101
|
MicroRNA-216a Inhibits NF-κB-Mediated Inflammatory Cytokine Production in Teleost Fish by Modulating p65. Infect Immun 2018; 86:IAI.00256-18. [PMID: 29632247 DOI: 10.1128/iai.00256-18] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/08/2023] Open
Abstract
Inflammation is the host self-protection mechanism to eliminate pathogen invasion. The excessive inflammatory response can result in uncontrolled inflammation, autoimmune diseases, or pathogen dissemination. Recent studies have widely shown that microRNAs (miRNAs) contribute to the regulation of inflammation in mammals by repressing gene expression at the posttranscriptional level. However, the miRNA-mediated mechanism in the inflammatory response in fish remains hazy. In the present study, the regulatory mechanism of the miR-216a-mediated inflammatory response in teleost fish was addressed. We found that the expression of miR-216a could be significantly upregulated in the miiuy croaker after challenge with Vibrio anguillarum and lipopolysaccharide. Bioinformatics predictions demonstrated a potential binding site of miR-216a in the 3' untranslated region of the p65 gene, and the result was further confirmed by luciferase assay. Moreover, both the mRNA and protein levels of p65 in macrophages were downregulated by miR-216a. Deletion mutant analysis of the miR-216a promoter showed that the Ap1 and Sp1 transcription factor binding sites are indispensable for the transcription of miR-216a. Further study revealed that overexpression of miR-216a suppresses inflammatory cytokine expression and negatively regulates NF-κB signaling, which inhibit an excessive inflammatory response. The collective results indicate that miR-216a plays a role as a negative regulator involved in modulating the bacterium-induced inflammatory response.
Collapse
|
102
|
Velagapudi R, Ajileye OO, Okorji U, Jain P, Aderogba MA, Olajide OA. Agathisflavone isolated from Anacardium occidentale suppresses SIRT1-mediated neuroinflammation in BV2 microglia and neurotoxicity in APPSwe-transfected SH-SY5Y cells. Phytother Res 2018; 32:1957-1966. [PMID: 29786910 DOI: 10.1002/ptr.6122] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 01/01/2023]
Abstract
Agathisflavone is a bioactive compound in Anacardium occidentale. In this study, we investigated inhibition neuroinflammation in BV2 microglia by agathisflavone. Neuroprotective activity of the compound was investigated in differentiated SH-SY5Y cells. Experiments in lipopolysaccharide (LPS)-activated BV2 microglia showed that pretreatment with agathisflavone (5-20 μM) produced significant reduction in the release of tumour necrosis factor-α, interleukin-6, interleukin-1β, NO, and PGE2 from the cells. Immunoblotting experiments also revealed that agathisflavone reduced levels of iNOS and COX-2 protein. Further studies revealed that agathisflavone reduced neuroinflammation by targeting critical steps in NF-κB signalling in BV2 microglia. Treatment of SH-SY5Y cells with conditioned medium from LPS-activated BV2 microglia produced a significant reduction in neuronal viability. However, conditioned medium from BV2 cells that were stimulated with LPS in the presence of agathisflavone did not induce neurotoxicity. Agathisflavone also produced neuroprotection in APPSwe plasmid-transfected SH-SY5Y neurons. The compound further attenuated LPS-induced and APPSwe plasmid-induced reduction in SIRT1 in BV2 microglia and SH-SY5Y, respectively. In the presence of EX527, agathisflavone lost its anti-inflammatory and neuroprotective activities. Our results suggest that agathisflavone inhibits neuroinflammation in BV2 microglia by targeting NF-κB signalling pathway. The compound also reduces neurotoxicity through mechanisms that are possibly linked to SIRT1 in the microglia and neurons.
Collapse
Affiliation(s)
- Ravikanth Velagapudi
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| | - Olusiji O Ajileye
- Department of Chemistry, Faculty of Science, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Uchechukwu Okorji
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| | - Priya Jain
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| | - Mutalib A Aderogba
- Department of Chemistry, Faculty of Science, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Olumayokun A Olajide
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| |
Collapse
|
103
|
Wei Z, Yoshihara E, He N, Hah N, Fan W, Pinto AFM, Huddy T, Wang Y, Ross B, Estepa G, Dai Y, Ding N, Sherman MH, Fang S, Zhao X, Liddle C, Atkins AR, Yu RT, Downes M, Evans RM. Vitamin D Switches BAF Complexes to Protect β Cells. Cell 2018; 173:1135-1149.e15. [PMID: 29754817 PMCID: PMC5987229 DOI: 10.1016/j.cell.2018.04.013] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/01/2017] [Accepted: 04/11/2018] [Indexed: 12/20/2022]
Abstract
A primary cause of disease progression in type 2 diabetes (T2D) is β cell dysfunction due to inflammatory stress and insulin resistance. However, preventing β cell exhaustion under diabetic conditions is a major therapeutic challenge. Here, we identify the vitamin D receptor (VDR) as a key modulator of inflammation and β cell survival. Alternative recognition of an acetylated lysine in VDR by bromodomain proteins BRD7 and BRD9 directs association to PBAF and BAF chromatin remodeling complexes, respectively. Mechanistically, ligand promotes VDR association with PBAF to effect genome-wide changes in chromatin accessibility and enhancer landscape, resulting in an anti-inflammatory response. Importantly, pharmacological inhibition of BRD9 promotes PBAF-VDR association to restore β cell function and ameliorate hyperglycemia in murine T2D models. These studies reveal an unrecognized VDR-dependent transcriptional program underpinning β cell survival and identifies the VDR:PBAF/BAF association as a potential therapeutic target for T2D.
Collapse
MESH Headings
- Animals
- Calcitriol/analogs & derivatives
- Calcitriol/pharmacology
- Chromatin Assembly and Disassembly
- Chromosomal Proteins, Non-Histone/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Humans
- Insulin/blood
- Insulin/metabolism
- Insulin-Secreting Cells/cytology
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- Mutagenesis, Site-Directed
- Oxidative Phosphorylation/drug effects
- Protein Binding
- RNA Interference
- RNA, Guide, CRISPR-Cas Systems/genetics
- RNA, Small Interfering/metabolism
- Receptors, Calcitriol/antagonists & inhibitors
- Receptors, Calcitriol/genetics
- Receptors, Calcitriol/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic/drug effects
- Vitamin D/pharmacology
Collapse
Affiliation(s)
- Zong Wei
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eiji Yoshihara
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nanhai He
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nasun Hah
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Weiwei Fan
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Timothy Huddy
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yuhao Wang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Brittany Ross
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Gabriela Estepa
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yang Dai
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ning Ding
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mara H Sherman
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sungsoon Fang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Xuan Zhao
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
104
|
Ubale RV, Shastri PN, Oettinger C, D’Souza MJ. Pulmonary Administration of Microparticulate Antisense Oligonucleotide (ASO) for the Treatment of Lung Inflammation. AAPS PharmSciTech 2018; 19:1908-1919. [PMID: 29663290 DOI: 10.1208/s12249-018-1002-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/19/2018] [Indexed: 01/01/2023] Open
Abstract
Targeted delivery to the lung for controlling lung inflammation is an area that we have explored in this study. The purpose was to use microparticles containing an antisense oligonucleotide (ASO) to NF-κB to inhibit the production of proinflammatory cytokines. Microparticles were prepared using the B-290 Buchi Spray Dryer using albumin as the microparticle matrix. Physicochemical characterization of the microparticles showed the size ranged from 2 to 5 μm, the charge was - 38.4 mV, and they had a sustained release profile over 72 h. Uptake of FITC-labeled ASO-loaded microparticles versus FITC-labeled ASO solution by RAW264.7 murine macrophage cells was 5-10-fold higher. After pulmonary delivery of microparticles to Sprague-Dawley rats, the microparticles were uniformly distributed throughout the lung and were retained in the lungs until 48 h. Serum cytokine (TNF-α and IL-1β) levels of rats after induction of lung inflammation by lipopolysaccharide were measured until 72 h. Animals receiving ASO-loaded microparticles were successful in significantly controlling lung inflammation during this period as compared to animals receiving no treatment. This study was successful in proving that microparticulate ASO therapy was capable of controlling lung inflammation.
Collapse
|
105
|
Wu F, Wei X, Wu Y, Kong X, Hu A, Tong S, Liu Y, Gong F, Xie L, Zhang J, Xiao J, Zhang H. Chloroquine Promotes the Recovery of Acute Spinal Cord Injury by Inhibiting Autophagy-Associated Inflammation and Endoplasmic Reticulum Stress. J Neurotrauma 2018; 35:1329-1344. [PMID: 29316847 DOI: 10.1089/neu.2017.5414] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Spinal cord injury (SCI) is a severe nervous system disease that may lead to lifelong disability. Studies have shown that autophagy plays a key role in various diseases; however, the mechanisms regulating cross-talk between autophagy, inflammation, and endoplasmic reticulum (ER) stress during SCI recovery remain unclear. This study was designed to investigate the mechanism by which chloroquine (CQ) inhibits autophagy-associated inflammation and ER stress in rats during their recovery from acute SCI. We evaluated the locomotor function, level of autophagy, and levels of inflammatory cytokines and ER-stress-associated proteins and examined the degradation of the key regulator of inflammation inhibitor of kappa B alpha (I-κBα) through autophagy by analyzing the colocalization of I-κBα, p62, and microtubule-associated protein 1 light chain 3-II. In addition, overexpression of the p62 and activating transcription factor 4 (ATF4) silencing plasmids was used to verify the important roles for autophagic degradation and ER stress. In this study, locomotor function is improved, and autophagy and inflammation are significantly inhibited by, CQ treatment in the model rats. In addition, CQ significantly inhibits the degradation of ubiquitinated I-κBα and blocks the nuclear translocation of nuclear factor kappa B p65 and expression of inflammatory factors. Overexpression of p62 increases I-κBα degradation and improves inflammatory responses. Moreover, CQ treatment also inhibits the activation of ER stress in the rat SCI model, and the ATF4 signaling pathway is required for ER-stress-induced activation of autophagy. These findings reveal a novel mechanism underlying the beneficial effects of CQ on the recovery of SCI, particularly the mechanisms regulating cross-talk between autophagy, inflammation, and ER stress.
Collapse
Affiliation(s)
- Fenzan Wu
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China .,2 Department of Orthopaedics, Cixi People's Hospital, Wenzhou Medical University , Ningbo, China
| | - Xiaojie Wei
- 2 Department of Orthopaedics, Cixi People's Hospital, Wenzhou Medical University , Ningbo, China
| | - Yanqing Wu
- 3 The Institute of Life Sciences, Wenzhou University , Wenzhou, China
| | - Xiaoxia Kong
- 4 Institute of Hypoxia Research, School of Basic Medical Sciences, Wenzhou Medical University , Wenzhou, China
| | - Aiping Hu
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China
| | - Songlin Tong
- 2 Department of Orthopaedics, Cixi People's Hospital, Wenzhou Medical University , Ningbo, China
| | - Yanlong Liu
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China
| | - Fanhua Gong
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China
| | - Ling Xie
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China
| | - Jinjing Zhang
- 2 Department of Orthopaedics, Cixi People's Hospital, Wenzhou Medical University , Ningbo, China
| | - Jian Xiao
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China .,2 Department of Orthopaedics, Cixi People's Hospital, Wenzhou Medical University , Ningbo, China
| | - Hongyu Zhang
- 1 Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University , Wenzhou, China .,5 Science and Research Center, Dongyang People's Hosipital, Wenzhou Medical University , Jinhua, China
| |
Collapse
|
106
|
Chishti AA, Baumstark-Khan C, Koch K, Kolanus W, Feles S, Konda B, Azhar A, Spitta LF, Henschenmacher B, Diegeler S, Schmitz C, Hellweg CE. Linear Energy Transfer Modulates Radiation-Induced NF-kappa B Activation and Expression of its Downstream Target Genes. Radiat Res 2018; 189:354-370. [PMID: 29369006 DOI: 10.1667/rr14905.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Nuclear factor kappaB (NF-κB) is a central transcription factor in the immune system and modulates cell survival in response to radiotherapy. Activation of NF-κB was shown to be an early step in the cellular response to ultraviolet A (UVA) and ionizing radiation exposure in human cells. NF-κB activation by the genotoxic stress-dependent sub-pathway after exposure to different radiation qualities had been evaluated to a very limited extent. In addition, the resulting gene expression profile, which shapes the cellular and tissue response, is unknown. Therefore, in this study the activation of NF-κB after exposure to low- and high-linear energy transfer (LET) radiation and the expression of its target genes were analyzed in human embryonic kidney (HEK) cells. The activation of NF-κB via canonical and genotoxic stress-induced pathways was visualized by the cell line HEK-pNF-κB-d2EGFP/Neo L2 carrying the destabilized enhanced green fluorescent protein (d2EGFP) as reporter. The NF-κB-dependent d2EGFP expression after irradiation with X rays and heavy ions was evaluated by flow cytometry. Because of differences in the extent of NF-κB activation after irradiation with X rays (significant NF-κB activation for doses >4 Gy) and heavy ions (significant NF-κB activation at doses as low as 1 Gy), it was expected that radiation quality (LET) played an important role in the cellular radiation response. In addition, the relative biological effectiveness (RBE) of NF-κB activation and reduction of cellular survival were compared for heavy ions having a broad LET range (∼0.3-9,674 keV/μm). Furthermore, the effect of LET on NF-κB target gene expression was analyzed by real-time reverse transcriptase quantitative PCR (RT-qPCR). The maximal RBE for NF-κB activation and cell killing occurred at an LET value of 80 and 175 keV/μm, respectively. There was a dose-dependent increase in expression of NF-κB target genes NF-κB1A and CXCL8. A qPCR array of 84 NF-κB target genes revealed that TNF and a set of CXCL genes (CXCL1, CXCL2, CXCL8, CXCL10), CCL2, VCAM1, CD83, NF-κB1, NF-κB2 and NF-κBIA were strongly upregulated after exposure to X rays and neon ions (LET 92 keV/μm). After heavy-ion irradiations, it was noted that the expression of NF-κB target genes such as chemokines and CD83 was highest at an LET value that coincided with the LET resulting in maximal NF-κB activation, whereas expression of the NF-κB inhibitory gene NFKBIA was induced transiently by all radiation qualities investigated. Taken together, these findings clearly demonstrate that NF-κB activation and NF-κB-dependent gene expression by heavy ions are highest in the LET range of ∼50-200 keV/μm. The upregulated chemokines and cytokines (CXCL1, CXCL2, CXCL10, CXCL8/IL-8 and TNF) could be important for cell-cell communication among hit as well as nonhit cells (bystander effect).
Collapse
Affiliation(s)
- Arif Ali Chishti
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Christa Baumstark-Khan
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Kristina Koch
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Waldemar Kolanus
- b Life and Medical Sciences (LIMES) Institute, University of Bonn, Karlrobert-Kreiten-Straße 13, 53115 Bonn, Germany
| | - Sebastian Feles
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Bikash Konda
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Abid Azhar
- c The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi-75270, Pakistan
| | - Luis F Spitta
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Bernd Henschenmacher
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Sebastian Diegeler
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Claudia Schmitz
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Christine E Hellweg
- a German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| |
Collapse
|
107
|
Ushio A, Eto K. RBM3 expression is upregulated by NF‐κB p65 activity, protecting cells from apoptosis, during mild hypothermia. J Cell Biochem 2018; 119:5734-5749. [DOI: 10.1002/jcb.26757] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/25/2018] [Indexed: 01/19/2023]
Affiliation(s)
- Ayako Ushio
- Department of Biological SciencesGraduate School of Science and TechnologyKumamoto UniversityKumamotoJapan
| | - Ko Eto
- Department of Biological SciencesGraduate School of Science and TechnologyKumamoto UniversityKumamotoJapan
| |
Collapse
|
108
|
The Role of Activator Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers (Basel) 2018; 10:cancers10040093. [PMID: 29597249 PMCID: PMC5923348 DOI: 10.3390/cancers10040093] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/21/2018] [Accepted: 03/25/2018] [Indexed: 12/14/2022] Open
Abstract
The Activator Protein-1 (AP-1) transcription factor (TF) family, composed of a variety of members including c-JUN, c-FOS and ATF, is involved in mediating many biological processes such as proliferation, differentiation and cell death. Since their discovery, the role of AP-1 TFs in cancer development has been extensively analysed. Multiple in vitro and in vivo studies have highlighted the complexity of these TFs, mainly due to their cell-type specific homo- or hetero-dimerization resulting in diverse transcriptional response profiles. However, as a result of the increasing knowledge of the role of AP-1 TFs in disease, these TFs are being recognized as promising therapeutic targets for various malignancies. In this review, we focus on the impact of deregulated expression of AP-1 TFs in CD30-positive lymphomas including Classical Hodgkin Lymphoma and Anaplastic Large Cell Lymphoma.
Collapse
|
109
|
Neonatal Colonic Inflammation Epigenetically Aggravates Epithelial Inflammatory Responses to Injury in Adult Life. Cell Mol Gastroenterol Hepatol 2018; 6:65-78. [PMID: 29928672 PMCID: PMC6008258 DOI: 10.1016/j.jcmgh.2018.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/01/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Early life adversity is considered a risk factor for the development of gastrointestinal diseases, including inflammatory bowel disease. We hypothesized that early life colonic inflammation causes susceptibility to aggravated overexpression of interleukin (IL)1β. METHODS We developed a 2-hit rat model in which neonatal inflammation (NI) and adult inflammation (AI) were induced by trinitrobenzene sulfonic acid. RESULTS Aggravated immune responses were observed in NI + AI rats, including a sustained up-regulation of IL1β and other cytokines. In parallel with exacerbated loss of inhibitor of kappa B alpha expression, NI + AI rats showed hyperacetylation of histone H4K12 and increased V-Rel Avian Reticuloendotheliosis Viral Oncogene Homolog A binding on the IL1B promoter, accompanied by high levels of norepinephrine/epinephrine. Propranolol, a β-blocker, markedly ameliorated the inflammatory response and IL1β overexpression by mitigating against epigenetic modifications. Adrenalectomy abrogated NI-induced disease susceptibility whereas yohimbine sensitized the epithelium for exacerbated immune response. The macrophages of NI rats produced more IL1β than controls after exposure to lipopolysaccharide (LPS), suggesting hypersensitization; incubation with LPS plus Foradil (Sigma, St. Louis, MO), a β2-agonist, induced a greater IL1β expression than LPS alone. Epinephrine and Foradil also exacerbated LPS-induced IL1β activation in human THP-1-derived macrophages, by increasing acetylated H4K12, and these increases were abrogated by propranolol. CONCLUSIONS NI sensitizes the colon epithelium for exacerbated IL1β activation by increasing stress hormones that induce histone hyperacetylation, allowing greater access of nuclear factor-κB to the IL1B promoter and rendering the host susceptible to aggravated immune responses. Our findings suggest that β blockers have a therapeutic potential for inflammatory bowel disease susceptibility and establish a novel paradigm whereby NI induces epigenetic susceptibility to inflammatory bowel disease.
Collapse
Key Words
- AI, adult inflammation
- ChIP, chromatin immunoprecipitation
- Ctl, control
- Early Life Adversity
- Epinephrine
- H4K12ac, acetylated HRK12
- HDAC, histone deacetylase
- Histone Acetylation
- IBD, inflammatory bowel disease
- IL, interleukin
- Inflammatory Bowel Disease
- IκB, inhibitor of kappa B alpha
- LPS, lipopolysaccharide
- MPO, myeloperoxidase
- NF-κB
- NF-κB, nuclear factor-κB
- NI, neonatal inflammation
- PCR, polymerase chain reaction
- PMA, phorbol 12-myristate 13-acetate
- RNAP II, RNA polymerase II
- RelA, V-Rel Avian Reticuloendotheliosis Viral Oncogene Homolog A
- TNBS, 2,4,6-trinitrobenzene sulfonic acid
- Tnf, tumor necrosis factor
- mRNA, messenger RNA
Collapse
|
110
|
The potential applications of mushrooms against some facets of atherosclerosis: A review. Food Res Int 2018; 105:517-536. [DOI: 10.1016/j.foodres.2017.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/08/2017] [Accepted: 11/19/2017] [Indexed: 12/16/2022]
|
111
|
Maurer K, Ramen S, Shi L, Song L, Sullivan KE. Rapid induction of expression by LPS is accompanied by favorable chromatin and rapid binding of c-Jun. Mol Immunol 2018; 95:99-106. [PMID: 29433067 DOI: 10.1016/j.molimm.2018.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/20/2023]
Abstract
The response to infection is managed in mammals by a coordinated immune response. Innate responses are rapid and hard wired and have been demonstrated to be regulated at the level of chromatin accessibility. This study examined primary human monocyte responses to LPS as a model of innate responses to bacteria. We utilized inhibitors of chromatin modifying enzymes to understand the inter-relationships of the chromatin complexes regulating transcription. Multiplex digital gene detection was utilized to quantitate changes in mRNA levels for genes induced by LPS. In the first 30 min, genes that were highly induced by LPS as a group exhibited minimal effect of the chemical inhibitors of chromatin modifications. At 60 min, the more highly expressed genes were markedly more inhibitable. The effects of the inhibitors were almost entirely concordant in spite of different mechanisms of action. Two focus groups of genes with either high LPS inducibility at 30 min or high LPS inducibility at 60 min (but not at 30 min) were further examined by ChIP assay. NFκB p65 binding was increased at the promoters of 30- and 60-min highly inducible genes equivalently. Binding of c-Jun was increased after LPS in the 30-min inducible gene set but not the 60-min inducible gene set. H3K4me3 and H4ac were not detectably altered by LPS stimulation. Baseline H3K4me3 and H4ac were higher in the 30-min highly inducible gene set compared to the 60-min highly inducible gene set. NFκB and JNK inhibitors led to diminished H4ac after LPS. The effects of DRB and C646 were greater for LPS-induced IL6 transcription at 30 min and LPS-stimulated H4ac compared to TNF where transcription was largely unaffected by the inhibitors. In conclusion, genes with very rapidly induced expression after LPS exhibited more favorable chromatin characteristics at baseline and were less inhibitable than genes induced at the later time points.
Collapse
Affiliation(s)
- Kelly Maurer
- The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Swathi Ramen
- The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lihua Shi
- The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Li Song
- The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- The Children's Hospital of Philadelphia, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
112
|
Koronowicz AA, Drozdowska M, Wielgos B, Piasna-Słupecka E, Domagała D, Dulińska-Litewka J, Leszczyńska T. The effect of "NutramilTM Complex," food for special medical purpose, on breast and prostate carcinoma cells. PLoS One 2018; 13:e0192860. [PMID: 29444163 PMCID: PMC5812662 DOI: 10.1371/journal.pone.0192860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/31/2018] [Indexed: 11/18/2022] Open
Abstract
NutramilTM Complex is a multicomponent food product that meets the requirements of a food for special medical purpose. As a complete, high-energy diet it consists of properly balanced nutrients, vitamins and minerals. The aim of this study was to assess the effect of NutramilTM Complex on breast and prostate carcinoma cells. Our results showed that NutramilTM Complex reduced the viability and proliferation of breast and prostate cancer cells and that this process was associated with the induction of apoptosis via activation of caspase signalling. Data showed elevated levels of p53 tumour suppressor, up-regulation of p38 MAPK and SAPK / JNK proteins and downregulation of anti-apoptotic ERK1/2, AKT1 and HSP27. Treatment with NutramilTM Complex also affected the expression of the BCL2 family genes. Results also showed down-regulation of anti-apoptotic BCL-2 and up-regulation of pro-apoptotic members such as BAX, BAD, BID. In addition, we also observed regulation of many other genes, including Iκβα, Chk1 and Chk2, associated with apoptotic events. Taken together, our results suggest activation of the mitochondrial apoptotic pathway as most likely mechanism of anti-carcinogenic activity of NutramilTM Complex.
Collapse
Affiliation(s)
- Aneta A. Koronowicz
- Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland
| | - Mariola Drozdowska
- Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland
| | | | - Ewelina Piasna-Słupecka
- Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland
| | - Dominik Domagała
- Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland
| | | | - Teresa Leszczyńska
- Department of Human Nutrition, Faculty of Food Technology, University of Agriculture, Krakow, Poland
| |
Collapse
|
113
|
Yang T, Zhang F, Zhai L, He W, Tan Z, Sun Y, Wang Y, Liu L, Ning C, Zhou W, Ao H, Wang C, Yu Y. Transcriptome of Porcine PBMCs over Two Generations Reveals Key Genes and Pathways Associated with Variable Antibody Responses post PRRSV Vaccination. Sci Rep 2018; 8:2460. [PMID: 29410429 PMCID: PMC5802836 DOI: 10.1038/s41598-018-20701-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a virus susceptible to antibody dependent enhancement, causing reproductive failures in sows and preweaning mortality of piglets. Modified-live virus (MLV) vaccines are used to control PRRS in swine herds. However, immunized sows and piglets often generate variable antibody levels. This study aimed to detect significant genes and pathways involved in antibody responsiveness of pregnant sows and their offspring post-PRRSV vaccination. RNA sequencing was conducted on peripheral blood-mononuclear cells (PBMCs), which were isolated from pregnant sows and their piglets with high (HA), median (MA), and low (LA) PRRS antibody levels following vaccination. 401 differentially expressed genes (DEGs) were identified in three comparisons (HA versus MA, HA versus LA, and MA versus LA) of sow PBMCs. Two novel pathways (complement and coagulation cascade pathway; and epithelial cell signaling in H. pylori infection pathway) revealed by DEGs in HA versus LA and MA versus LA were involved in chemotactic and proinflammatory responses. TNF-α, CCL4, and NFKBIA genes displayed the same expression trends in subsequent generation post-PRRS-MLV vaccination. Findings of the study suggest that two pathways and TNF-α, CCL4, and NFKBIA could be considered as key pathways and potential candidate genes for PRRSV vaccine responsiveness, respectively.
Collapse
Affiliation(s)
- Ting Yang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Fengxia Zhang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Liwei Zhai
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Weiyong He
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zhen Tan
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Yangyang Sun
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuan Wang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Lei Liu
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Chao Ning
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China
| | - Weiliang Zhou
- Tianjin Ninghe Primary Pig Breeding Farm, Ninghe, 301500, Tianjin, China
| | - Hong Ao
- State Key Laboratory for Animal Nutrition, Key Laboratory for Domestic Animal Genetic Resources and Breeding of the Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chuduan Wang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China.
| | - Ying Yu
- Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture & National Engineering Laboratory for Animal Breeding, College of Animal Sciences and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
114
|
Cavaleri F. Presenting a New Standard Drug Model for Turmeric and Its Prized Extract, Curcumin. Int J Inflam 2018; 2018:5023429. [PMID: 29568482 PMCID: PMC5820622 DOI: 10.1155/2018/5023429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023] Open
Abstract
Various parts of the turmeric plant have been used as medicinal treatment for various conditions from ulcers and arthritis to cardiovascular disease and neuroinflammation. The rhizome's curcumin extract is the most studied active constituent, which exhibits an expansive polypharmacology with influence on many key inflammatory markers. Despite the expansive reports of curcucmin's therapeutic value, clinical reliability and research repeatability with curcumin treatment are still poor. The pharmacology must be better understood and reliably mapped if curcumin is to be accepted and used in modern medical applications. Although the polypharmacology of this extract has been considered, in mainstream medicine, to be a drawback, a perspective change reveals a comprehensive and even synergistic shaping of the NF-kB pathway, including transactivation. Much of the inconsistent research data and unreliable clinical outcomes may be due to a lack of standardization which also pervades research standard samples. The possibility of other well-known curcumin by-products contributing in the polypharmacology is also discussed. A new flowchart of crosstalk in transduction pathways that lead to shaping of nuclear NF-kB transactivation is generated and a new calibration or standardization protocol for the extract is proposed which could lead to more consistent data extraction and improved reliability in therapy.
Collapse
Affiliation(s)
- Franco Cavaleri
- Biologic Pharmamedical Research, 688-2397 King George Blvd., White Rock, BC, Canada V4A7E9
| |
Collapse
|
115
|
Abstract
PURPOSE OF REVIEW To summarize recent clinical and preclinical studies on extracranial pathophysiologies in migraine. It challenges the opinion-based notion that the headache phase of migraine occurs without input from peripheral nociceptors or is caused solely by activation of intracranial nociceptors supplying dural and cerebral vasculature. RECENT FINDINGS Data that support a scenario by which migraine can originate extracranially include the perception of imploding headache that hurts outside the cranium, the existence of a network of sensory fibers that bifurcate from parent axons of intracranial meningeal nociceptors and reach extracranial tissues such as periosteum and pericranial muscles by crossing the calvarial bones through the sutures, the discovery of proinflammatory genes that are upregulated and anti-inflammatory genes that are down regulated in extracranial tissue of chronic migraine patients, and evidence that administration of OnabotulinumtoxinA to peripheral tissues outside the calvaria reduces frequency of migraine headache. SUMMARY These findings seeks to shift clinical practice from prophylactically treating chronic migraine solely with medications that reduce neuronal excitability to treating irritated nociceptors or affected tissues. The findings also seeks to shift current research from focusing solely on central nervous system alterations and activation of meningeal nociceptors as a prerequisite for studying migraine.
Collapse
|
116
|
Publisher’s note. Colloids Surf B Biointerfaces 2017; 160:423-428. [DOI: 10.1016/j.colsurfb.2017.09.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 09/20/2017] [Accepted: 09/24/2017] [Indexed: 01/22/2023]
|
117
|
Ahn S, Singh P, Jang M, Kim YJ, Castro-Aceituno V, Simu SY, Kim YJ, Yang DC. Gold nanoflowers synthesized using Acanthopanacis cortex extract inhibit inflammatory mediators in LPS-induced RAW264.7 macrophages via NF-κB and AP-1 pathways. Colloids Surf B Biointerfaces 2017; 162:398-404. [PMID: 29245117 DOI: 10.1016/j.colsurfb.2017.11.037] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We reported the rapid synthesis (<8s) of gold nanoparticles at room temperature using Acanthopanacis cortex extract (A-AuNPs). We characterized the A-AuNPs using several analytical techniques and found that nano-flower type A-AuNPs, which are known to possess a coarse surface with a high surface to volume ratio, conferring these particles with high binding capacity for various biological molecules. After confirming the stability of the nanoparticles, we investigated the anti-inflammatory effect of A-AuNPs in LPS-stimulated RAW264.7 cells. These nanoparticles inhibited LPS-induced iNOS and COX-2 protein as well as gene expression level, along with reduction of NO and PGE2 production. Furthermore, we observed that the A-AuNPs inhibited translocation of NF-κB and AP-1 through phosphorylation of MAPK signaling by western blot analysis. In summary, we synthesized gold nanoflowers in an economical and eco-friendly way using Acanthopanacis cortex extract and the resultant flower-like A-AuNPs had anti-inflammatory activity, highlighting their potential as therapeutic candidates for suppression of inflammatory-mediated diseases.
Collapse
Affiliation(s)
- Sungeun Ahn
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea; Graduate School of Biotechnology and Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Priyanka Singh
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea; Graduate School of Biotechnology and Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Mi Jang
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Yu-Jin Kim
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Verónica Castro-Aceituno
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Shakina Yesmin Simu
- Graduate School of Biotechnology and Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Yeon Ju Kim
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea.
| | - Deok-Chun Yang
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea; Graduate School of Biotechnology and Ginseng Bank, College of Life Sciences, Kyung Hee University, Yongin, 17104, Republic of Korea.
| |
Collapse
|
118
|
Li L, Hou Y, Yu J, Lu Y, Chang L, Jiang M, Wu X. Synergism of ursolic acid and cisplatin promotes apoptosis and enhances growth inhibition of cervical cancer cells via suppressing NF-κB p65. Oncotarget 2017; 8:97416-97427. [PMID: 29228621 PMCID: PMC5722573 DOI: 10.18632/oncotarget.22133] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/17/2017] [Indexed: 01/03/2023] Open
Abstract
Objective This study was designed to investigate the effect of combination of ursolic acid (UA) with cisplatin (DDP) on cervical cancer cell proliferation and apoptosis. Methods The mRNA and protein expressions of nuclear factor-kappa B (NF-κB) p65 in cervical cancer cells were examined using RT-PCR and western blot. MTT and colony formation assays were performed to examine the DDP toxicity and the proliferation ability of cervical cancer cells. Cell morphology was observed by means of Hoechst33258 and transmission electron microscopy (TEM). The apoptosis rate and cell cycle were assessed through flow cytometry assay. Western blot was used to detect the expression of apoptosis-related molecules. Results The mRNA and protein expressions of NF-κB p65 in cervical cancer cells were significantly higher than that in cervical epithelial cells. The combined treatment of UA and DDP inhibited cervical cancer cell growth and promoted apoptosis more effectively than DDP treatment or UA treatment alone (P < 0.05). Compared with the DDP group and UA group, the expressions of Bcl-2 and NF-κB p65 in DDP +UA group were decreased, while the expressions of Bax, Caspase-3 and PARP cleavage were observably increased. The expression of nuclear NF-κB p65 significantly reduced in UA group and DDP +UA group. si-p65 group displayed a decrease of cell proliferation ability and led to a significant reduction in the number of SiHa cell colony formation. Conclusion The combination of UA with DDP could more effectively inhibit SiHa cells proliferation and facilitate cell apoptosis through suppressing NF-κB p65.
Collapse
Affiliation(s)
- Lan Li
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| | - Yu Hou
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| | - Jing Yu
- Department of Gynaecology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| | - Yulin Lu
- Nursing School, Kunming Medical University, Kunming 650118, China
| | - Li Chang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| | - Meiping Jiang
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| | - Xingrao Wu
- Department of Radiation Oncology, The Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming 650118, China
| |
Collapse
|
119
|
Poma P, Labbozzetta M, D'Alessandro N, Notarbartolo M. NF-κB Is a Potential Molecular Drug Target in Triple-Negative Breast Cancers. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2017; 21:225-231. [PMID: 28388298 DOI: 10.1089/omi.2017.0020] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer continues to cause significant burden in global health morbidity and mortality. Triple-negative breast cancers (TNBCs) are highly aggressive with poor prognosis and are characterized by lack of expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor (Her-2). TNBCs are often resistant to cytotoxic chemotherapy and pose major difficulty in achieving personalized medicine due to their molecular heterogeneity. There is increasing evidence that the aberrant activation of nuclear factor (NF)-κB signaling is a frequent characteristic of TNBCs. We evaluated the effects of different potential NF-κB inhibitors, such as bisindolylmaleimide I (BIS, a selective protein kinase C [PKC] inhibitor), MG132 (a proteasome inhibitor), curcumin (endowed with pleiotropic activities), and dehydroxymethylepoxyquinomicin (an inhibitor of NF-κB translocation into the nucleus) on the constitutive activation of NF-κB present in three TNBC cell lines (SUM 149, SUM 159, and MDA-MB-231). We also evaluated whether MDA-9/Syntenin plays a role in NF-κB activation, as observed in other cancer types. Indeed, silencing experiments with a siRNA anti-MDA-9/Syntenin produced a very strong reduction of NF-κB activation in all the three TNBC cell lines. We conclude that different approaches targeting NF-κB activation might potentially prove useful for innovation in anticancer drug development for TNBCs. Further research that bridge preclinical and clinical investigations with NF-κB inhibitors would be timely and warranted.
Collapse
Affiliation(s)
- Paola Poma
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care "G. D'Alessandro," School of Medicine, University of Palermo , Palermo, Italy
| | - Manuela Labbozzetta
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care "G. D'Alessandro," School of Medicine, University of Palermo , Palermo, Italy
| | - Natale D'Alessandro
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care "G. D'Alessandro," School of Medicine, University of Palermo , Palermo, Italy
| | - Monica Notarbartolo
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care "G. D'Alessandro," School of Medicine, University of Palermo , Palermo, Italy
| |
Collapse
|
120
|
Chen S, Maini R, Bai X, Nangreave RC, Dedkova LM, Hecht SM. Incorporation of Phosphorylated Tyrosine into Proteins: In Vitro Translation and Study of Phosphorylated IκB-α and Its Interaction with NF-κB. J Am Chem Soc 2017; 139:14098-14108. [PMID: 28898075 PMCID: PMC5901656 DOI: 10.1021/jacs.7b05168] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Phosphorylated proteins play important roles in the regulation of many different cell networks. However, unlike the preparation of proteins containing unmodified proteinogenic amino acids, which can be altered readily by site-directed mutagenesis and expressed in vitro and in vivo, the preparation of proteins phosphorylated at predetermined sites cannot be done easily and in acceptable yields. To enable the synthesis of phosphorylated proteins for in vitro studies, we have explored the use of phosphorylated amino acids in which the phosphate moiety bears a chemical protecting group, thus eliminating the negative charges that have been shown to have a negative effect on protein translation. Bis-o-nitrobenzyl protection of tyrosine phosphate enabled its incorporation into DHFR and IκB-α using wild-type ribosomes, and the elaborated proteins could subsequently be deprotected by photolysis. Also investigated in parallel was the re-engineering of the 23S rRNA of Escherichia coli, guided by the use of a phosphorylated puromycin, to identify modified ribosomes capable of incorporating unprotected phosphotyrosine into proteins from a phosphotyrosyl-tRNACUA by UAG codon suppression during in vitro translation. Selection of a library of modified ribosomal clones with phosphorylated puromycin identified six modified ribosome variants having mutations in nucleotides 2600-2605 of 23S rRNA; these had enhanced sensitivity to the phosphorylated puromycin. The six clones demonstrated some sequence homology in the region 2600-2605 and incorporated unprotected phosphotyrosine into IκB-α using a modified gene having a TAG codon in the position corresponding to amino acid 42 of the protein. The purified phosphorylated protein bound to a phosphotyrosine specific antibody and permitted NF-κB binding to a DNA duplex sequence corresponding to its binding site in the IL-2 gene promoter. Unexpectedly, phosphorylated IκB-α also mediated the exchange of exogenous DNA into an NF-κB-cellular DNA complex isolated from the nucleus of activated Jurkat cells.
Collapse
Affiliation(s)
- Shengxi Chen
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Rumit Maini
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Xiaoguang Bai
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Ryan C. Nangreave
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Larisa M. Dedkova
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Sidney M. Hecht
- Biodesign Center for BioEnergetics, and School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
121
|
Qu M, Yu J, Liu H, Ren Y, Ma C, Bu X, Lan Q. The Candidate Tumor Suppressor Gene SLC8A2 Inhibits Invasion, Angiogenesis and Growth of Glioblastoma. Mol Cells 2017; 40:761-772. [PMID: 29047259 PMCID: PMC5682253 DOI: 10.14348/molcells.2017.0104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/14/2017] [Accepted: 08/20/2017] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma is the most frequent and most aggressive brain tumor in adults. Solute carrier family 8 member 2 (SLC8A2) is only expressed in normal brain, but not present in other human normal tissues or in gliomas. Therefore, we hypothesized that SLC8A2 might be a glioma tumor suppressor gene and detected the role of SLC8A2 in glioblastoma and explored the underlying molecular mechanism. The glioblastoma U87MG cells stably transfected with the lentivirus plasmid containg SLC8A2 (U87MG-SLC8A2) and negative control (U87MG-NC) were constructed. In the present study, we found that the tumorigenicity of U87MG in nude mice was totally inhibited by SLC8A2. Overexpression of SLC8A2 had no effect on cell proliferation or cell cycle, but impaired the invasion and migration of U87MG cells, most likely through inactivating the extracellular signal-related kinases (ERK)1/2 signaling pathway, inhibiting the nuclear translocation and DNA binding activity of nuclear factor kappa B (NF-κB), reducing the level of matrix metalloproteinases (MMPs) and urokinase-type plasminogen activator (uPA)-its receptor (uPAR) system (ERK1/2-NF-κB-MMPs/uPA-uPAR), and altering the protein levels of epithelial to mesenchymal transitions (EMT)-associated proteins E-cardherin, vimentin and Snail. In addition, SLC8A2 inhibited the angiogenesis of U87MG cells, probably through combined inhibition of endothelium-dependent and endothelium-nondependent angiogenesis (vascular mimicry pattern). Totally, SLC8A2 serves as a tumor suppressor gene and inhibits invasion, angiogenesis and growth of glioblastoma.
Collapse
Affiliation(s)
- Mingqi Qu
- Department of Neurosurgery, Henan Provincial People’s Hospital,
P.R. China
- Department of Neurosurgery, People’s Hospital of Zhengzhou University,
P.R. China
| | - Ju Yu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University,
P.R. China
| | - Hongyuan Liu
- Department of Neurosurgery, Mianyang Central Hospital,
P.R. China
| | - Ying Ren
- Department of Pathology, People’s Hospital of Zhengzhou University,
P.R. China
| | - Chunxiao Ma
- Department of Neurosurgery, Henan Provincial People’s Hospital,
P.R. China
- Department of Neurosurgery, People’s Hospital of Zhengzhou University,
P.R. China
| | - Xingyao Bu
- Department of Neurosurgery, Henan Provincial People’s Hospital,
P.R. China
- Department of Neurosurgery, People’s Hospital of Zhengzhou University,
P.R. China
| | - Qing Lan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University,
P.R. China
| |
Collapse
|
122
|
Rice KM, Manne ND, Arvapalli R, Ginjupalli GK, Blough ER. Diabetes alters vascular mechanotransduction data: Pressure-induced regulation of Nf-kapa-B p65 and translational associated signaling in the rat inferior vena cava. Data Brief 2017; 14:676-685. [PMID: 28932772 PMCID: PMC5596213 DOI: 10.1016/j.dib.2017.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 01/21/2023] Open
Abstract
Diabetic patients have a high rate of vein graft failure due to attrition or vessel occlusion that cause recurrent ischemic events or vein graft. Veins grafted into a high-pressure arterial environment must undergo vascular remodeling to better handle the altered hemodynamics and intravascular increased pressure. Multiple cellular and molecular events are purported to be associated with vascular remodeling of veins. Understanding the effect diabetes has on vascular mechano-transductive response is critical to decreasing graft failure rates. This article represents data regarding a study published in Cardiovascular Diabetology [1] and Open Journal of Endocrine and Metabolic Diseases [2] with the purpose of evaluating the effect of pressurization on rat inferior venae cavae (IVC). Here we provide the information about the method and processing of raw data related to our prior publish work and Data in Brief articles [3], [4]. The data contained in this article evaluates the contribution of NF-kB signaling and associated proteins. IVC from lean and obese animals were exposed to a 30 min of perfusion at 120 mm Hg pressure and evaluated for changes in expression and (IkB-alpha, NF-kB p50, NF-kB p105, NF-kB p65, Traf2, caspase 12), phosphorylation of (IkB-alpha (ser 32), Fox01 (ser 256), and Fox04 (ser 193)) proteins thought to be involved in the regulation of vascular mechanotransduction.
Collapse
Affiliation(s)
- Kevin M. Rice
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA
- Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
- Biotechnology Graduate Program West Virginia State University, Institute, WV, USA
- Department of Health and Human Service, School of Kinesiology, Marshall University, Huntington, WV, USA
| | | | | | | | - Eric R. Blough
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA
- Biotechnology Graduate Program West Virginia State University, Institute, WV, USA
- Department of Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
123
|
Tran QT, Wong WF, Chai CL. Labdane diterpenoids as potential anti-inflammatory agents. Pharmacol Res 2017; 124:43-63. [PMID: 28751221 DOI: 10.1016/j.phrs.2017.07.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 01/20/2023]
|
124
|
Lohman BK, Steinel NC, Weber JN, Bolnick DI. Gene Expression Contributes to the Recent Evolution of Host Resistance in a Model Host Parasite System. Front Immunol 2017; 8:1071. [PMID: 28955327 PMCID: PMC5600903 DOI: 10.3389/fimmu.2017.01071] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/16/2017] [Indexed: 12/31/2022] Open
Abstract
Heritable population differences in immune gene expression following infection can reveal mechanisms of host immune evolution. We compared gene expression in infected and uninfected threespine stickleback (Gasterosteus aculeatus) from two natural populations that differ in resistance to a native cestode parasite, Schistocephalus solidus. Genes in both the innate and adaptive immune system were differentially expressed as a function of host population, infection status, and their interaction. These genes were enriched for loci controlling immune functions known to differ between host populations or in response to infection. Coexpression network analysis identified two distinct processes contributing to resistance: parasite survival and suppression of growth. Comparing networks between populations showed resistant fish have a dynamic expression profile while susceptible fish are static. In summary, recent evolutionary divergence between two vertebrate populations has generated population-specific gene expression responses to parasite infection, affecting parasite establishment and growth.
Collapse
Affiliation(s)
- Brian K Lohman
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, United States
| | - Natalie C Steinel
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, United States.,Department of Medical Education, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Jesse N Weber
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, United States.,Division of Biological Sciences, The University of Montana, Missoula, MT, United States
| | - Daniel I Bolnick
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
125
|
Cheng BF, Gao YX, Lian JJ, Guo DD, Liu TT, Xie YF, Wang L, Yang HJ, Wang M, Feng ZW. Anti-inflammatory effects of pitavastatin in interleukin-1β-induced SW982 human synovial cells. Int Immunopharmacol 2017; 50:224-229. [DOI: 10.1016/j.intimp.2017.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/09/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023]
|
126
|
Zhang HJ, Wang XZ, Cao Q, Gong GH, Quan ZS. Design, synthesis, anti-inflammatory activity, and molecular docking studies of perimidine derivatives containing triazole. Bioorg Med Chem Lett 2017; 27:4409-4414. [DOI: 10.1016/j.bmcl.2017.08.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/06/2017] [Accepted: 08/08/2017] [Indexed: 02/01/2023]
|
127
|
Fan C, Wu LH, Zhang GF, Xu F, Zhang S, Zhang X, Sun L, Yu Y, Zhang Y, Ye RD. 4'-Hydroxywogonin suppresses lipopolysaccharide-induced inflammatory responses in RAW 264.7 macrophages and acute lung injury mice. PLoS One 2017; 12:e0181191. [PMID: 28792498 PMCID: PMC5549707 DOI: 10.1371/journal.pone.0181191] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/26/2017] [Indexed: 12/22/2022] Open
Abstract
4'-Hydroxywogonin (4'-HW), a flavonoid, has been isolated from various plants and shown to inhibit NO production in macrophages. However, the molecular mechanisms and its in vivo activity have not been determined. Our study aimed to investigate the mechanisms underlying the anti-inflammatory effects of 4'-HW in vitro and in vivo. We showed that 4'-HW potently reduced the expression levels of COX-2 and iNOS as well as their products, prostaglandin E2 (PGE2) and nitric oxide (NO) respectively, in LPS-stimulated RAW 264.7 macrophages. 4'-HW also suppressed LPS-induced pro-inflammatory cytokines at mRNA and protein levels. Moreover, 4'-HW blocked the interaction of TAK1 and TAB1 in LPS-stimulated RAW 264.7 macrophages, resulting in an inhibition of the TAK1/IKK/NF-κB signaling pathway. Furthermore, 4'-HW also reduced the phosphorylation of MAPKs and PI3/Akt signaling pathways in LPS-stimulated RAW 264.7 macrophages. 4'-HW was also significantly decreased the intracellular reactive oxygen species (ROS) level. The effect of 4'-HW was confirmed in vivo. 4'-HW exhibited potent protective effect against LPS-induced ALI in mice. These findings indicate that 4'-HW suppresses the LPS-induced response in vitro and in vivo. It is likely that the inhibition of the TAK1/IKK/NF-κB, MAPKs and PI3/AKT signaling pathways contribute to the anti-inflammatory effects of 4'-HW. Our study suggests that 4'-HW may be an important functional constituent in the plants and has the potential value to be developed as a novel anti-inflammatory agent.
Collapse
Affiliation(s)
- Chao Fan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Le-Hao Wu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Gu-Fang Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfang Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Shuo Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuli Zhang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Lei Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Yu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (YZ); (RDY)
| | - Richard D. Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Institute of Chinese Medical Sciences, University of Macau, Macau Special Administrative Region, China
- * E-mail: (YZ); (RDY)
| |
Collapse
|
128
|
Cho YC, Kim BR, Le HTT, Cho S. Anti‑inflammatory effects on murine macrophages of ethanol extracts of Lygodium japonicum spores via inhibition of NF‑κB and p38. Mol Med Rep 2017; 16:4362-4370. [PMID: 29067444 DOI: 10.3892/mmr.2017.7070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/28/2017] [Indexed: 11/06/2022] Open
Abstract
The spores of Lygodium japonicum (Thunb.) Sw. (L. japonicum) have been used in traditional Chinese medicine for the treatment of various inflammatory diseases. However, the molecular mechanisms underlying their anti‑inflammatory effects have yet to be elucidated. In the present study, we investigated the anti‑inflammatory effects of ethanol extracts of L. japonicum spores (ELJ) by measuring the production of inflammatory mediators, and explored the molecular mechanisms underlying the effects of ELJ in murine macrophages in vitro using immunoblotting analyses. At non‑cytotoxic concentrations of (50‑300 µg/ml), ELJ was revealed to significantly suppress the production of nitric oxide (NO) and tumor necrosis factor (TNF)‑α in lipopolysaccharide (LPS)‑stimulated murine RAW 264.7 macrophages; ELJ repressed the production of interleukin (IL)‑6 only at high concentrations (≥200 µg/ml). The ELJ‑mediated decrease in NO production was demonstrated to depend on the downregulation of inducible NO synthase mRNA and protein expression. Conversely, the mRNA and protein expression of cyclooxygenase‑2 were not affected by ELJ. In addition, ELJ was revealed to inhibit the mRNA expression of IL‑6, IL‑1β, and TNF‑α in LPS‑stimulated RAW 264.7 macrophages. The effects of ELJ on proinflammatory mediators may have been due to the stabilization of inhibitor of κBα and the inhibition of p38 mitogen‑activated protein kinase (MAPK). These results suggested that ELJ may suppress LPS‑induced inflammatory responses in murine macrophages in vitro, through the negative regulation of p38 MAPK and nuclear factor (NF)‑κB. Therefore, ELJ may have potential as a novel candidate for the development of therapeutic strategies aimed at alleviating inflammation.
Collapse
Affiliation(s)
- Young-Chang Cho
- Laboratory of Molecular Pharmacological Cell Biology, College of Pharmacy, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Ba Reum Kim
- Laboratory of Molecular Pharmacological Cell Biology, College of Pharmacy, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Hien Thi Thu Le
- Laboratory of Molecular Pharmacological Cell Biology, College of Pharmacy, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Sayeon Cho
- Laboratory of Molecular Pharmacological Cell Biology, College of Pharmacy, Chung‑Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
129
|
Wang Y, Weng H, Song JF, Deng YH, Li S, Liu HB. Activation of the HMGB1‑TLR4‑NF‑κB pathway may occur in patients with atopic eczema. Mol Med Rep 2017; 16:2714-2720. [PMID: 28713916 PMCID: PMC5547948 DOI: 10.3892/mmr.2017.6942] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 05/12/2017] [Indexed: 01/01/2023] Open
Abstract
High mobility group protein B1 (HMGB1) has been reported to serve important roles in various pathological conditions. Toll‑like receptor 4 (TLR4), as one of the HMGB1 receptors, has been reported to be involved in the development of certain inflammatory diseases by activating nuclear factor NF‑κ‑B (NF‑κB). However, there are few studies investigating the effects of HMGB1, TLR4 and NF‑κB on human inflammatory dermatoses. In the present study, the distribution and characteristics of HMGB1, TLR4 and NF‑κB p65 expression in psoriasis and atopic eczema (AE) were investigated. In addition, immunohistochemical analysis was performed to evaluate their expression and distribution in normal skin, and in patients with AE or psoria-sis. Spearman's correlation analysis was used to predicate their relevancy. The present study identified that the p65 level in epithelial nuclei in AE skin was increased compared with normal and psoriasis skin (P<0.01). The level of extracellular HMGB1 in AE skin was also increased compared with normal and psoriasis skin (P<0.01). Meanwhile, TLR4 expression on the epithelial membranes of AE skin was increased compared with psoriasis skin (P<0.01). Furthermore, the level of extracellular HMGB1 was positively correlated with epithelial membrane TLR4 (r=0.3856; P<0.05) and epithelial nuclear p65 (r=0.5894; P<0.01) in AE skin. These results indicated that the HMGB1‑TLR4‑NF‑κB signaling pathway is activated in AE and may account for its pathogenesis, but not in psoriasis. Therefore, HMGB1, TLR4 and NF‑κB p65 have the potential to be targets for the treatment of human inflammatory dermatoses, including AE.
Collapse
Affiliation(s)
- Yong Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| | - Hui Weng
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jian Fei Song
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| | - Yun Hua Deng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shuang Li
- Department of Clinical Laboratory, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi 541002, P.R. China
| | - Hong Bo Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P.R. China
| |
Collapse
|
130
|
Li M, Guan H. Noncoding RNAs Regulating NF-κB Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 927:317-36. [PMID: 27376741 DOI: 10.1007/978-981-10-1498-7_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As transcription factors that regulate expression of a variety of genes essential for diverse physiological and pathological processes, nuclear factor kappa B (NF-κB) family molecules play important roles in the development and progression of malignant tumor, and constitutive activation of NF-κB has been evidenced in various types of tumor tissues. Underlying its pathologic role, deregulated expression and/or transactivating activity of NF-κB usually involves multiple layers of molecular mechanisms. Noncoding RNAs, including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are known to modulate expression and biological functions of regulatory proteins in a variety of cancer contexts. In this chapter, the regulatory role of miRNAs and lncRNAs in NF-κB signaling in malignant diseases will be discussed.
Collapse
Affiliation(s)
- Mengfeng Li
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, China.
| | - Hongyu Guan
- Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan Road II, Guangzhou, China
| |
Collapse
|
131
|
Liu DT, Brewer MS, Chen S, Hong W, Zhu Y. Transcriptomic signatures for ovulation in vertebrates. Gen Comp Endocrinol 2017; 247:74-86. [PMID: 28111234 PMCID: PMC5410184 DOI: 10.1016/j.ygcen.2017.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 01/01/2023]
Abstract
The central roles of luteinizing hormone (LH), progestin and their receptors for initiating ovulation have been well established. However, signaling pathways and downstream targets such as proteases that are essential for the rupture of follicular cells are still unclear. Recently, we found anovulation in nuclear progestin receptor (Pgr) knockout (Pgr-KO) zebrafish, which offers a new model for examining genes and pathways that are important for ovulation and fertility. In this study, we examined expression of all transcripts using RNA-Seq in preovulatory follicular cells collected following the final oocyte maturation, but prior to ovulation, from wild-type (WT) or Pgr-KO fish. Differential expression analysis revealed 3567 genes significantly differentially expressed between WT and Pgr-KO fish (fold change⩾2, p<0.05). Among those, 1543 gene transcripts were significantly more expressed, while 2024 genes were significantly less expressed, in WT than those in Pgr-KO. We then retrieved and compared transcriptional data from online databases and further identified 661 conserved genes in fish, mice, and humans that showed similar levels of high (283 genes) or low (387) expression in animals that were ovulating compared to those with no ovulation. For the first time, ovulatory genes and their involved biological processes and pathways were also visualized using Enrichment Map and Cytoscape. Intriguingly, enrichment analysis indicated that the genes with higher expression were involved in multiple ovulatory pathways and processes such as inflammatory response, angiogenesis, cytokine production, cell migration, chemotaxis, MAPK, focal adhesion, and cytoskeleton reorganization. In contrast, the genes with lower expression were mainly involved in DNA replication, DNA repair, DNA methylation, RNA processing, telomere maintenance, spindle assembling, nuclear acid transport, catabolic processes, and nuclear and cell division. Our results indicate that a large set of genes (>3000) is differentially regulated in the follicular cells in zebrafish prior to ovulation, terminating programs such as growth and proliferation, and beginning processes including the inflammatory response and apoptosis. Further studies are required to establish relationships among these genes and an ovulatory circuit in the zebrafish model.
Collapse
Affiliation(s)
- Dong Teng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Michael S Brewer
- Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Shixi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Wanshu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States.
| |
Collapse
|
132
|
Transcriptomics, NF-κB Pathway, and Their Potential Spaceflight-Related Health Consequences. Int J Mol Sci 2017; 18:ijms18061166. [PMID: 28561779 PMCID: PMC5485990 DOI: 10.3390/ijms18061166] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 01/05/2023] Open
Abstract
In space, living organisms are exposed to multiple stress factors including microgravity and space radiation. For humans, these harmful environmental factors have been known to cause negative health impacts such as bone loss and immune dysfunction. Understanding the mechanisms by which spaceflight impacts human health at the molecular level is critical not only for accurately assessing the risks associated with spaceflight, but also for developing effective countermeasures. Over the years, a number of studies have been conducted under real or simulated space conditions. RNA and protein levels in cellular and animal models have been targeted in order to identify pathways affected by spaceflight. Of the many pathways responsive to the space environment, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) network appears to commonly be affected across many different cell types under the true or simulated spaceflight conditions. NF-κB is of particular interest, as it is associated with many of the spaceflight-related health consequences. This review intends to summarize the transcriptomics studies that identified NF-κB as a responsive pathway to ground-based simulated microgravity or the true spaceflight condition. These studies were carried out using either human cell or animal models. In addition, the review summarizes the studies that focused specifically on NF-κB pathway in specific cell types or organ tissues as related to the known spaceflight-related health risks including immune dysfunction, bone loss, muscle atrophy, central nerve system (CNS) dysfunction, and risks associated with space radiation. Whether the NF-κB pathway is activated or inhibited in space is dependent on the cell type, but the potential health impact appeared to be always negative. It is argued that more studies on NF-κB should be conducted to fully understand this particular pathway for the benefit of crew health in space.
Collapse
|
133
|
Khalafalla FG, Khan MW. Inflammation and Epithelial-Mesenchymal Transition in Pancreatic Ductal Adenocarcinoma: Fighting Against Multiple Opponents. CANCER GROWTH AND METASTASIS 2017; 10:1179064417709287. [PMID: 28579826 PMCID: PMC5436837 DOI: 10.1177/1179064417709287] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer and one of the most lethal human cancers. Inflammation is a critical component in PDAC initiation and progression. Inflammation also contributes to the aggressiveness of PDAC indirectly via induction of epithelial-mesenchymal transition (EMT), altogether leading to enhanced resistance to chemotherapy and poor survival rates. This review gives an overview of the key pro-inflammatory signaling pathways involved in PDAC pathogenesis and discusses the role of inflammation in induction of EMT and development of chemoresistance in patients with PDAC.
Collapse
|
134
|
Periploca forrestii saponin ameliorates CIA via suppressing proinflammatory cytokines and nuclear factor kappa-B pathways. PLoS One 2017; 12:e0176672. [PMID: 28463993 PMCID: PMC5412996 DOI: 10.1371/journal.pone.0176672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/16/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Periploca forrestii Schltr has been used as a Chinese folk medicine for the treatment of rheumatism, arthralgia and fractures. However, the anti-arthritic activity of Periploca forrestii saponin (PFS) and the active compound has still not been revealed. This study aimed to investigate the protective effects and mechanisms of PFS on collagen type II (CII) collagen-induced arthritis (CIA) mice. We sought to investigate whether PFS and Periplocin could regulate osteoclastogenesis, and if so, further investigation on its mechanism of action. METHODS Arthritis was induced in female BALB/c mice by CIA method. PFS was administered at a dose of 50 mg/kg body weight once daily for five weeks. The effects of treatment in mice were assessed by histological and biochemical evaluation in sera and paws. Anti-osteoclastogenic action of PFS and Periplocin was identified using an osteoclast formation model induced by RANKL. RESULTS PFS ameliorated paw erythema and swelling, inhibited bone erosion in ankle joint histopathological examination. PFS treatment resulted in decreased IgG2a, and increased IgG1 levels in the serum of CIA mice. Decreased TNF-α, and increased interleukin (IL)-4 and IL-22 levels were also found in PFS-treated mice. PFS inhibited the I-κBα phosphorylation, blocked nuclear factor (NF)-κB/p65 phosphorylation and abrogated AP-1/c-Fos activity. PFS downregulated toll-like receptor (TLR) 4, STAT3 and MMP-9 expression in CIA mice and RANKL-induced osteoclastogenesis. PFS and Periplocin inhibited RANKL-induced osteoclast formation in a dose dependent manner within nongrowth inhibitory concentration, and PFS decreased osteoclastogenesis-related marker expression, including cathepsin K and MMP-9. CONCLUSION This study revealed that the protective mechanism of PFS on CIA was associated with regulatory effects on proinflammatory factors and further on the crosstalk between NF-κB and c-Fos/AP-1 in vivo and in vitro. Therefore, PFS is a promising therapeutic alternative for the treatment of RA, evidencing the need to conduct further studies that can identify their active components in treating and preventing RA.
Collapse
|
135
|
Huang X, Zang Y, Zhang M, Yuan X, Li M, Gao X. Nuclear Factor of κB1 Is a Key Regulator for the Transcriptional Activation of Milk Synthesis in Bovine Mammary Epithelial Cells. DNA Cell Biol 2017; 36:295-302. [DOI: 10.1089/dna.2016.3610] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Xin Huang
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| | - Yanli Zang
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| | - Minghui Zhang
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| | - Xiaohan Yuan
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| | - Meng Li
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| | - Xuejun Gao
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, China
| |
Collapse
|
136
|
Hidasi AO, Groh KJ, Suter MJF, Schirmer K. Clobetasol propionate causes immunosuppression in zebrafish (Danio rerio) at environmentally relevant concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2017; 138:16-24. [PMID: 27987419 DOI: 10.1016/j.ecoenv.2016.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/26/2016] [Accepted: 11/28/2016] [Indexed: 06/06/2023]
Abstract
Synthetic glucocorticoids (GCs) are potential endocrine disrupting compounds that have been detected in the aquatic environment around the world in the low ng/L (nanomolar) range. GCs are used as immunosuppressants in medicine. It is of high interest whether clobetasol propionate (CP), a highly potent GC, suppresses the inflammatory response in fish after exposure to environmentally relevant concentrations. Bacterial lipopolysaccharide (LPS) challenge was used to induce inflammation and thus mimic pathogen infection. Zebrafish embryos were exposed to ≤1000nM CP from ~1h post fertilization (hpf) to 96 hpf, and CP uptake, survival after LPS challenge, and expression of inflammation-related genes were examined. Our initial experiments were carried out using 0.001% DMSO as a solvent vehicle, but we observed that DMSO interfered with the LPS challenge assay, and thus masked the effects of CP. Therefore, DMSO was not used in the subsequent experiments. The internal CP concentration was quantifiable after exposure to ≥10nM CP for 96h. The bioconcentration factor (BCF) of CP was determined to be between 16 and 33 in zebrafish embryos. CP-exposed embryos showed a significantly higher survival rate in the LPS challenge assay after exposure to ≥0.1nM in a dose dependent manner. This effect is an indication of immunosuppression. Furthermore, the regulation pattern of several genes related to LPS challenge in mammals supported our results, providing evidence that LPS-mediated inflammatory pathways are conserved from mammals to teleost fish. Anxa1b, a GC-action related anti-inflammatory gene, was significantly down-regulated after exposure to ≥0.05nM CP. Our results show for the first time that synthetic GCs can suppress the innate immune system of fish at environmentally relevant concentrations. This may reduce the chances of fish to survive in the environment, as their defense against pathogens is weakened.
Collapse
Affiliation(s)
- Anita O Hidasi
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Department of Environmental Toxicology, Dübendorf 8600, Switzerland; EPFL, School of Architecture, Civil and Environmental Engineering, Lausanne 1015, Switzerland
| | - Ksenia J Groh
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Department of Environmental Toxicology, Dübendorf 8600, Switzerland
| | - Marc J-F Suter
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Department of Environmental Toxicology, Dübendorf 8600, Switzerland; ETHZ, Institute of Biogeochemistry and Pollutant Dynamics, Zürich 8092, Switzerland
| | - Kristin Schirmer
- Eawag, Swiss Federal Institute of Aquatic Science and Technology, Department of Environmental Toxicology, Dübendorf 8600, Switzerland; EPFL, School of Architecture, Civil and Environmental Engineering, Lausanne 1015, Switzerland; ETHZ, Institute of Biogeochemistry and Pollutant Dynamics, Zürich 8092, Switzerland.
| |
Collapse
|
137
|
Li Y, Urban A, Midura D, Simon HG, Wang QT. Proteomic characterization of epicardial-myocardial signaling reveals novel regulatory networks including a role for NF-κB in epicardial EMT. PLoS One 2017; 12:e0174563. [PMID: 28358917 PMCID: PMC5373538 DOI: 10.1371/journal.pone.0174563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/10/2017] [Indexed: 01/09/2023] Open
Abstract
Signaling between the epicardium and underlying myocardium is crucial for proper heart development. The complex molecular interactions and regulatory networks involved in this communication are not well understood. In this study, we integrated mass spectrometry with bioinformatics to systematically characterize the secretome of embryonic chicken EPDC-heart explant (EHE) co-cultures. The 150-protein secretome dataset established greatly expands the knowledge base of the molecular players involved in epicardial-myocardial signaling. We identified proteins and pathways that are implicated in epicardial-myocardial signaling for the first time, as well as new components of pathways that are known to regulate the crosstalk between epicardium and myocardium. The large size of the dataset enabled bioinformatics analysis to deduce networks for the regulation of specific biological processes and predicted signal transduction nodes within the networks. We performed functional analysis on one of the predicted nodes, NF-κB, and demonstrate that NF-κB activation is an essential step in TGFβ2/PDGFBB-induced cardiac epithelial-to-mesenchymal transition. In summary, we have generated a global perspective of epicardial-myocardial signaling for the first time, and our findings open exciting new avenues for investigating the molecular basis of heart development and regeneration.
Collapse
Affiliation(s)
- Yanyang Li
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Alexander Urban
- Department of Pediatrics, The Feinberg School of Medicine, Northwestern University, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
| | - Devin Midura
- Department of Pediatrics, The Feinberg School of Medicine, Northwestern University, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
| | - Hans-Georg Simon
- Department of Pediatrics, The Feinberg School of Medicine, Northwestern University, Stanley Manne Children’s Research Institute, Chicago, Illinois, United States of America
- * E-mail: (QTW); (HGS)
| | - Q. Tian Wang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: (QTW); (HGS)
| |
Collapse
|
138
|
Japanese Encephalitis Virus NS5 Inhibits Type I Interferon (IFN) Production by Blocking the Nuclear Translocation of IFN Regulatory Factor 3 and NF-κB. J Virol 2017; 91:JVI.00039-17. [PMID: 28179530 DOI: 10.1128/jvi.00039-17] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/31/2017] [Indexed: 12/24/2022] Open
Abstract
The type I interferon (IFN) response is part of the first-line defense against viral infection. To initiate replication, viruses have developed powerful evasion strategies to counteract host IFN responses. In the present study, we found that the Japanese encephalitis virus (JEV) NS5 protein could inhibit double-stranded RNA (dsRNA)-induced IFN-β expression in a dose-dependent manner. Our data further demonstrated that JEV NS5 suppressed the activation of the IFN transcriptional factors IFN regulatory factor 3 (IRF3) and NF-κB. However, there was no defect in the phosphorylation of IRF3 and degradation of IκB, an upstream inhibitor of NF-κB, upon NS5 expression, indicating a direct inhibition of the nuclear localization of IRF3 and NF-κB by NS5. Mechanistically, NS5 was shown to interact with the nuclear transport proteins KPNA2, KPNA3, and KPNA4, which competitively blocked the interaction of KPNA3 and KPNA4 with their cargo molecules, IRF3 and p65, a subunit of NF-κB, and thus inhibited the nuclear translocation of IRF3 and NF-κB. Furthermore, overexpression of KPNA3 and KPNA4 restored the activity of IRF3 and NF-κB and increased the production of IFN-β in NS5-expressing or JEV-infected cells. Additionally, an upregulated replication level of JEV was shown upon KPNA3 or KPNA4 overexpression. These results suggest that JEV NS5 inhibits the induction of type I IFN by targeting KPNA3 and KPNA4.IMPORTANCE JEV is the major cause of viral encephalitis in South and Southeast Asia, with high mortality. However, the molecular mechanisms contributing to the severe pathogenesis are poorly understood. The ability of JEV to counteract the host innate immune response is potentially one of the mechanisms responsible for JEV virulence. Here we demonstrate the ability of JEV NS5 to interfere with the dsRNA-induced nuclear translocation of IRF3 and NF-κB by competitively inhibiting the interaction of IRF3 and NF-κB with nuclear transport proteins. Via this mechanism, JEV NS5 suppresses the induction of type I IFN and the antiviral response in host cells. These findings reveal a novel strategy for JEV to escape the host innate immune response and provide new insights into the pathogenesis of JEV.
Collapse
|
139
|
Wang Q, Yan C, Xin M, Han L, Zhang Y, Sun M. Sirtuin 1 (Sirt1) Overexpression in BaF3 Cells Contributes to Cell Proliferation Promotion, Apoptosis Resistance and Pro-Inflammatory Cytokine Production. Med Sci Monit 2017; 23:1477-1482. [PMID: 28346398 PMCID: PMC5380195 DOI: 10.12659/msm.900754] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background B lymphocyte hyperactivity is a main characteristic of systemic lupus erythematosus (SLE), and B lymphocytes play a prominent pathogenic role in the development and progression of SLE. The aim of this study was to investigate the role of Sirtuin 1 (Sirt1) in B lymphocytes. Material/Methods Mouse B lymphocytes BaF3 was transfected with Sirt1 vector or shRNA against Sirt1. Then the transfected cells viability and apoptosis were respectively determined by MTT assay and flow cytometry. In addition, the mRNA levels of three pro-inflammatory cytokines and p53 were detected by RT-PCR. Furthermore, the expression levels of nuclear factor-kappa B (NF-κB) pathway proteins were measured by Western blot. Results Overexpression of Sirt1 significantly increased cell proliferation (p<0.05 or p<0.01) and significantly suppressed apoptosis (p<0.05). The mRNA level expressions of interleukin 1 (IL-1), IL-6, and tumor necrosis factor-α (TNF-α) were significantly upregulated (p<0.05 or p<0.01), whereas p53 was significantly downregulated (p<0.05) by Sirt1 overexpression. In addition, the inhibitory subunit of NF-κB (IκBα) and p65 were significantly activated and phosphorylated (p<0.01 or p<0.001), and B-Cell CLL/Lymphoma 3 (Bcl-3) was significantly upregulated (p<0.05) by Sirt1 overexpression. Conclusions These results suggested that Sirt1 overexpression could promote BaF3 cell proliferation, inhibit apoptosis, and upregulate pro-inflammatory cytokines. The NF-κB pathway might be involved in these effects of Sirt1 on BaF3 cells, and Sirt1 might be a potential risk factor of SLE.
Collapse
Affiliation(s)
- Qian Wang
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Chao Yan
- Department of Rheumatology, Qilu Hospital of Shandong University (Qingdao Branch), Qingdao, Shandong, China (mainland)
| | - Miaomiao Xin
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Li Han
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Yunqing Zhang
- Department of Rheumatology, Qilu Hospital of Shandong University (Qingdao Branch), Qingdao, Shandong, China (mainland)
| | - Mingshu Sun
- Department of Rheumatology and Clinical Immunology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
140
|
Kim MJ, Kim WS, Kim DO, Byun JE, Huy H, Lee SY, Song HY, Park YJ, Kim TD, Yoon SR, Choi EJ, Ha H, Jung H, Choi I. Macrophage migration inhibitory factor interacts with thioredoxin-interacting protein and induces NF-κB activity. Cell Signal 2017; 34:110-120. [PMID: 28323005 DOI: 10.1016/j.cellsig.2017.03.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/09/2017] [Accepted: 03/16/2017] [Indexed: 12/27/2022]
Abstract
The nuclear factor kappa B (NF-κB) pathway is pivotal in controlling survival and apoptosis of cancer cells. Macrophage migration inhibitory factor (MIF), a cytokine that regulates the immune response and tumorigenesis under inflammatory conditions, is upregulated in various tumors. However, the intracellular functions of MIF are unclear. In this study, we found that MIF directly interacted with thioredoxin-interacting protein (TXNIP), a tumor suppressor and known inhibitor of NF-κB activity, and MIF significantly induced NF-κB activation. MIF competed with TXNIP for NF-κB activation, and the intracellular MIF induced NF-κB target genes, including c-IAP2, Bcl-xL, ICAM-1, MMP2 and uPA, by inhibiting the interactions between TXNIP and HDACs or p65. Furthermore, we identified the interaction motifs between MIF and TXNIP via site-directed mutagenesis of their cysteine (Cys) residues. Cys57 and Cys81 of MIF and Cys36 and Cys120 of TXNIP were responsible for the interaction. MIF reversed the TXNIP-induced suppression of cell proliferation and migration. Overall, we suggest that MIF induces NF-κB activity by counter acting the inhibitory effect of TXNIP on the NF-κB pathway via direct interaction with TXNIP. These findings reveal a novel intracellular function of MIF in the progression of cancer.
Collapse
Affiliation(s)
- Mi Jeong Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Won Sam Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Dong Oh Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Jae-Eun Byun
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hangsak Huy
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Soo Yun Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Hae Young Song
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Young-Jun Park
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Tae-Don Kim
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Suk Ran Yoon
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Eun-Ji Choi
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hyunjung Ha
- Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Haiyoung Jung
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea.
| | - Inpyo Choi
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseong-gu, Daejeon 34141, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
141
|
Melnik BC, Schmitz G. Milk's Role as an Epigenetic Regulator in Health and Disease. Diseases 2017; 5:diseases5010012. [PMID: 28933365 PMCID: PMC5456335 DOI: 10.3390/diseases5010012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/02/2017] [Accepted: 03/07/2017] [Indexed: 12/16/2022] Open
Abstract
It is the intention of this review to characterize milk's role as an epigenetic regulator in health and disease. Based on translational research, we identify milk as a major epigenetic modulator of gene expression of the milk recipient. Milk is presented as an epigenetic "doping system" of mammalian development. Milk exosome-derived micro-ribonucleic acids (miRNAs) that target DNA methyltransferases are implicated to play the key role in the upregulation of developmental genes such as FTO, INS, and IGF1. In contrast to miRNA-deficient infant formula, breastfeeding via physiological miRNA transfer provides the appropriate signals for adequate epigenetic programming of the newborn infant. Whereas breastfeeding is restricted to the lactation period, continued consumption of cow's milk results in persistent epigenetic upregulation of genes critically involved in the development of diseases of civilization such as diabesity, neurodegeneration, and cancer. We hypothesize that the same miRNAs that epigenetically increase lactation, upregulate gene expression of the milk recipient via milk-derived miRNAs. It is of critical concern that persistent consumption of pasteurized cow's milk contaminates the human food chain with bovine miRNAs, that are identical to their human analogs. Commercial interest to enhance dairy lactation performance may further increase the epigenetic miRNA burden for the milk consumer.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, Faculty of Human Sciences, University of Osnabrück, Am Finkenhügel 7a, D-49076 Osnabrück, Germany.
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, University of Regensburg, Franz-Josef-Strauß-Allee 11, D-93053 Regensburg, Germany.
| |
Collapse
|
142
|
Park J, Min JS, Chae U, Lee JY, Song KS, Lee HS, Lee HJ, Lee SR, Lee DS. Anti-inflammatory effect of oleuropein on microglia through regulation of Drp1-dependent mitochondrial fission. J Neuroimmunol 2017; 306:46-52. [PMID: 28385187 DOI: 10.1016/j.jneuroim.2017.02.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 01/25/2023]
Abstract
Oleuropein is a primary phenolic compound found in olive leaf and Fraxinus rhynchophylla. Here, we investigated the impact of oleuropein on LPS-induced BV-2 microglial cells. Oleuropein suppressed the LPS-induced increase in pro-inflammatory mediators, such as nitric oxide, and pro-inflammatory cytokines, via inhibition of ERK/p38/NF-κB activation and reactive oxygen species (ROS) generation. Furthermore, it suppressed LPS-induced excessive mitochondrial fission, which regulates mitochondrial ROS generation and pro-inflammatory response by diminishing Drp1 dephosphorylation. Collectively, we demonstrated that oleuropein suppresses pro-inflammatory response of microglia by inhibiting Drp1-dependent mitochondrial fission. Our findings suggest a potential role of oleuropein in microglial inflammation-mediated neurodegenerative disorders.
Collapse
Affiliation(s)
- Junghyung Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Ju-Sik Min
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea; Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Unbin Chae
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Yeop Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea; Traditional Korean Medicine Technology Division, National Development Institute of Korean Medicine, Gyeongsangbuk-do, Republic of Korea
| | - Kyung-Sik Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hong Jun Lee
- Biomedical Research Institute, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
143
|
Global decrease in the expression of signalling pathways’ genes in murine uterus during preimplantation pregnancy. Reprod Biol 2017; 17:89-96. [DOI: 10.1016/j.repbio.2017.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/12/2016] [Accepted: 01/04/2017] [Indexed: 01/25/2023]
|
144
|
|
145
|
Suppression of LPS-induced NF-κB activity in macrophages by the synthetic aurone, (Z)-2-((5-(hydroxymethyl) furan-2-yl) methylene) benzofuran-3(2H)-one. Int Immunopharmacol 2017; 43:116-128. [DOI: 10.1016/j.intimp.2016.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/23/2016] [Accepted: 12/03/2016] [Indexed: 11/21/2022]
|
146
|
Deng Z, Wang Z, Jin J, Wang Y, Bao N, Gao Q, Zhao J. SIRT1 protects osteoblasts against particle-induced inflammatory responses and apoptosis in aseptic prosthesis loosening. Acta Biomater 2017; 49:541-554. [PMID: 27890623 DOI: 10.1016/j.actbio.2016.11.051] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/19/2016] [Accepted: 11/22/2016] [Indexed: 02/07/2023]
Abstract
We hypothesized that SIRT1 downregulation in osteoblasts induced by wear particles was one of the reasons for particle-induced osteolysis (PIO) in total joint arthroplasty failure. In the present study, the expression of SIRT1 was examined in osteoblasts treated with TiAl6V4 particles (TiPs) and CoCrMo particles (CoPs) from materials used in prosthetics and specimens from PIO animal models. To address whether SIRT1 downregulation triggers inflammatory responses and apoptosis in osteoblasts, the effect of a SIRT1 activator, resveratrol on the expression of inflammatory cytokines and apoptosis in particle-treated osteoblasts was tested. The results demonstrated that SIRT1 expression was significantly downregulated in particle-treated osteoblasts and PIO animal models. Both pharmacological activation and overexpression of SIRT1 dramatically reduced the particle-induced expression of inflammatory cytokines and osteoblast apoptosis through NF-κB and p53 signaling, respectively. Furthermore, in PIO animal models, resveratrol significantly reduced the severity of osteolysis. Collectively, the results of the present study indicated that SIRT1 plays a vital role in the pathogenesis of aseptic loosening, and further treatment targeted at SIRT1 possibly lead to novel approaches for prevention of aseptic prosthesis loosening. STATEMENT OF SIGNIFICANCE Aseptic loosening is the most common cause of total hip arthroplasty (THA) and total knee arthroplasty (TKA) failure and revision surgery. However, there is still no effective therapeutic target in the clinical treatment. Besides, the underlying mechanism of aseptic loosening is largely unknown. The result of our study indicated that SIRT1 has the ability to effectively regulate the wear particle-induced inflammatory responses, apoptosis, osteolysis in particle-stimulated osteoblasts and particle-induced osteolysis animal models. Our study provides a potential target for the prevention and treatment of aseptic loosening and further investigated the underlying mechanism of aseptic loosening, which may make contribution to decrease the incidence of THA and TKA failure in the clinical practice.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China; Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Zhenheng Wang
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| | - Jiewen Jin
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Yong Wang
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Nirong Bao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| | - Qian Gao
- Center for Translational Medicine, Nanjing University Medical School, Nanjing, Jiangsu, PR China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, PR China.
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
147
|
Fowler KA, Jania CM, Tilley SL, Panoskaltsis-Mortari A, Baldwin AS, Serody JS, Coghill JM. Targeting the Canonical Nuclear Factor-κB Pathway with a High-Potency IKK2 Inhibitor Improves Outcomes in a Mouse Model of Idiopathic Pneumonia Syndrome. Biol Blood Marrow Transplant 2017; 23:569-580. [PMID: 28161607 DOI: 10.1016/j.bbmt.2017.01.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/20/2017] [Indexed: 10/20/2022]
Abstract
Idiopathic pneumonia syndrome (IPS) is a noninfectious inflammatory disorder of the lungs that occurs most often after fully myeloablative allogeneic hematopoietic stem cell transplantation (HSCT). IPS can be severe and is associated with high 1-year mortality rates despite existing therapies. The canonical nuclear factor-(NF) κB signaling pathway has previously been linked to several inflammatory disorders of the lung, including asthma and lung allograft rejection. It has never been specifically targeted as a novel IPS treatment approach, however. Here, we report that the IκB kinase 2 (IKK2) antagonist BAY 65-5811 or "compound A," a highly potent and specific inhibitor of the NF-κB pathway, was able to improve median survival times and recipient oxygenation in a well-described mouse model of IPS. Compound A impaired the production of the proinflammatory chemokines CCL2 and CCL5 within the host lung after transplantation. This resulted in significantly lower numbers of donor lung infiltrating CD4+ and CD8+ T cells and reduced pulmonary inflammatory cytokine production after allograft. Compound A's beneficial effects appeared to be specific for limiting pulmonary injury, as the drug was unable to improve outcomes in a B6 into B6D2 haplotype-matched murine HSCT model in which recipient mice succumb to lethal acute graft-versus-host disease of the gastrointestinal tract. Collectively, our data suggest that the targeting of the canonical NF-κB pathway with a small molecule IKK2 antagonist may represent an effective and novel therapy for the specific management of acute lung injury that can occur after allogeneic HSCT.
Collapse
Affiliation(s)
- Kenneth A Fowler
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Corey M Jania
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephen L Tilley
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Serody
- Lineberger Comprehensive Cancer Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - James M Coghill
- Lineberger Comprehensive Cancer Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
148
|
Promising Role of Toll-Like Receptor 8 Agonist in Concert with Prostratin for Activation of Silent HIV. J Virol 2017; 91:JVI.02084-16. [PMID: 27928016 DOI: 10.1128/jvi.02084-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 11/29/2016] [Indexed: 01/03/2023] Open
Abstract
The persistence of latently HIV-infected cells in patients under combined antiretroviral treatment (cART) remains the major hurdle for HIV eradication. Thus far, individual compounds have not been sufficiently potent to reactivate latent virus and guarantee its elimination in vivo. Thus, we hypothesized that transcriptional enhancers, in concert with compounds triggering the innate immune system, are more efficient in reversing latency by creating a Th1 supportive milieu that acts against latently HIV-infected cells at various levels. To test our hypothesis, we screened six compounds on a coculture of latently infected cells (J-lat) and monocyte-derived dendritic cells (MDDCs). The protein kinase C (PKC) agonist prostratin, with a Toll-like receptor 8 (TLR8) agonist, resulted in greater reversion of HIV latency than any single compound. This combinatorial approach led to a drastic phenotypic and functional maturation of the MDDCs. Tumor necrosis factor (TNF) and cell-cell interactions were crucial for the greater reversion observed. Similarly, we found a greater potency of the combination of prostratin and TLR8 agonist in reversing HIV latency when applying it to primary cells of HIV-infected patients. Thus, we demonstrate here the synergistic interplay between TLR8-matured MDDCs and compounds acting directly on latently HIV-infected cells, targeting different mechanisms of latency, by triggering various signaling pathways. Moreover, TLR8 triggering may reverse exhaustion of HIV-specific cytotoxic T lymphocytes that might be essential for killing or constraining the latently infected cells. IMPORTANCE Curing HIV is the Holy Grail. The so-called "shock and kill" strategy relies on drug-mediated reversion of HIV latency and the subsequent death of those cells under combined antiretroviral treatment. So far, no compound achieves efficient reversal of latency or eliminates this latent reservoir. The compounds may not target all of the latency mechanisms in all latently infected cells. Moreover, HIV-associated exhaustion of the immune system hinders the efficient elimination of the reactivated cells. In this study, we demonstrated synergistic latency reversion by combining agonists for protein kinase C and Toll-like receptor 8 in a coculture of latently infected cells with myeloid dendritic cells. The drug prostratin stimulates directly the transcriptional machinery of latently infected cells, and the TLR8 agonist acts indirectly by maturing dendritic cells. These findings highlight the importance of the immune system and its activation, in combination with direct-acting compounds, to reverse latency.
Collapse
|
149
|
Ma Y, Zhang B, Wang D, Qian L, Song X, Wang X, Yang C, Zhao G. HTLV-1 basic leucine zipper factor downregulates cyclin D1 expression via interactions with NF-κB. Int J Mol Med 2017; 39:764-770. [PMID: 28204810 DOI: 10.3892/ijmm.2017.2868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 01/20/2017] [Indexed: 11/06/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus. It can cause adult T cell leukemia (ATL) and other diseases. The HTLV-1 basic leucine zipper (bZIP) factor (HBZ), which is encoded by the minus-strand of the provirus, is expressed in all cases of ATL and involved in T cell proliferation. However, the exact mechanism underlying its growth-promoting activity is poorly understood. Herein, we demonstrated that HBZ suppressed cyclin D1 expression by inhibiting the nuclear factor (NF)-κB signaling pathway. Among the potential mechanisms of cyclin D1 inhibition mediated by HBZ, we found that HBZ suppressed cyclin D1 promoter activity. Luciferase assay analysis revealed that HBZ repressed cyclin D1 promoter activity by suppressing NF-κB‑driven transcription mediated by the p65 subunit. Using an immunoprecipitation assay, we found that HBZ could bind to p65, but not p50. Finally, we showed that HBZ selectively interacted with p65 via its AD+bZIP domains. By suppressing cyclin D1 expression, HBZ can alter cell cycle progression of HTLV-1-infected cells, which may be critical for oncogenesis.
Collapse
Affiliation(s)
- Yunyun Ma
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Bo Zhang
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Dong Wang
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Lili Qian
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Xianmei Song
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Xueyin Wang
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Chaokuan Yang
- Henan Medical College, Zhengzhou, Henan 451191, P.R. China
| | - Guoqiang Zhao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
150
|
Uchio R, Higashi Y, Kohama Y, Kawasaki K, Hirao T, Muroyama K, Murosaki S. A hot water extract of turmeric ( Curcuma longa) suppresses acute ethanol-induced liver injury in mice by inhibiting hepatic oxidative stress and inflammatory cytokine production. J Nutr Sci 2017; 6:e3. [PMID: 28620478 PMCID: PMC5465857 DOI: 10.1017/jns.2016.43] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 11/20/2016] [Accepted: 11/28/2016] [Indexed: 01/10/2023] Open
Abstract
Turmeric (Curcuma longa) is a widely used spice that has various biological effects, and aqueous extracts of turmeric exhibit potent antioxidant activity and anti-inflammatory activity. Bisacurone, a component of turmeric extract, is known to have similar effects. Oxidative stress and inflammatory cytokines play an important role in ethanol-induced liver injury. This study was performed to evaluate the influence of a hot water extract of C. longa (WEC) or bisacurone on acute ethanol-induced liver injury. C57BL/6 mice were orally administered WEC (20 mg/kg body weight; BW) or bisacurone (60 µg/kg BW) at 30 min before a single dose of ethanol was given by oral administration (3·0 g/kg BW). Plasma levels of aspartate aminotransferase and alanine aminotransferase were markedly increased in ethanol-treated mice, while the increase of these enzymes was significantly suppressed by prior administration of WEC. The increase of alanine aminotransferase was also significantly suppressed by pretreatment with bisacurone. Compared with control mice, animals given WEC had higher hepatic tissue levels of superoxide dismutase and glutathione, as well as lower hepatic tissue levels of thiobarbituric acid-reactive substances, TNF-α protein and IL-6 mRNA. These results suggest that oral administration of WEC may have a protective effect against ethanol-induced liver injury by suppressing hepatic oxidation and inflammation, at least partly through the effects of bisacurone.
Collapse
Key Words
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- BW, body weight
- Bisacurone
- Ethanol-induced liver injury
- GSH, glutathione
- GSSG, oxidised glutathione
- Inflammatory cytokines
- O2•−, superoxide anion radical
- Oxidative stress
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- TBARS, thiobarbituric acid-reactive substances
- Turmeric (Curcuma longa)
- WEC, hot water extract of Curcuma longa
Collapse
Affiliation(s)
- Ryusei Uchio
- Research & Development Institute, House Wellness Foods Corporation, 3–20 Imoji, Itami 664-0011, Japan
| | - Yohei Higashi
- Research & Development Institute, House Wellness Foods Corporation, 3–20 Imoji, Itami 664-0011, Japan
| | - Yusuke Kohama
- Central Research & Development Institute, House Foods Group Inc., 1–4 Takanodai, Yotsukaido 284-0033, Japan
| | - Kengo Kawasaki
- Research & Development Institute, House Wellness Foods Corporation, 3–20 Imoji, Itami 664-0011, Japan
| | - Takashi Hirao
- Central Research & Development Institute, House Foods Group Inc., 1–4 Takanodai, Yotsukaido 284-0033, Japan
| | - Koutarou Muroyama
- Research & Development Institute, House Wellness Foods Corporation, 3–20 Imoji, Itami 664-0011, Japan
| | - Shinji Murosaki
- Research & Development Institute, House Wellness Foods Corporation, 3–20 Imoji, Itami 664-0011, Japan
| |
Collapse
|