcutaneous tumors measuring >1.0×1.0 cm were selected for study. Mice were administered intraperitoneal injections of fentanyl (0.05 mg/kg, group F1; 0.1 mg/kg, group F2; 0.2 mg/kg, group F3; and 0.4 mg/kg, group F4) for 14 consecutive days. Non-fentanyl-treated mice (group C) and normal saline-treated mice (group N) served as the control groups. Tumor growth was monitored by calculating the time-shift of the growth curve. Morphological changes were also observed using microscopy. The expression of NF-κB, B-cell lymphoma-2 (Bcl-2), B-cell associated X protein (Bax), vascular endothelial growth factor-A (VEGF-A) and matrix metalloproteinase-9 (MMP-9) in the subcutaneous tumor tissue was also analyzed by reverse transcription-polymerase chain reaction and western blot analysis, and confirmed using immunohistochemistry. The relative tumor volumes of groups F1, F2, F3 and F4 were significantly reduced compared with groups C and N. Furthermore, subcutaneous tumor cells exhibited nuclear swelling, chromatin condensation, reduced chromatin and nuclear fragmentation in the F1, F2, F3 and F4 groups. The number of NF-κB+, Bcl-2+, VEGF-A+ and MMP-9+ subcutaneous tumor cells was reduced, whereas the number of Bax+ cells was increased in the F1, F2, F3 and F4 groups. Additionally, in these groups, tumor expression of NF-κB, Bcl-2, VEGF-A and MMP-9 transcripts and proteins was downregulated, while Bax messenger RNA and protein expression levels were upregulated. The results revealed that fentanyl inhibits the growth of subcutaneous human gastric carcinoma tumors in mice. Therefore, it could be hypothesized that this antineoplastic activity may result from the inhibition of NF-κB activation, suppression of downstream VEGF-A and MMP-9 expression, and normalization of the pro-apoptotic Bax/Bcl-2 ratio.
Collapse
Affiliation(s)
- Guodong He
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China; Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - L I Li
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Enjian Guan
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Chen
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Y I Qin
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
164
|
The simple neuroendocrine-immune regulatory network in oyster Crassostrea gigas mediates complex functions. Sci Rep 2016; 6:26396. [PMID: 27193598 PMCID: PMC4872224 DOI: 10.1038/srep26396] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 04/29/2016] [Indexed: 01/01/2023] Open
Abstract
The neuroendocrine-immune (NEI) regulatory network is a complex system, which plays an indispensable role in the immunity of the host. In the present study, the bioinformatical analysis of the transcriptomic data from oyster Crassostrea gigas and further biological validation revealed that oyster TNF (CgTNF-1 CGI_10018786) could activate the transcription factors NF-κB and HSF (heat shock transcription factor) through MAPK signaling pathway, and then regulate apoptosis, redox reaction, neuro-regulation and protein folding in oyster haemocytes. The activated immune cells then released neurotransmitters including acetylcholine, norepinephrine and [Met(5)]-enkephalin to regulate the immune response by arising the expression of three TNF (CGI_10005109, CGI_10005110 and CGI_10006440) and translocating two NF-κB (Cgp65, CGI_10018142 and CgRel, CGI_10021567) between the cytoplasm and nuclei of haemocytes. Neurotransmitters exhibited the immunomodulation effects by influencing apoptosis and phagocytosis of oyster haemocytes. Acetylcholine and norepinephrine could down-regulate the immune response, while [Met(5)]-enkephalin up-regulate the immune response. These results suggested that the simple neuroendocrine-immune regulatory network in oyster might be activated by oyster TNF and then regulate the immune response by virtue of neurotransmitters, cytokines and transcription factors.
Collapse
|
165
|
Qin S, Qin L, Zhang C, Liu L, Sun W, Li N, Wu R, Wang X. p120-Catenin modulating nuclear factor-κB activation is partially RhoA/ROCKdependent in scratch injury. Wound Repair Regen 2016; 23:231-40. [PMID: 25693631 DOI: 10.1111/wrr.12270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 02/12/2015] [Indexed: 12/01/2022]
Abstract
p120-catenin (p120) is known as a cadherin-associated protein that participates in tumor metastasis and invasion, as well as an anti-inflammatory mediator. Recently, its anti-inflammatory role is drawing increasing attention, but the regulatory mechanisms are still unknown. Here, we report that p120 modulated inflammatory responses partially depends on RhoA/ROCK pathway in scratch-induced injury in human bronchial epithelial cells (BECs). For the first time, we found that p120 was significantly reduced in BECs after scratching, which could induce interleukin-8 (IL-8) production through nuclear factor-κB (NF-κB) activation accompanied with IκBα phosphorylation. Over-expression of p120 3A could inhibit NF-κB activation and IL-8 mRNA expression and protein synthesis after scratching, while p120 knockdown by small interfering RNA could promote NF-κB activation and IL-8 mRNA expression and protein synthesis after scratching. Furthermore, we found that RhoA was the binding partner of p120 in BECs. Although total RhoA and p120-binded RhoA remained unchanged, the RhoA activity was increased after scratching. Chemical blockade of RhoA/ROCK signaling (Y27632) inhibited scratch-induced nuclear translocation of NF-κB p65. Over-expression of p120 3A attenuated scratch-induced RhoA activation, whereas silence of p120 significantly elevated scratch-induced RhoA activation in BCEs. Conclusively, these results indicate an anti-inflammatory effect of p120 in bronchial epithelial cells through its modulation of NF-κB signaling depending on RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Shenghui Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Lingzhi Qin
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Chao Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Liwei Liu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Wenjia Sun
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Naping Li
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Renliang Wu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| | - Xi Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Pulmonary Disease of Ministry of Health of China, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
166
|
Perry CJ, Blake P, Buettner C, Papavassiliou E, Schain AJ, Bhasin MK, Burstein R. Upregulation of inflammatory gene transcripts in periosteum of chronic migraineurs: Implications for extracranial origin of headache. Ann Neurol 2016; 79:1000-13. [PMID: 27091721 DOI: 10.1002/ana.24665] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/29/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Chronic migraine (CM) is often associated with chronic tenderness of pericranial muscles. A distinct increase in muscle tenderness prior to onset of occipital headache that eventually progresses into a full-blown migraine attack is common. This experience raises the possibility that some CM attacks originate outside the cranium. The objective of this study was to determine whether there are extracranial pathophysiologies in these headaches. METHODS We biopsied and measured the expression of gene transcripts (mRNA) encoding proteins that play roles in immune and inflammatory responses in affected (ie, where the head hurts) calvarial periosteum of (1) patients whose CMs are associated with muscle tenderness and (2) patients with no history of headache. RESULTS Expression of proinflammatory genes (eg, CCL8, TLR2) in the calvarial periosteum significantly increased in CM patients attesting to muscle tenderness, whereas expression of genes that suppress inflammation and immune cell differentiation (eg, IL10RA, CSF1R) decreased. INTERPRETATION Because the upregulated genes were linked to activation of white blood cells, production of cytokines, and inhibition of NF-κB, and the downregulated genes were linked to prevention of macrophage activation and cell lysis, we suggest that the molecular environment surrounding periosteal pain fibers is inflamed and in turn activates trigeminovascular nociceptors that reach the affected periosteum through suture branches of intracranial meningeal nociceptors and/or somatic branches of the occipital nerve. This study provides the first set of evidence for localized extracranial pathophysiology in CM. Ann Neurol 2016;79:1000-1013.
Collapse
Affiliation(s)
| | - Pamela Blake
- Headache Center of Greater Heights, Memorial Hermann Greater Heights Hospital, Houston, TX
| | - Catherine Buettner
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA.,Harvard Medical School, Boston, MA
| | - Efstathios Papavassiliou
- Harvard Medical School, Boston, MA.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | - Aaron J Schain
- Harvard Medical School, Boston, MA.,Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, MA
| | - Manoj K Bhasin
- Harvard Medical School, Boston, MA.,Division of Genomics, Proteomics, Bioinformatics, and Systems Biology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Rami Burstein
- Harvard Medical School, Boston, MA.,Department of Anesthesia and Critical Care, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
167
|
Ghag G, Wolf LM, Reed RG, Van Der Munnik NP, Mundoma C, Moss MA, Rangachari V. Fully reduced granulin-B is intrinsically disordered and displays concentration-dependent dynamics. Protein Eng Des Sel 2016; 29:177-86. [PMID: 26957645 PMCID: PMC4830411 DOI: 10.1093/protein/gzw005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/25/2016] [Accepted: 01/29/2016] [Indexed: 02/06/2023] Open
Abstract
Granulins (Grns) are a family of small, cysteine-rich proteins that are generated upon proteolytic cleavage of their precursor, progranulin (Pgrn). All seven Grns (A-G) contain 12 conserved cysteines that form 6 intramolecular disulfide bonds, rendering this family of proteins unique. Grns are known to play multi-functional roles, including wound healing, embryonic growth, and inflammation and are implicated in neurodegenerative diseases. Despite their manifold functions, there exists a dearth of information regarding their structure-function relationship. Here, we sought to establish the role of disulfide bonds in promoting structure by investigating the fully reduced GrnB (rGrnB). We report that monomeric rGrnB is an intrinsically disordered protein (IDP) at low concentrations. rGrnB undergoes dimerization at higher concentrations to form a fuzzy complex without a net gain in the structure-a behavior increasingly identified as a hallmark of some IDPs. Interestingly, we show that rGrnB is also able to activate NF-κB in human neuroblastoma cells in a concentration-dependent manner. This activation correlates with the observed monomer-dimer dynamics. Collectively, the presented data establish that the intrinsic disorder of rGrnB governs conformational dynamics within the reduced form of the protein, and suggest that the overall structure of Grns could be entirely dictated by disulfide bonds.
Collapse
Affiliation(s)
- Gaurav Ghag
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | | | - Randi G Reed
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | | | - Claudius Mundoma
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Melissa A Moss
- Biomedical Engineering Program and Department of Chemical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Vijayaraghavan Rangachari
- Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| |
Collapse
|
168
|
Song SM, Ham YM, Ko YJ, Ko EY, Oh DJ, Kim CS, Kim D, Kim KN, Yoon WJ. Anti-inflammatory activities of the products of supercritical fluid extraction from Litsea japonica fruit in RAW 264.7 cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
169
|
Inhibition of Acute Lung Injury by TNFR-Fc through Regulation of an Inflammation-Oxidative Stress Pathway. PLoS One 2016; 11:e0151672. [PMID: 26990441 PMCID: PMC4798551 DOI: 10.1371/journal.pone.0151672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/02/2016] [Indexed: 01/11/2023] Open
Abstract
Background Acute lung injury (ALI), characterized by disruption of the lung alveolar-capillary membrane barrier and resultant pulmonary edema, and associated with a proteinaceous alveolar exudate, is a leading cause of morbidity and mortality. Currently, inflammation-oxidative stress interaction between TNF-α and NF-κB was identified as a key pathway of ALI. We hypothesized that a TNFR-Fc fusion protein would have beneficial effects in experimental ALI, and sought to test this idea in mice by blocking TNF-α. Methods and Results Intratracheal instillation of lipopolysaccharide (LPS) into the lungs of ALI mice led to histiocyte apoptosis, and detection of serum and bronchoalveolar lavage fluid (BALF) cytokines, feedback between NF-κB and TNF-α, lung albumin leakage, lung damage, IκB kinase (IKK) and NF-κB activation, I-κB degradation, and oxidative injury. LPS administration raised pulmonary inflammation as reflected by increased inflammatory cytokines, alveoli protein concentration, and ALI scores. IKK is phosphorylated following LPS challenge, leading to I-κB degradation and NF-κB p65 phosphorylation. Furthermore, NF-κB is translocated into the nucleus and up-regulates TNF-α gene transcription. Infusion of TNFR-Fc 24h before LPS challenge significantly abrogated the increase of inflammatory cytokines, especially serum TNF-α concentration, as well as pulmonary alveoli protein levels, and diminished IKK and NF-κB activation and I-κB degradation. The nuclear translocation of NF-κB was inhibited, following by down-regulation of TNF-α gene transcription. In addition, LPS intratracheal instillation induced marked oxidative damage, such as a decrease in total anti-oxidation products and an increase in malondialdehyde (MDA), as well as up-regulation of oxidation enzymes. Histologic analysis and apoptosis scores revealed that the extent of tissue lesions was significantly reduced, but not abrogated, by TNF-α blockade. Conclusion Treatment with LPS alone increased inflammation and oxidative stress in ALI mice, while administration of TNFR-Fc 24h before LPS challenge broke the feedback between NF-κB and TNF-α, resulting in decreased pulmonary inflammation/oxidative damage and tissue destruction. These results suggest a potential role for TNF-α therapy to treat clinical ALI.
Collapse
|
170
|
Platt MO, Shockey WA. Endothelial cells and cathepsins: Biochemical and biomechanical regulation. Biochimie 2016; 122:314-23. [PMID: 26458976 PMCID: PMC4747805 DOI: 10.1016/j.biochi.2015.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/07/2015] [Indexed: 01/12/2023]
Abstract
Cathepsins are mechanosensitive proteases that are regulated not only by biochemical factors, but are also responsive to biomechanical forces in the cardiovascular system that regulate their expression and activity to participate in cardiovascular tissue remodeling. Their elastinolytic and collagenolytic activity have been implicated in atherosclerosis, abdominal aortic aneurysms, and in heart valve disease, all of which are lined by endothelial cells that are the mechanosensitive monolayer of cells that sense and respond to fluid shear stress as the blood flows across the surfaces of the arteries and valve leaflets. Inflammatory cytokine signaling is integrated with biomechanical signaling pathways by the endothelial cells to transcribe, translate, and activate either the cysteine cathepsins to remodel the tissue or to express their inhibitors to maintain healthy cardiovascular tissue structure. Other cardiovascular diseases should now be included in the study of the cysteine cathepsin activation because of the additional biochemical cues they provide that merges with the already existing hemodynamics driving cardiovascular disease. Sickle cell disease causes a chronic inflammation including elevated TNFα and increased numbers of circulating monocytes that alter the biochemical stimulation while the more viscous red blood cells due to the sickling of hemoglobin alters the hemodynamics and is associated with accelerated elastin remodeling causing pediatric strokes. HIV-mediated cardiovascular disease also occurs earlier in than the broader population and the influence of HIV-proteins and antiretrovirals on endothelial cells must be considered to understand these accelerated mechanisms in order to identify new therapeutic targets for prevention.
Collapse
Affiliation(s)
- Manu O Platt
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Suite 3015, Atlanta, GA 30332, USA.
| | - W Andrew Shockey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Suite 3015, Atlanta, GA 30332, USA.
| |
Collapse
|
171
|
Csizmok V, Follis AV, Kriwacki RW, Forman-Kay JD. Dynamic Protein Interaction Networks and New Structural Paradigms in Signaling. Chem Rev 2016; 116:6424-62. [PMID: 26922996 DOI: 10.1021/acs.chemrev.5b00548] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Understanding signaling and other complex biological processes requires elucidating the critical roles of intrinsically disordered proteins (IDPs) and regions (IDRs), which represent ∼30% of the proteome and enable unique regulatory mechanisms. In this review, we describe the structural heterogeneity of disordered proteins that underpins these mechanisms and the latest progress in obtaining structural descriptions of conformational ensembles of disordered proteins that are needed for linking structure and dynamics to function. We describe the diverse interactions of IDPs that can have unusual characteristics such as "ultrasensitivity" and "regulated folding and unfolding". We also summarize the mounting data showing that large-scale assembly and protein phase separation occurs within a variety of signaling complexes and cellular structures. In addition, we discuss efforts to therapeutically target disordered proteins with small molecules. Overall, we interpret the remodeling of disordered state ensembles due to binding and post-translational modifications within an expanded framework for allostery that provides significant insights into how disordered proteins transmit biological information.
Collapse
Affiliation(s)
- Veronika Csizmok
- Molecular Structure & Function, The Hospital for Sick Children , Toronto, ON M5G 0A4, Canada
| | - Ariele Viacava Follis
- Department of Structural Biology, St. Jude Children's Research Hospital , Memphis, Tennessee 38105, United States
| | - Richard W Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital , Memphis, Tennessee 38105, United States.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center , Memphis, Tennessee 38163, United States
| | - Julie D Forman-Kay
- Molecular Structure & Function, The Hospital for Sick Children , Toronto, ON M5G 0A4, Canada.,Department of Biochemistry, University of Toronto , Toronto, ON M5S 1A8, Canada
| |
Collapse
|
172
|
Shih RH, Wang CY, Yang CM. NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review. Front Mol Neurosci 2015; 8:77. [PMID: 26733801 PMCID: PMC4683208 DOI: 10.3389/fnmol.2015.00077] [Citation(s) in RCA: 627] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022] Open
Abstract
The NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells) transcription factor family is a pleiotropic regulator of many cellular signaling pathways, providing a mechanism for the cells in response to a wide variety of stimuli linking to inflammation. The stimulated cells will be regulated by not only the canonical but also non-canonical NF-κB pathways. To initiate both of these pathways, IκB-degradation triggers NF-κB release and the nuclear translocated-heterodimer (or homodimer) can associate with the κB sites of promoter to regulate the gene transcriptions. NF-κB ubiquitously expresses in neurons and the constitutive NF-κB activation is associated with processing of neuronal information. NF-κB can regulate the transcription of genes such as chemokines, cytokines, proinflammatory enzymes, adhesion molecules, proinflammatory transcription factors, and other factors to modulate the neuronal survival. In neuronal insult, NF-κB constitutively active in neuron cell bodies can protect neurons against different injuries and regulate the neuronal inflammatory reactions. Besides neurons, NF-κB transcription factors are abundant in glial cells and cerebral blood vessels and the diverse functions of NF-κB also regulate the inflammatory reaction around the neuronal environment. NF-κB transcription factors are abundant in the brain and exhibit diverse functions. Several central nerve system (CNS) diseases are linked to NF-κB activated by inflammatory mediators. The RelA and c-Rel expression produce opposite effects on neuronal survival. Importantly, c-Rel expression in CNS plays a critical role in anti-apoptosis and reduces the age-related behaviors. Moreover, the different subunits of NF-κB dimer formation can modulate the neuroninflammation, neuronal protection, or neurotoxicity. The diverse functions of NF-κB depend on the subunits of the NF-κB dimer-formation which enable us to develop a therapeutic approach to neuroinflammation based on a new concept of inflammation as a strategic tool in neuronal cells. However, the detail role of NF-κB in neuroinflammation, remains to be clarified. In the present article, we provide an updated review of the current state of our knowledge about relationship between NF-κB and neuroinflammation.
Collapse
Affiliation(s)
- Ruey-Horng Shih
- Institute of Neuroscience, National Chengchi University Taipei, Taiwan
| | - Chen-Yu Wang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University Tao-Yuan, Taiwan
| |
Collapse
|
173
|
Devarie-Baez NO, Silva Lopez EI, Furdui CM. Biological chemistry and functionality of protein sulfenic acids and related thiol modifications. Free Radic Res 2015; 50:172-94. [PMID: 26340608 DOI: 10.3109/10715762.2015.1090571] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Selective modification of proteins at cysteine residues by reactive oxygen, nitrogen or sulfur species formed under physiological and pathological states is emerging as a critical regulator of protein activity impacting cellular function. This review focuses primarily on protein sulfenylation (-SOH), a metastable reversible modification connecting reduced cysteine thiols to many products of cysteine oxidation. An overview is first provided on the chemistry principles underlining synthesis, stability and reactivity of sulfenic acids in model compounds and proteins, followed by a brief description of analytical methods currently employed to characterize these oxidative species. The following chapters present a selection of redox-regulated proteins for which the -SOH formation was experimentally confirmed and linked to protein function. These chapters are organized based on the participation of these proteins in the regulation of signaling, metabolism and epigenetics. The last chapter discusses the therapeutic implications of altered redox microenvironment and protein oxidation in disease.
Collapse
Affiliation(s)
- Nelmi O Devarie-Baez
- a Department of Internal Medicine, Section on Molecular Medicine , Wake Forest School of Medicine , Winston-Salem , NC , USA
| | - Elsa I Silva Lopez
- a Department of Internal Medicine, Section on Molecular Medicine , Wake Forest School of Medicine , Winston-Salem , NC , USA
| | - Cristina M Furdui
- a Department of Internal Medicine, Section on Molecular Medicine , Wake Forest School of Medicine , Winston-Salem , NC , USA
| |
Collapse
|
174
|
Mondragón E, Maher LJ. Anti-Transcription Factor RNA Aptamers as Potential Therapeutics. Nucleic Acid Ther 2015; 26:29-43. [PMID: 26509637 PMCID: PMC4753637 DOI: 10.1089/nat.2015.0566] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factors (TFs) are DNA-binding proteins that play critical roles in regulating gene expression. These proteins control all major cellular processes, including growth, development, and homeostasis. Because of their pivotal role, cells depend on proper TF function. It is, therefore, not surprising that TF deregulation is linked to disease. The therapeutic drug targeting of TFs has been proposed as a frontier in medicine. RNA aptamers make interesting candidates for TF modulation because of their unique characteristics. The products of in vitro selection, aptamers are short nucleic acids (DNA or RNA) that bind their targets with high affinity and specificity. Aptamers can be expressed on demand from transgenes and are intrinsically amenable to recognition by nucleic acid-binding proteins such as TFs. In this study, we review several natural prokaryotic and eukaryotic examples of RNAs that modulate the activity of TFs. These examples include 5S RNA, 6S RNA, 7SK, hepatitis delta virus-RNA (HDV-RNA), neuron restrictive silencer element (NRSE)-RNA, growth arrest-specific 5 (Gas5), steroid receptor RNA activator (SRA), trophoblast STAT utron (TSU), the 3' untranslated region of caudal mRNA, and heat shock RNA-1 (HSR1). We then review examples of unnatural RNA aptamers selected to inhibit TFs nuclear factor-kappaB (NF-κB), TATA-binding protein (TBP), heat shock factor 1 (HSF1), and runt-related transcription factor 1 (RUNX1). The field of RNA aptamers for DNA-binding proteins continues to show promise.
Collapse
Affiliation(s)
- Estefanía Mondragón
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
175
|
Kim HS, Shin MJ, Lee B, Oh KS, Choo H, Pae AN, Roh EJ, Nam G. Synthesis and Biological Evaluation of 2-Phenylimino-5((5-phenylfuran-2-yl)methylene)thiazolidin-4-ones as IKK2 Inhibitors. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hee Sook Kim
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
| | - Min Jae Shin
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
| | - Byungho Lee
- School of Science; University of Science and Technology; Daejeon 305-333 Korea
| | - Kwang-Seok Oh
- School of Science; University of Science and Technology; Daejeon 305-333 Korea
| | - Hyunah Choo
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
- Department of Biological Chemistry; Korea University of Science and Technology (UST); Daejeon 305-350 Korea
| | - Ae Nim Pae
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
- Department of Biological Chemistry; Korea University of Science and Technology (UST); Daejeon 305-350 Korea
| | - Eun Joo Roh
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
- Department of Biological Chemistry; Korea University of Science and Technology (UST); Daejeon 305-350 Korea
| | - Ghilsoo Nam
- Center for Neuro-Medicine, Brain Science Institute; Korea Institutes of Science and Technology (KIST); Seoul 136-791 Korea
- Department of Biological Chemistry; Korea University of Science and Technology (UST); Daejeon 305-350 Korea
| |
Collapse
|
176
|
Activating transcription factor 3 represses inflammatory responses by binding to the p65 subunit of NF-κB. Sci Rep 2015; 5:14470. [PMID: 26412238 PMCID: PMC4585983 DOI: 10.1038/srep14470] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/28/2015] [Indexed: 12/14/2022] Open
Abstract
Activating transcription factor 3 (ATF3) is induced by inflammatory responses, cell death, cytokines, and oxidative stress conditions. ATF3 is a negative regulator in the Toll-like receptor 4 signalling pathway. The principal molecule in this pathway is nuclear factor κB (NF-κB) that translocates into the nucleus to initiate the transcription of inflammatory mediators. However, scarce data are available regarding the interaction of ATF3 and p65, a part of the NF-κB dimer. Therefore, we studied the mechanism of regulation of p65 by ATF3 in RAW 264.7 cells. First, LPS-mediated NF-κB activation was confirmed, and then the direct interaction of ATF3 and p65 was observed through immunoprecipitation (IP). The presence of histone deacetylase 1 (HDAC1) was also detected in the complex. In ATF3 deficient cells, NF-κB activity was up-regulated and HDAC1 was not detected by IP. These observations suggest that p65 is attenuated by ATF3 such that ATF3 recruits HDAC1 to the ATF3/p65 complex and facilitates the deacetylation of p65. Likewise, inflammatory response genes were induced by translocated NF-κB in ATF3-deficient cells. Cumulatively, we uncovered a novel mechanism for the negative regulation of NF-κB by ATF3 via direct interaction with p65.
Collapse
|
177
|
Pyrroloquinoline Quinone Decelerates Rheumatoid Arthritis Progression by Inhibiting Inflammatory Responses and Joint Destruction via Modulating NF-κB and MAPK Pathways. Inflammation 2015; 39:248-256. [DOI: 10.1007/s10753-015-0245-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
178
|
Rajmani RS, Gandham RK, Gupta SK, Sahoo AP, Singh PK, Saxena S, Kumar R, Chaturvedi U, Tiwari AK. Administration of IκB-kinase inhibitor PS1145 enhances apoptosis in DMBA-induced tumor in male Wistar rats. Cell Biol Int 2015; 39:1317-28. [DOI: 10.1002/cbin.10510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Affiliation(s)
- R. S. Rajmani
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ravi Kumar Gandham
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shishir Kumar Gupta
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Aditya P. Sahoo
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Prafull Kumar Singh
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shikha Saxena
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Rajiv Kumar
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Uttara Chaturvedi
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ashok K. Tiwari
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| |
Collapse
|
179
|
Xu K, Harrison RE. Down-regulation of Stathmin Is Required for the Phenotypic Changes and Classical Activation of Macrophages. J Biol Chem 2015; 290:19245-60. [PMID: 26082487 PMCID: PMC4521045 DOI: 10.1074/jbc.m115.639625] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
Macrophages are important cells of innate immunity with specialized capacity for recognition and elimination of pathogens and presentation of antigens to lymphocytes for adaptive immunity. Macrophages become activated upon exposure to pro-inflammatory cytokines and pathogenic stimuli. Classical activation of macrophages with interferon-γ (IFNγ) and lipopolysaccharide (LPS) triggers a wide range of signaling events and morphological changes to induce the immune response. Our previous microtubule (MT) proteomic work revealed that the stathmin association with MTs is considerably reduced in activated macrophages, which contain significantly more stabilized MTs. Here, we show that there is a global decrease in stathmin levels, an MT catastrophe protein, in activated macrophages using both immunoblotting and immunofluorescent microscopy. This is an LPS-specific response that induces proteasome-mediated degradation of stathmin. We explored the functions of stathmin down-regulation in activated macrophages by generating a stable cell line overexpressing stathmin-GFP. We show that stathmin-GFP overexpression impacts MT stability, impairs cell spreading, and reduces activation-associated phenotypes. Furthermore, overexpressing stathmin reduces complement receptor 3-mediated phagocytosis and cellular activation, implicating a pivotal inhibitory role for stathmin in classically activated macrophages.
Collapse
Affiliation(s)
- Kewei Xu
- From the Departments of Cell and Systems Biology and Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Rene E Harrison
- From the Departments of Cell and Systems Biology and Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| |
Collapse
|
180
|
Yu OM, Brown JH. G Protein-Coupled Receptor and RhoA-Stimulated Transcriptional Responses: Links to Inflammation, Differentiation, and Cell Proliferation. Mol Pharmacol 2015; 88:171-80. [PMID: 25904553 PMCID: PMC4468647 DOI: 10.1124/mol.115.097857] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/22/2015] [Indexed: 01/06/2023] Open
Abstract
The low molecular weight G protein RhoA (rat sarcoma virus homolog family member A) serves as a node for transducing signals through G protein-coupled receptors (GPCRs). Activation of RhoA occurs through coupling of G proteins, most prominently, G12/13, to Rho guanine nucleotide exchange factors. The GPCR ligands that are most efficacious for RhoA activation include thrombin, lysophosphatidic acid, sphingosine-1-phosphate, and thromboxane A2. These ligands also stimulate proliferation, differentiation, and inflammation in a variety of cell and tissues types. The molecular events underlying these responses are the activation of transcription factors, transcriptional coactivators, and downstream gene programs. This review describes the pathways leading from GPCRs and RhoA to the regulation of activator protein-1, NFκB (nuclear factor κ-light-chain-enhancer of activated B cells), myocardin-related transcription factor A, and Yes-associated protein. We also focus on the importance of two prominent downstream transcriptional gene targets, the inflammatory mediator cyclooxygenase 2, and the matricellular protein cysteine-rich angiogenic inducer 61 (CCN1). Finally, we describe the importance of GPCR-induced activation of these pathways in the pathophysiology of cancer, fibrosis, and cardiovascular disease.
Collapse
Affiliation(s)
- Olivia M Yu
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| | - Joan Heller Brown
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| |
Collapse
|
181
|
Wang Z, Zhang Z, Yuan J, Li LI. Altered TNFAIP3 mRNA expression in peripheral blood mononuclear cells from patients with rheumatoid arthritis. Biomed Rep 2015; 3:675-680. [PMID: 26405544 DOI: 10.3892/br.2015.486] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/21/2015] [Indexed: 12/29/2022] Open
Abstract
The tumor necrosis factor α-induced protein-3 (TNFAIP3) gene functions in negative immunoregulation and its single-nucleotide polymorphisms (SNPs) are associated with rheumatoid arthritis (RA) disease. However, its expression level in immune cells from RA patients remains unclear. The aim of the present study was to investigate whether the expression of TNFAIP3 is changed in patients with RA. Reverse transcription-quantitative polymerase chain reaction analysis was used to determine TNFAIP3 mRNA expression in peripheral blood mononuclear cells (PBMCs) from patients with RA and healthy controls. TNFAIP3 expression was decreased in RA patients compared with the healthy controls. The expression level of the TNFAIP3 gene negatively correlated with the RA score, anti-cyclic citrullinated peptide (CCP) antibody levels and C-reactive protein levels. Furthermore, RA patients with positive results of anti-CCP antibodies had a lower expression of TNFAIP3 than those without anti-CCP antibodies. In conclusion, the present results suggest that the insufficient expression of the TNFAIP3 gene in PBMCs may correlate with the diagnosis of RA.
Collapse
Affiliation(s)
- Zhaoyan Wang
- Clinical Laboratory, Qingdao Municipal Hospital, Qingdao, Shandong 266011, P.R. China
| | - Zongliang Zhang
- Department of Urology, Qingdao Municipal Hospital, Qingdao, Shandong 266011, P.R. China
| | - Jiangshui Yuan
- Clinical Laboratory, Qingdao Municipal Hospital, Qingdao, Shandong 266011, P.R. China
| | - L I Li
- Clinical Laboratory, Qingdao Municipal Hospital, Qingdao, Shandong 266011, P.R. China
| |
Collapse
|
182
|
Subhash VV, Ho B. Inflammation and proliferation - a causal event of host response to Helicobacter pylori infection. MICROBIOLOGY (READING, ENGLAND) 2015; 161:1150-1160. [PMID: 25721850 DOI: 10.1099/mic.0.000066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Helicobacter pylori is a major aetiological agent in the development of various gastroduodenal diseases. Its persistence in gastric mucosa is determined by the interaction between various host, microbial and environmental factors. The bacterium colonizes the gastric epithelium and induces activation of various chemokine mediators, including NFκB, the master regulator of inflammation. H. pylori infection is also associated with an increase in expression of cell cycle regulators, thereby leading to mucosal cell hyper-proliferation. Thus, H. pylori-associated infections manifest activation of key host response events, which inadvertently could lead to the establishment of chronic infection and neoplastic progression. This article reviews and elaborates the current knowledge in H. pylori-induced activation of various host signalling pathways that could promote cancer development. Special focus is placed on the inflammatory and proliferative responses that could serve as suitable biomarkers of infection, since a sustained cell proliferation in an environment rich in inflammatory cells is characteristic in H. pylori-associated gastric malignancies. Here, the role of ERK and WNT signalling in H. pylori-induced activation of inflammatory and proliferative responses respectively is discussed in detail. An in depth analysis of the underlying signalling pathways and interacting partners causing alterations in these crucial host responses could contribute to the development of successful therapeutic strategies for the prevention, management and treatment of H. pylori infection.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| | - Bow Ho
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| |
Collapse
|
183
|
Esposito E, Sticozzi C, Ravani L, Drechsler M, Muresan XM, Cervellati F, Cortesi R, Valacchi G. Effect of new curcumin-containing nanostructured lipid dispersions on human keratinocytes proliferative responses. Exp Dermatol 2015; 24:449-54. [DOI: 10.1111/exd.12696] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Elisabetta Esposito
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Claudia Sticozzi
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Laura Ravani
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Markus Drechsler
- Macromolecular Chemistry II; University of Bayreuth; Bayreuth Germany
| | - Ximena M. Muresan
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Franco Cervellati
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Rita Cortesi
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| | - Giuseppe Valacchi
- Department of Life Sciences and Biotechnology; University of Ferrara; Ferrara Italy
| |
Collapse
|
184
|
Shihab PK, Al-Roub A, Al-Ghanim M, Al-Mass A, Behbehani K, Ahmad R. TLR2 and AP-1/NF-kappaB are involved in the regulation of MMP-9 elicited by heat killed Listeria monocytogenes in human monocytic THP-1 cells. JOURNAL OF INFLAMMATION-LONDON 2015; 12:32. [PMID: 25931987 PMCID: PMC4415258 DOI: 10.1186/s12950-015-0077-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 03/09/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND MMP-9 is crucial for a normal immune response, but excessive release of this enzyme leads to severe tissue damage. Listeria monocytogenes (LM) is an opportunistic food-borne pathogen causing listerosis, meningitis and sepsis. Heat killed Listeria monocytogenes (HKLM) activates immune system and leads production of cytokines and chemokines. However, nothing is known about the involvement of HKLM in MMP-9 regulation. Therefore we investigated the role of HKLM in the regulation of MMP-9 gene expression in THP-1 cells. METHODS Commercially available heat killed Listeria monocytogenes was used in this study. HKLM-induced MMP-9 expression was assessed with quantitative real-time qPCR and ELISA. Action of HKLM in different signaling pathways were studied by using THP-1-XBlue™ cells (THP-1-cells with NF-κB/AP-1 reporter construct), THP-1-XBlue™-defMyD cells (MyD88(-/-) THP-1 cells), anti-TLR2 mAb and pharmacological inhibitors. Phospho and total proteins were determined by Western blotting. RESULTS Increased MMP-9 production (mRNA: 395-Fold; Protein: 8141 pg/ml; P < 0.05) was observed in HKLM stimulated THP-1 cells as compared to the un-stimulated THP-1 cells. This production of MMP-9 was completely abrogated by anti-TLR2 blocking mAb (P = 0.0024). Furthermore, THP-1-XBlue™-defMyD cells were unable to produce MMP-9 in response to HKLM. HKLM- induced activation of NF-kappaB/AP-1 was also observed in THP-1-XBlue™ Cells. In addition, inhibitors of JNK (SP600125), MEK/ERK (U0126; PD98056), p38 MAPK (SB203580) and NF-kappaB (BAY 11-7085, Triptolide and Resveratrol) significantly suppressed (P < 0.05) HKLM-stimulated MMP-9 expression. CONCLUSION Our results indicate that HKLM activates TLR2 and NF-κB/AP-1 signaling pathways, leading to up-regulation of MMP-9 production in THP-1 cells. Thus, MMP-9 could be an appropriate therapeutic target to stop severe tissue damage caused by infection or chronic inflammation.
Collapse
Affiliation(s)
- Puthiyaveetil Kochumon Shihab
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Areej Al-Roub
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Moneera Al-Ghanim
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Anfal Al-Mass
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Kazem Behbehani
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| | - Rasheed Ahmad
- Immunology & Innovative Cell therapy Unit, Dasman Diabetes Institute, Al-Soor Street, P.O. Box 1180, Dasman, 15462 Kuwait
| |
Collapse
|
185
|
Singh V, Gupta D, Arora R. NF-kB as a key player in regulation of cellular radiation responses and identification of radiation countermeasures. Discoveries (Craiova) 2015; 3:e35. [PMID: 32309561 PMCID: PMC7159829 DOI: 10.15190/d.2015.27] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor (NF)-κB is a transcription factor that plays significant role in immunity, cellular survival and inhibition of apoptosis, through the induction of genetic networks. Depending on the stimulus and the cell type, the members of NF-κB related family (RelA, c-Rel, RelB, p50, and p52), forms different combinations of homo and hetero-dimers. The activated complexes (Es) translocate into the nucleus and bind to the 10bp κB site of promoter region of target genes in stimulus specific manner. In response to radiation, NF-κB is known to reduce cell death by promoting the expression of anti-apoptotic proteins and activation of cellular antioxidant defense system. Constitutive activation of NF-κB associated genes in tumour cells are known to enhance radiation resistance, whereas deletion in mice results in hypersensitivity to IR-induced GI damage. NF-κB is also known to regulate the production of a wide variety of cytokines and chemokines, which contribute in enhancing cell proliferation and tissue regeneration in various organs, such as the GI crypts stem cells, bone marrow etc., following exposure to IR. Several other cytokines are also known to exert potent pro-inflammatory effects that may contribute to the increase of tissue damage following exposure to ionizing radiation. Till date there are a series of molecules or group of compounds that have been evaluated for their radio-protective potential, and very few have reached clinical trials. The failure or less success of identified agents in humans could be due to their reduced radiation protection efficacy.
In this review we have considered activation of NF-κB as a potential marker in screening of radiation countermeasure agents (RCAs) and cellular radiation responses. Moreover, we have also focused on associated mechanisms of activation of NF-κB signaling and their specified family member activation with respect to stimuli. Furthermore, we have categorized their regulated gene expressions and their function in radiation response or modulation. In addition, we have discussed some recently developed radiation countermeasures in relation to NF-κB activation
Collapse
Affiliation(s)
- Vijay Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Damodar Gupta
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Rajesh Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
186
|
Zhou J, Ching YQ, Chng WJ. Aberrant nuclear factor-kappa B activity in acute myeloid leukemia: from molecular pathogenesis to therapeutic target. Oncotarget 2015; 6:5490-5500. [PMID: 25823927 PMCID: PMC4467382 DOI: 10.18632/oncotarget.3545] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/15/2015] [Indexed: 02/07/2023] Open
Abstract
The overall survival of patients with acute myeloid leukemia (AML) has not been improved significantly over the last decade. Molecularly targeted agents hold promise to change the therapeutic landscape in AML. The nuclear factor kappa B (NF-κB) controls a plethora of biological process through switching on and off its long list of target genes. In AML, constitutive NF-κB has been detected in 40% of cases and its aberrant activity enable leukemia cells to evade apoptosis and stimulate proliferation. These facts suggest that NF-κB signaling pathway plays a fundamental role in the development of AML and it represents an attractive target for the intervention of AML. This review summarizes our current knowledge of NF-κB signaling transduction including canonical and non-canonical NF-κB pathways. Then we specifically highlight what factors contribute to the aberrant activation of NF-κB activity in AML, followed by an overview of 8 important clinical trials of the first FDA approved proteasome inhibitor, Bortezomib (Velcade), which is a NF-κB inhibitor too, in combination with other therapeutic agents in patients with AML. Finally, this review discusses the future directions of NF-κB inhibitor in treatment of AML, especially in targeting leukemia stem cells (LSCs).
Collapse
Affiliation(s)
- Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
| | - Ying Qing Ching
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, Republic of Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), Singapore, Republic of Singapore
| |
Collapse
|
187
|
Cho YC, Ju A, Kim BR, Cho S. Anti-inflammatory effects of Crataeva nurvala Buch. Ham. are mediated via inactivation of ERK but not NF-κB. JOURNAL OF ETHNOPHARMACOLOGY 2015; 162:140-147. [PMID: 25571844 DOI: 10.1016/j.jep.2014.12.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 12/02/2014] [Accepted: 12/24/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Crataeva nurvala Buch. Ham. is an important medicinal plant in India, and its extracts and components were used to treat various inflammatory diseases, such as urinary tract infection, rheumatoid arthritis, and colitis. However, no systemic studies about anti-inflammatory effects of Crataeva nurvala Buch. Ham. and its underlying mechanisms of action have been reported. This study aimed to explore the anti-inflammatory effects of ethanol extracts of Crataeva nurvala Buch. Ham. (ECN). MATERIALS AND METHODS The non-cytotoxic and maximal effective concentration of ECN was determined by measuring the formation of formazan from water-soluble tetrazolium salt in living cells. The inhibitory effect of ECN on nitric oxide (NO) synthesis was measured using Griess reagent, and Enzyme-linked immunosorbent assay (ELISA) was used to measure secreted tumor necrosis factor (TNF)-α and interleukin (IL)-6 protein levels. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) and Western blotting analysis were used to assess the mRNA and protein expression of each inflammatory mediator or relating signaling protein, respectively. RESULTS A non-cytotoxic concentration of ECN (≤200 μg/ml) significantly reduced the production of NO and IL-6, but not TNF-α, in lipopolysaccharides (LPS)-stimulated RAW 264.7 macrophages. Decreased production of NO by ECN was correlated with reduced expression of iNOS at the mRNA and protein levels. However, cyclooxygenase (COX)-2 expressions at mRNA and protein level were not regulated by ECN. The mRNA expression of IL-6 and IL-1β, but not TNF-α, was also inhibited by ECN treatment in LPS-stimulated RAW 264.7 macrophages. Reduced production of inflammatory mediators by ECN was followed by decreased activity of mitogen-activated protein kinases (MAPKs), especially extracellular signal-regulated kinase (ERK), but not nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). CONCLUSIONS These results indicate that ECN inhibits LPS-induced inflammatory responses via negative regulation of ERK in murine macrophages, suggesting that ECN is a candidate for alleviating severe inflammation.
Collapse
Affiliation(s)
- Young-Chang Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Anna Ju
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Ba Reum Kim
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Sayeon Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
188
|
Grodzki ACG, Poola B, Pasupuleti N, Nantz MH, Lein PJ, Gorin F. A novel carboline derivative inhibits nitric oxide formation in macrophages independent of effects on tumor necrosis factor α and interleukin-1β expression. J Pharmacol Exp Ther 2015; 352:438-47. [PMID: 25538105 PMCID: PMC4352593 DOI: 10.1124/jpet.114.220186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/19/2014] [Indexed: 12/19/2022] Open
Abstract
Neuropathic pain is a maladaptive immune response to peripheral nerve injury that causes a chronic painful condition refractory to most analgesics. Nitric oxide (NO), which is produced by nitric oxide synthases (NOSs), has been implicated as a key factor in the pathogenesis of neuropathic pain. β-Carbolines are a large group of natural and synthetic indole alkaloids, some of which block activation of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), a predominant transcriptional regulator of NOS expression. Here, we characterize the inhibitory effects of a novel 6-chloro-8-(glycinyl)-amino-β-carboline (8-Gly carb) on NO formation and NF-κB activation in macrophages. 8-Gly carb was significantly more potent than the NOS inhibitor NG-nitro-L-arginine methyl ester in inhibiting constitutive and inducible NO formation in primary rat macrophages. 8-Gly carb interfered with NF-κB-mediated gene expression in differentiated THP1-XBlue cells, a human NF-κB reporter macrophage cell line, but only at concentrations severalfold higher than needed to significantly inhibit NO production. 8-Gly carb also had no effect on tumor necrosis factor α (TNFα)-induced phosphorylation of the p38 mitogen-activated protein kinase in differentiated THP1 cells, and did not inhibit lipopolysaccharide- or TNFα-stimulated expression of TNFα and interleukin-1β. These data demonstrate that relative to other carbolines and pharmacologic inhibitors of NOS, 8-Gly carb exhibits a unique pharmacological profile by inhibiting constitutive and inducible NO formation independent of NF-κB activation and cytokine expression. Thus, this novel carboline derivative holds promise as a parent compound, leading to therapeutic agents that prevent the development of neuropathic pain mediated by macrophage-derived NO without interfering with cytokine expression required for neural recovery following peripheral nerve injury.
Collapse
Affiliation(s)
- Ana Cristina G Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| | - Bhaskar Poola
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| | - Nagarekha Pasupuleti
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| | - Michael H Nantz
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| | - Fredric Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine (A.C.G.G., F.G., P.J.L.), and Department of Neurology, School of Medicine (N.P., F.G.), University of California, Davis, California; and Department of Chemistry, University of Louisville, Louisville, Kentucky (B.P., M.H.N.)
| |
Collapse
|
189
|
Ge H, Yuan W, Liu J, He Q, Ding S, Pu J, He B. Functional relevance of protein glycosylation to the pro-inflammatory effects of extracellular matrix metalloproteinase inducer (EMMPRIN) on monocytes/macrophages. PLoS One 2015; 10:e0117463. [PMID: 25658763 PMCID: PMC4319903 DOI: 10.1371/journal.pone.0117463] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/23/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Extracellular matrix metalloproteinase inducer (EMMPRIN) is an important pro-inflammatory protein involved in the cellular functions of monocytes/macrophages. We have hypothesized that high-level heterogeneousness of protein glycosylation of EMMPRIN may have functional relevance to its biological effects and affect the inflammatory activity of monocytes/macrophages. METHODS The glycosylation patterns of EMMPRIN expressed by monocytes/macrophages (THP-1 cells) in response to different extracellular stimuli were observed, and the structures of different glycosylation forms were identified. After the purification of highly- and less-glycosylated proteins respectively, the impacts of different glycosylation forms on the pro-inflammatory effects of EMMPRIN were examined in various aspects, such as cell adhesion to endothelial cells, cell migrations, cytokine expression, and activation of inflammatory signalling pathway. RESULTS 1) It was mainly the highly-glycosylated form of EMMPRIN (HG-EMMPRIN) that increased after being exposed to inflammatory signals (PMA and H2O2). 2) Glycosylation of EMMPRIN in monocytes/macrophages led to N-linked-glycans being added to the protein, with the HG form containing complex-type glycans and the less-glycosylated form (LG) the simple type. 3) Only the HG-EMMPRIN but not the LG-EMMPRIN exhibited pro-inflammatory effects and stimulated inflammatory activities of the monocytes/macrophages (i.e., activation of ERK1/2 and NF-κB pathway, enhanced monocyte-endothelium adhesion, cell migration and matrix metalloproteinase -9 expression). CONCLUSIONS Post-transcriptional glycosylation represents an important mechanism that determines the biological effects of EMMPRIN in monocytes/macrophages. Glycosylation of EMMPRIN may serve as a potential target for regulating the inflammatory activities of monocytes/macrophages.
Collapse
Affiliation(s)
- Heng Ge
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Yuan
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jidong Liu
- Department of Cardiac Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing He
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Song Ding
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ben He
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
190
|
Wang Y, Wei H, Wang X, Du L, Zhang A, Zhou H. Cellular activation, expression analysis and functional characterization of grass carp IκBα: evidence for its involvement in fish NF-κB signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2015; 42:408-412. [PMID: 25434741 DOI: 10.1016/j.fsi.2014.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 06/04/2023]
Abstract
IκBα is a well-known member of the inhibitors of kappa B (IκB) family that controls NF-κB signaling by blocking NF-κB translocation from cytoplasm to nucleus. In the present study, an IκBα homologue was identified from grass carp (gcIκBα), showing the structural characteristics of IκB family. Moreover, mRNA expression of this molecule in grass carp periphery blood lymphocytes (PBLs) was enhanced significantly by both LPS and PHA in a time- and dose-dependent manner, indicating the involvement of gcIκBα in fish immune response. Further analysis demonstrated that LPS but not PHA induced gcIκBα phosphorylation and protein degradation in PBLs, implying different signaling pathways mediated by LPS and PHA in gcIκBα expression regulation in grass carp PBLs. In particular, the time-dependent oscillation of gcIκBα phosphorylation and total protein levels induced by LPS is in accordance with the characteristics of mammalian IκBα phosphorylation followed by protein degradation during NF-κB activation. In support of this notion, overexpression of gcIκBα was able to block both basal and LPS-stimulated NF-κB activity in grass carp kidney cell line, indicating the negatively regulatory role of gcIκBα in NF-κB activity as seen in mammals. Therefore, our results not only reveal a dynamic variation of NF-κB activity based on the activation and expression of IκBα for the first time, but also provide the direct evidence for the involvement of IκBα in NF-κB signaling in fish immune cells.
Collapse
Affiliation(s)
- Yanan Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - He Wei
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Xinyan Wang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Linyong Du
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Anying Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
191
|
Arepalli SK, Choi M, Jung JK, Lee H. Novel NF-κB inhibitors: a patent review (2011 – 2014). Expert Opin Ther Pat 2015; 25:319-34. [DOI: 10.1517/13543776.2014.998199] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
192
|
Khalil H, Loukili N, Regamey A, Cuesta-Marban A, Santori E, Huber M, Widmann C. The caspase-3/p120 RasGAP module generates a NF-κB repressor in response to cellular stress. J Cell Sci 2015. [DOI: 10.1242/jcs.174409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The NF-κB transcription factor is a master regulator of inflammation. Short-term NF-κB activation is generally beneficial. However, sustained NF-κB may be detrimental, directly causing apoptosis of cells or leading to a persistent damaging inflammatory response. NF-κB activity in stressed cells needs therefore to be controlled for homeostasis maintenance. Here we show that fragment N that is produced by the caspase-3/p120 RasGAP sensor in mildly stressed cells is a potent NF-κB inhibitor. Fragment N decreases the transcriptional activity of NF-κB by promoting its export from the nucleus. Cells unable to generate fragment N displayed increased NF-κB activation upon stress. Knock-in mice expressing the uncleavable RasGAP mutant showed exaggerated NF-κB activation when their epidermis was treated with anthralin, a drug used for the treatment of psoriasis. Our study provides biochemical and genetic evidence of the importance of the caspase-3/p120 RasGAP stress-sensing module in the control of stress-induced NF-κB activation.
Collapse
Affiliation(s)
- Hadi Khalil
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Noureddine Loukili
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Alexandre Regamey
- Department of Dermatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Alvaro Cuesta-Marban
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Elettra Santori
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Marcel Huber
- Department of Dermatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| |
Collapse
|
193
|
Zhou HR, He K, Landgraf J, Pan X, Pestka JJ. Direct activation of ribosome-associated double-stranded RNA-dependent protein kinase (PKR) by deoxynivalenol, anisomycin and ricin: a new model for ribotoxic stress response induction. Toxins (Basel) 2014; 6:3406-25. [PMID: 25521494 PMCID: PMC4280541 DOI: 10.3390/toxins6123406] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/29/2014] [Accepted: 12/08/2014] [Indexed: 01/04/2023] Open
Abstract
Double-stranded RNA (dsRNA)-activated protein kinase (PKR) is a critical upstream mediator of the ribotoxic stress response (RSR) to the trichothecene deoxynivalenol (DON) and other translational inhibitors. Here, we employed HeLa cell lysates to: (1) characterize PKR’s interactions with the ribosome and ribosomal RNA (rRNA); (2) demonstrate cell-free activation of ribosomal-associated PKR and (3) integrate these findings in a unified model for RSR. Robust PKR-dependent RSR was initially confirmed in intact cells. PKR basally associated with 40S, 60S, 80S and polysome fractions at molar ratios of 7, 2, 23 and 3, respectively. Treatment of ATP-containing HeLa lysates with DON or the ribotoxins anisomycin and ricin concentration-dependently elicited phosphorylation of PKR and its substrate eIF2α. These phosphorylations could be blocked by PKR inhibitors. rRNA immunoprecipitation (RNA-IP) of HeLa lysates with PKR-specific antibody and sequencing revealed that in the presence of DON or not, the kinase associated with numerous discrete sites on both the 18S and 28S rRNA molecules, a number of which contained double-stranded hairpins. These findings are consistent with a sentinel model whereby multiple PKR molecules basally associate with the ribosome positioning them to respond to ribotoxin-induced alterations in rRNA structure by dimerizing, autoactivating and, ultimately, evoking RSR.
Collapse
Affiliation(s)
- Hui-Ren Zhou
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA.
| | - Kaiyu He
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.
| | - Jeff Landgraf
- Research Technology Support Facility, Michigan State University, East Lansing, MI 48824, USA.
| | - Xiao Pan
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| | - James J Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
194
|
Liu MW, Su MX, Zhang W, Wang YQ, Chen M, Wang L, Qian CY. Protective effect of Xuebijing injection on paraquat-induced pulmonary injury via down-regulating the expression of p38 MAPK in rats. Altern Ther Health Med 2014; 14:498. [PMID: 25511395 PMCID: PMC4301062 DOI: 10.1186/1472-6882-14-498] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
Abstract
Background Exposure to paraquat results in acute lung injury. A systemic inflammatory response has been widely established as a contributor to paraquat-induced acute lung injury. Recent studies have reported that consumption of Xuebijing prevents inflammatory response-induced diseases. This study investigated whether consumption of Xuebijing protected rats against paraquat-induced acute lung injury. Methods Adult male Sprague Dawley rats were randomly divided into four groups: control group; paraquat group; paraquat + Xuebijing group; and paraquat + dexamethasone group. Rats in the paraquat, paraquat + Xuebijing and paraquat + dexamethasone groups were intraperitoneally injected with paraquat (30 mg/kg) or administered paraquat and Xuebijing at 8 mL/kg or dexamethasone at 5 mg/kg, respectively, via an injection into the tail vein. Lung p38 MAPK, NF-κB65, IkB, p-IκB-α, HIF-1α, Nrf2 and TGF-β1 expression were essayed using western blotting. IL-6, TNF-α, IL-1β, IL-10, TGF-β1 and PIIIP were measured using ELISA. ROS, oxidised glutathione and glutathione activity were measured. Results After inducing acute lung injury with paraquat for 24 h, Xuebijing was observed to block lung p-p38 MAPK, NF-κB65, HIF-1α, p-IκB-α and TGF-β1 expression, and increased Nrf2 and IkB expression. The numbers of neutrophils and lymphocytes and total number of cells were significantly lower in the Xuebijing group compared with the control group. IL-6, TNF-α, IL-1β, TGF-β1 and PIIIP levels were significantly decreased in the Xuebijing group. ROS and oxidised glutathione activity were markedly inhibited by Xuebijing. Histological evaluation showed attenuation of the effects of Xuebijing on paraquat-induced lung injury. Compared with the paraquat + dexamethasone group, the Xuebijing + paraquat group showed no significant differences. Conclusions Inhibiting the expression of p38 MAPK and NF-κB65 was crucial for the protective effects of Xuebijing on paraquat-induced acute lung injury. The findings suggest that Xuebijing could effectively ameliorate paraquat-induced acute lung injury in rats. Xuebijing was as effective as dexamethasone at improving paraquat-induced lung injury by regulating lung inflammation, lung function and oxidative stress responses.
Collapse
|
195
|
CPEB regulation of TAK1 synthesis mediates cytokine production and the inflammatory immune response. Mol Cell Biol 2014; 35:610-8. [PMID: 25452303 DOI: 10.1128/mcb.00800-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The cytoplasmic-element-binding (CPEB) protein is a sequence-specific RNA-binding protein that regulates cytoplasmic polyadenylation-induced translation. In mouse embryo fibroblasts (MEFs) lacking CPEB, many mRNAs encoding proteins involved in inflammation are misregulated. Correlated with this aberrant translation in MEFs, a macrophage cell line depleted of CPEB and treated with lipopolysaccharide (LPS) to stimulate the inflammatory immune response expresses high levels of interleukin-6 (IL-6), which is due to prolonged nuclear retention of NF-κB. Two proteins involved in NF-κB nuclear localization and IL-6 expression, IκBα and transforming growth factor beta-activated kinase 1 (TAK1), are present at excessively low and high steady-state levels, respectively, in LPS-treated CPEB-depleted macrophages. However, only TAK1 has an altered synthesis rate that is CPEB dependent and CPEB/TAK1 double depletion alleviates high IL-6 production. Peritoneal macrophages isolated from CPEB knockout (KO) mice treated with LPS in vitro also have prolonged NF-κB nuclear retention and produce high IL-6 levels. LPS-injected CPEB KO mice secrete prodigious amounts of IL-6 and other proinflammatory cytokines and exhibit hypersensitivity to endotoxic shock; these effects are mitigated when the animals are also injected with (5Z)-7-oxozeaenol, a potent and specific inhibitor of TAK1. These data show that CPEB control of TAK1 mRNA translation mediates the inflammatory immune response.
Collapse
|
196
|
Lee Y, Umasuthan N, Whang I, Revathy KS, Lee S, De Zoysa M, Oh C, Kang DH, Noh JK, Lee J. Two NF-κB inhibitor-alpha (IκBα) genes from rock bream (Oplegnathus fasciatus): molecular characterization, genomic organization and mRNA expression analysis after immune stimulation. FISH & SHELLFISH IMMUNOLOGY 2014; 41:633-642. [PMID: 25462458 DOI: 10.1016/j.fsi.2014.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/07/2014] [Accepted: 10/10/2014] [Indexed: 06/04/2023]
Abstract
IkBa is a member of IkB family, which sequesters NF-kB in an inactivate form in the cytoplasm and blocks the translocation of NF-kB to nucleus. The IkBa paralogs of rock bream (OfIkBa-A and OfIkBa-B) encoded IkBa proteins with typical features including, highly conserved IkB degradation motif, six ankyrin repeats and a PEST sequence. However, their amino acid identity and similarity were only 55.6 and 69.7%, respectively suggesting that these two genes could be the two different isoforms of IkBa. The number and size of the exons of OfIkBa-A and OfIkBa-B were conserved well with all the compared vertebrate species, although they have significantly different genomic sizes. Phylogenetic analysis revealed that OfIkBa-A and OfIkBa-B proteins cluster with IkBa family members; however, they were grouped with different subclades in IkBa family. Tissue specific expression of OfIkBa mRNA was constitutively detected in all the tested tissues, and they showed the higher transcription level in heart, liver, gill and peripheral blood cells, respectively. The injection of flagellin stimulated the mRNA expression of OfIkBa paralogs in head kidney and intestine. Moreover, the OfIkBa mRNA expression in gill and liver was significantly upregulated by LPS, poly I:C and Edwardsiella tarda challenges. The transcription of OfIkBa was up-regulated in early-phase of injection and then rapidly restored. These results suggest that the OfIkBa paralogs might be involved in rapid immune responsive reactions in rock bream against bacterial and viral pathogens.
Collapse
Affiliation(s)
- Youngdeuk Lee
- Korea Institute of Ocean Science & Technology, Ansan 426-744, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Yoon CS, Kim DC, Lee DS, Kim KS, Ko W, Sohn JH, Yim JH, Kim YC, Oh H. Anti-neuroinflammatory effect of aurantiamide acetate from the marine fungus Aspergillus sp. SF-5921: Inhibition of NF-κB and MAPK pathways in lipopolysaccharide-induced mouse BV2 microglial cells. Int Immunopharmacol 2014; 23:568-74. [DOI: 10.1016/j.intimp.2014.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 12/15/2022]
|
198
|
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Mitochondrial complex I impairment in PD is modeled in vitro by the susceptibility of dopaminergic neurons to the complex I inhibitor 1-methyl-4-phenylpyridinium (MPP+). In the present study, we demonstrate that microRNA-7 (miR-7), which is expressed in tyrosine hydroxylase-positive nigral neurons in mice and humans, protects cells from MPP+-induced toxicity in dopaminergic SH-SY5Y cells, differentiated human neural progenitor ReNcell VM cells, and primary mouse neurons. RelA, a component of nuclear factor-κB (NF-κB), was identified to be downregulated by miR-7 using quantitative proteomic analysis. Through a series of validation experiments, it was confirmed that RelA mRNA is a target of miR-7 and is required for cell death following MPP+ exposure. Further, RelA mediates MPP+-induced suppression of NF-κB activity, which is essential for MPP+-induced cell death. Accordingly, the protective effect of miR-7 is exerted through relieving NF-κB suppression by reducing RelA expression. These findings provide a novel mechanism by which NF-κB suppression, rather than activation, underlies the cell death mechanism following MPP+ toxicity, have implications for the pathogenesis of PD, and suggest miR-7 as a therapeutic target for this disease.
Collapse
|
199
|
Epidermal RelA Specifically Restricts Contact Allergen–Induced Inflammation and Apoptosis in Skin. J Invest Dermatol 2014; 134:2541-2550. [DOI: 10.1038/jid.2014.193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/18/2014] [Accepted: 04/04/2014] [Indexed: 11/08/2022]
|
200
|
Du MJ, Zhang HK, He AJ, Chang YS, Yang Y, Wang Y, Zhang CZ, Cao Y. Selection of peptide inhibitors for double-stranded RNA-dependent protein kinase PKR. BIOCHEMISTRY (MOSCOW) 2014; 78:1254-62. [PMID: 24460939 DOI: 10.1134/s0006297913110059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Protein kinase inhibitors have been developed and applied as antitumor drugs. The majority of these inhibitors are derived from ATP analogs with limited specificity towards the kinase target. Here we present our proof-of-principle study on peptide inhibitors for kinases. Two peptides were selected by phage display against double-stranded RNA-dependent protein kinase (PKR). In vitro assay revealed that these peptides exhibit an inhibitory effect on PKR-catalyzed phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF2α). The peptides also interrupt PKR activity in cells infected by viruses, as PKR activation is one of the hallmarks of host response to viral infection. Kinetic study revealed that one of the peptides, named P1, is a competitive inhibitor for PKR, while the other, named P2, exhibits a more complicated pattern of inhibition on PKR activity. Fragment-based docking of the PKR-peptide complex suggests that P1 occupies the substrate pocket of PKR and thus inhibits the binding between PKR and eIF2α, whereas P2 sits near the substrate pocket. The computational model of PKR-peptide complex agrees with their kinetic behavior. We surmise that peptide inhibitors for kinases have higher specificity than ATP analogs, and that they provide promising leads for the optimization of kinase inhibitors.
Collapse
Affiliation(s)
- M-J Du
- Key Laboratory of Microbial Functional Genomics of the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|