251
|
Sharifi L, Moshiri M, Dallal MM, Asgardoon MH, Nourizadeh M, Bokaie S, Mirshafiey A. The Inhibitory Role of M2000 (β-D-Mannuronic Acid) on Expression of Toll-like Receptor 2 and 4 in HT29 Cell Line. RECENT PATENTS ON INFLAMMATION & ALLERGY DRUG DISCOVERY 2019; 13:57-65. [PMID: 30539708 PMCID: PMC6778985 DOI: 10.2174/1872213x13666181211160238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 12/01/2018] [Accepted: 12/04/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND/OBJECTIVES Anti-inflammatory agents play a crucial role in controlling inflammatory diseases such as Inflammatory Bowel Disease (IBD) but their use is restricted due to their vast side effects. M2000 (β-D-mannuronic acid) is a new immunomodulatory drug. According to the capacity of M2000 in suppressing some molecules involved in Toll Like Receptors (TLRs) signaling and reducing oxidative stress we hypothesize that, this molecule may have a potential role in decreasing inflammatory responses in IBD. The aim of this study was to evaluate the cytotoxicity of M2000 and its effect on the gene expression of TLR2 and TLR4. METHODS HEK293 cell line was grown and divided into 96-well cell plate and MTT assay was performed. HT29 cells were cultured and treated with low and high doses of M2000. Total RNA was extracted and cDNA synthesized and quantitative real-time PCR was done to quantify the TLR2 and TLR4 mRNA expression. RESULTS We found that M2000 at the concentration of ≤ 1000µg/ml had no obvious cytotoxicity effect on the HEK293 cells. Also, low and high doses of M2000 could significantly down-regulate both TLR2 and TLR4 mRNA expression. Moreover, a significant reduction in gene expression of TLR2 and TLR4 in an inflammatory condition resulted in high doses of M2000 in the presence of LPS. CONCLUSION Our study which was conducted in colonic epithelial cell model, shows that M2000 can be considered as a new anti-inflammatory agent in IBD. However, more comprehensive experimental and clinical studies are required to recognize the molecular mechanism of M2000 and also its safety and efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Abbas Mirshafiey
- Address correspondence to this author at the Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Tel/Fax: +98 (21) 88954913; E-mail:
| |
Collapse
|
252
|
Ma J, Chen X, Xin G, Li X. Chronic exposure to the ionic liquid [C 8mim]Br induces inflammation in silver carp spleen: Involvement of oxidative stress-mediated p38MAPK/NF-κB signalling and microRNAs. FISH & SHELLFISH IMMUNOLOGY 2019; 84:627-638. [PMID: 30343007 DOI: 10.1016/j.fsi.2018.09.052] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 06/08/2023]
Abstract
The present study aimed to determine the chronic toxicity of 1-methyl-3-octylimidazolium bromide ([C8mim]Br) on the silver carp to further reveal the toxicological mechanisms of ionic liquids. Chronic exposure of silver carp to [C8mim]Br at concentrations of 1.095 and 4.380 mg/L for 60 d was conducted under laboratory conditions. The results revealed that chronic exposure to [C8mim]Br inhibited the activity of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) and reduced glutathione (GSH) levels while markedly increasing malondialdehyde (MDA) and protein carbonyl (PC) levels in fish spleen, indicating that [C8mim]Br treatment induced oxidative stress. Additionally, long-term exposure to [C8mim]Br markedly upregulated the expressions of nuclear factor-κB (NF-κB), inducible nitric oxide synthase (iNOS), interleukin-1β (IL-1β), IL-6, tumour necrosis factor-α (TNF-α), and interferon-γ (IFN-γ); altered the levels of transforming growth factor-β (TGF-β); and increased the mRNA levels of p38MAPK, c-fos, c-jun, and c-myc, suggesting that long-term exposure to [C8mim]Br might promote the inflammatory response in fish spleen and that p38MAPK/NF-κB signalling may potentially be involved in this process. Moreover, [C8mim]Br-exposure altered lysozyme activity and complement 3 (C3) and immunoglobulin M (IgM) content, indicating that chronic [C8mim]Br exposure also has immunotoxic effects on silver carp. Furthermore, we also found that [C8mim]Br exposure reduced miR-125b levels, altered miR-143 levels, and upregulated miR-155 and miR-21 levels, suggesting that these miRNAs may be involved in the [C8mim]Br-induced inflammatory response in fish spleen. In summary, the present study indicates that chronic exposure to [C8mim]Br induces inflammation in fish spleen and that oxidative stress-mediated p38MAPK/NF-κB signalling and miRNAs may play a key role in this process.
Collapse
Affiliation(s)
- Junguo Ma
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xi Chen
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Guangyuan Xin
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Xiaoyu Li
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| |
Collapse
|
253
|
Regulation of TLR signaling pathways by microRNAs: implications in inflammatory diseases. Cent Eur J Immunol 2018; 43:482-489. [PMID: 30799997 PMCID: PMC6384427 DOI: 10.5114/ceji.2018.81351] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/13/2017] [Indexed: 12/19/2022] Open
Abstract
The control of the immune response during the development of some diseases is crucial for the maintenance or restoration of homeostasis. Several mechanisms can initiate inflammation, one of which is the activation of toll-like receptors (TLRs), necessary to initiate the immune response to eliminate an infection. However, inappropriate activation can compromise immunological homeostasis, leading to pathologies such as autoimmune diseases, chronic inflammation, and even cancer. Regulatory mechanisms that intervene in the initiation or modulation of inflammation include microRNAs (miRNAs), which have emerged as key post-transcriptional regulators of proteins involved in distinct cellular processes, such as regulation of the immune response. The focus of this review is on the diverse roles of miRNAs in the regulation of TLR-signaling pathways by targeting multiple molecules, including TLRs, the signaling proteins and cytokines induced by TLRs. It will also address the relationships of these molecules with some diseases that involve inflammation such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), cancer, as well as bacterial or viral infections.
Collapse
|
254
|
van Dalen FJ, van Stevendaal MHME, Fennemann FL, Verdoes M, Ilina O. Molecular Repolarisation of Tumour-Associated Macrophages. Molecules 2018; 24:molecules24010009. [PMID: 30577495 PMCID: PMC6337345 DOI: 10.3390/molecules24010009] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
The tumour microenvironment (TME) is composed of extracellular matrix and non-mutated cells supporting tumour growth and development. Tumour-associated macrophages (TAMs) are among the most abundant immune cells in the TME and are responsible for the onset of a smouldering inflammation. TAMs play a pivotal role in oncogenic processes as tumour proliferation, angiogenesis and metastasis, and they provide a barrier against the cytotoxic effector function of T lymphocytes and natural killer (NK) cells. However, TAMs are highly plastic cells that can adopt either pro- or anti-inflammatory roles in response to environmental cues. Consequently, TAMs represent an attractive target to recalibrate immune responses in the TME. Initial TAM-targeted strategies, such as macrophage depletion or disruption of TAM recruitment, have shown beneficial effects in preclinical models and clinical trials. Alternatively, reprogramming TAMs towards a proinflammatory and tumouricidal phenotype has become an attractive strategy in immunotherapy. This work summarises the molecular wheelwork of macrophage biology and presents an overview of molecular strategies to repolarise TAMs in immunotherapy.
Collapse
Affiliation(s)
- Floris J van Dalen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Marleen H M E van Stevendaal
- Department of Bio-Organic Chemistry, Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
| | - Felix L Fennemann
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Olga Ilina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
255
|
Meroni M, Longo M, Rametta R, Dongiovanni P. Genetic and Epigenetic Modifiers of Alcoholic Liver Disease. Int J Mol Sci 2018; 19:E3857. [PMID: 30513996 PMCID: PMC6320903 DOI: 10.3390/ijms19123857] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/31/2018] [Accepted: 11/28/2018] [Indexed: 12/12/2022] Open
Abstract
Alcoholic liver disease (ALD), a disorder caused by excessive alcohol consumption is a global health issue. More than two billion people consume alcohol in the world and about 75 million are classified as having alcohol disorders. ALD embraces a wide spectrum of hepatic lesions including steatosis, alcoholic steatohepatitis (ASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). ALD is a complex disease where environmental, genetic, and epigenetic factors contribute to its pathogenesis and progression. The severity of alcohol-induced liver disease depends on the amount, method of usage and duration of alcohol consumption as well as on age, gender, presence of obesity, and genetic susceptibility. Genome-wide association studies and candidate gene studies have identified genetic modifiers of ALD that can be exploited as non-invasive biomarkers, but which do not completely explain the phenotypic variability. Indeed, ALD development and progression is also modulated by epigenetic factors. The premise of this review is to discuss the role of genetic variants and epigenetic modifications, with particular attention being paid to microRNAs, as pathogenic markers, risk predictors, and therapeutic targets in ALD.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| | - Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| | - Raffaela Rametta
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
256
|
He R, Li Y, Zhou L, Su X, Li Y, Pan P, Hu C. miR-146b overexpression ameliorates lipopolysaccharide-induced acute lung injury in vivo and in vitro. J Cell Biochem 2018; 120:2929-2939. [PMID: 30500983 DOI: 10.1002/jcb.26846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a type of acute lung injury (ALI), which causes high morbidity and mortality. So far, effective clinical treatment of ARDS is still limited. Recently, miR-146b has been reported to play a key role in inflammation. In the present study, we evaluated the functional role of miR-146b in ARDS using the murine model of lipopolysaccharide (LPS)-induced ALI. The miR-146b expression could be induced by LPS stimulation, and miR-146b overexpression was required in the maintenance of body weight and survival of ALI mice; after miR-146b overexpression, LPS-induced lung injury, pulmonary inflammation, total cell and neutrophil counts, proinflammatory cytokines, and chemokines in bronchial alveolar lavage (BAL) fluid were significantly reduced. The promotive effect of LPS on lung permeability through increasing total protein, albumin and IgM in BAL fluid could be partially reversed by miR-146b overexpression. Moreover, in murine alveolar macrophages, miR-146b overexpression reduced LPS-induced TNF-α and interleukin (IL)-1β releasing. Taken together, we demonstrated that miR-146b overexpression could effectively improve the LPS-induced ALI; miR-146b is a promising target in ARDS treatment.
Collapse
Affiliation(s)
- Ruoxi He
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Li Zhou
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
257
|
Wang X, Zheng Y, Ma Y, Du L, Chu F, Gu H, Dahlgren RA, Li Y, Wang H. Lipid metabolism disorder induced by up-regulation of miR-125b and miR-144 following β-diketone antibiotic exposure to F0-zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 164:243-252. [PMID: 30121499 DOI: 10.1016/j.ecoenv.2018.08.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 06/08/2023]
Abstract
β-Diketone antibiotics (DKAs) are widely used in human and veterinary medicine to prevent and treat a large variety of infectious diseases. Long-term DKA exposure to zebrafish can result in lipid metabolism disorders and liver function abnormalities. Based on our previous miRNA-seq analyses, miR-144 and miR-125b were identified as target genes regulating lipid metabolism. DKA-exposure at 12.5 and 25 mg/L significantly increased the expressions of miR-144 and miR-125b. The expression levels for the two miRNAs exhibited an inverse relationship with their lipid-metabolism-related target genes (ppardb, bcl2a, pparaa and pparda). Over-expression and inhibition of miR-144 and miR-125b were observed by micro-injection of agomir-144, agomir-125b, antagomir-144 and antagomir-125b. The over-expression of miR-144 and miR-125b enhanced lipid accumulation and further induced lipid-metabolism-disorder syndrome in F1-zebrafish. The expression of ppardb and bcl2a in whole-mount in situ hybridization was in general agreement with results from qRT-PCR and was concentration-dependent. Oil red O and H&E staining, as well as related physiological and biochemical indexes, showed that chronic DKA exposure resulted in lipid-metabolism-disorder in F0-adults, and in F1-larvae fat accumulation, increased lipid content, abnormal liver function and obesity. The abnormal levels of triglyceride (TG) and total cholesterol (TCH) in DKA-exposed zebrafish increased the risk of hyperlipidemia, atherosclerosis and coronary heart disease. These observations improve our understanding of mechanisms leading to liver disease from exposure to environmental pollution, thereby having relevant practical significance in health prevention, early intervention, and gene therapy for drug-induced diseases.
Collapse
Affiliation(s)
- Xuedong Wang
- Jiangsu Key Laboratory of Environmental Science and Engineering, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yan Ma
- College of Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Liyang Du
- Jiangsu Key Laboratory of Environmental Science and Engineering, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Fangyu Chu
- Jiangsu Key Laboratory of Environmental Science and Engineering, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Haidong Gu
- Jiangsu Key Laboratory of Environmental Science and Engineering, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Randy A Dahlgren
- Department of Land, Air and Water Resources, University of California, Davis, CA 95616, USA
| | - Yanyan Li
- College of Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Huili Wang
- Jiangsu Key Laboratory of Environmental Science and Engineering, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
258
|
Vergadi E, Vaporidi K, Tsatsanis C. Regulation of Endotoxin Tolerance and Compensatory Anti-inflammatory Response Syndrome by Non-coding RNAs. Front Immunol 2018; 9:2705. [PMID: 30515175 PMCID: PMC6255943 DOI: 10.3389/fimmu.2018.02705] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022] Open
Abstract
The onset and the termination of innate immune response must be tightly regulated to maintain homeostasis and prevent excessive inflammation, which can be detrimental to the organism, particularly in the context of sepsis. Endotoxin tolerance and compensatory anti-inflammatory response syndrome (CARS) describe a state of hypo-responsiveness characterized by reduced capacity of myeloid cells to respond to inflammatory stimuli, particularly those initiated by bacterial lipopolysaccharide (LPS). To achieve endotoxin tolerance, extensive reprogramming otherwise termed as “innate immune training”, is required that leads to both modifications of the intracellular components of TLR signaling and also to alterations in extracellular soluble mediators. Non-coding RNAs (ncRNAs) have been recognized as critical regulators of TLR signaling. Specifically, several microRNAs (miR-146, miR-125b, miR-98, miR-579, miR-132, let-7e and others) are induced upon TLR activation and reciprocally promote endotoxin tolerance and/or cross tolerance. Many other miRNAs have been also shown to negatively regulate TLR signaling. The long non-coding (lnc)RNAs (Mirt2, THRIL, MALAT1, lincRNA-21 and others) are also altered upon TLR activation and negatively regulate TLR signaling. Furthermore, the promotion or termination of myeloid cell tolerance is not only regulated by intracellular mediators but is also affected by other TLR-independent soluble signals that often achieve their effect via modulation of intracellular ncRNAs. In this article, we review recent evidence on the role of different ncRNAs in the context of innate immune cell tolerance and trained immunity, and evaluate their impact on immune system homeostasis.
Collapse
Affiliation(s)
- Eleni Vergadi
- Department of Paediatrics, Medical School, University of Crete, Heraklion, Greece.,Department of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, Medical School, University of Crete, Heraklion, Greece
| | - Christos Tsatsanis
- Department of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
259
|
Li S, Sun Y, Zhong L, Xiao Z, Yang M, Chen M, Wang C, Xie X, Chen X. The suppression of ox-LDL-induced inflammatory cytokine release and apoptosis of HCAECs by long non-coding RNA-MALAT1 via regulating microRNA-155/SOCS1 pathway. Nutr Metab Cardiovasc Dis 2018; 28:1175-1187. [PMID: 30314869 DOI: 10.1016/j.numecd.2018.06.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/23/2018] [Accepted: 06/12/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease. Accumulating evidence suggests that long non-coding RNAs (lncRNAs) and microRNAs have emerged as critical regulators of atherosclerosis; however, whether they have crosstalk on this issue remains elusive. Here, we investigated the potential associations between lncRNA-MALAT1 and miR-155 on the regulation of atherosclerosis. METHODS Quantitative real-time PCR was employed to assess the expression of MALAT1, IL-6 and IL-8. ELISA was performed to measure the secretion of IL-6 and IL-8. MTT assay was used to determine the proliferation of Human Coronary Artery Endothelial Cells (HCAECs). Flow cytometry was used to measure the cell apoptosis. Western blot was used to assess the expression of apoptosis-related proteins and the phosphorylation of STAT1 and STAT3. RESULTS We found that the pro-inflammatory cytokine release and the apoptosis of HCAECs were elevated upon ox-LDL treatment, while MALAT1 expression was also up regulated. Knocking down of MALAT1 boosted ox-LDL-induced cytokine release and apoptosis of HCAECs. The binding site of miR-155 in MALAT1 sequence was confirmed by dual luciferase assay. Furthermore, miR-155 inhibition significantly repressed ox-LDL mediated inflammation and apoptosis of HCAECs via SOCS1. At last, we found that MALAT1 could suppress the inflammatory cytokine release and cell apoptosis via sponging miR-155 to increase SOCS1 level, which in turn restrained JAK-STAT pathway. CONCLUSION In summary, this study revealed the mechanisms by which MALAT1 worked as a putative atherosclerosis suppressor via miR-155 and SOCS1. Therefore, modulation of MALAT1/miR-155/SOCS1 axis might alleviate the inflammation persisted in atherosclerosis.
Collapse
Affiliation(s)
- S Li
- Department of Geriatrics, Third Hospital of Changsha, Changsha 410015, PR China; Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China
| | - Y Sun
- Department of Geriatrics, Third Hospital of Changsha, Changsha 410015, PR China
| | - L Zhong
- Department of Geriatrics, Third Hospital of Changsha, Changsha 410015, PR China
| | - Z Xiao
- Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China
| | - M Yang
- Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China
| | - M Chen
- Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China
| | - C Wang
- Department of Geriatrics, Third Hospital of Changsha, Changsha 410015, PR China
| | - X Xie
- Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China
| | - X Chen
- Department of Cardiovascular, Xiangya Hospital, Central South University, Changsha 410015, PR China.
| |
Collapse
|
260
|
Garrido-Mesa J, Rodríguez-Nogales A, Algieri F, Vezza T, Hidalgo-Garcia L, Garrido-Barros M, Utrilla MP, Garcia F, Chueca N, Rodriguez-Cabezas ME, Garrido-Mesa N, Gálvez J. Immunomodulatory tetracyclines shape the intestinal inflammatory response inducing mucosal healing and resolution. Br J Pharmacol 2018; 175:4353-4370. [PMID: 30184260 DOI: 10.1111/bph.14494] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 08/12/2018] [Accepted: 08/15/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Immunomodulatory tetracyclines are well-characterized drugs with a pharmacological potential beyond their antibiotic properties. Specifically, minocycline and doxycycline have shown beneficial effects in experimental colitis, although pro-inflammatory actions have also been described in macrophages. Therefore, we aimed to characterize the mechanism behind their effect in acute intestinal inflammation. EXPERIMENTAL APPROACH A comparative pharmacological study was initially used to elucidate the most relevant actions of immunomodulatory tetracyclines: doxycycline, minocycline and tigecycline; other antibiotic or immunomodulatory drugs were assessed in bone marrow-derived macrophages and in dextran sodium sulfate (DSS)-induced mouse colitis, where different barrier markers, inflammatory mediators, microRNAs, TLRs, and the gut microbiota composition were evaluated. The sequential immune events that mediate the intestinal anti-inflammatory effect of minocycline in DSS-colitis were then characterized. KEY RESULTS Novel immunomodulatory activity of tetracyclines was identifed; they potentiated the innate immune response and enhanced resolution of inflammation. This is also the first report describing the intestinal anti-inflammatory effect of tigecycline. A minor therapeutic benefit seems to derive from their antibiotic properties. Conversely, immunomodulatory tetracyclines potentiated macrophage cytokine release in vitro, and while improving mucosal recovery in colitic mice, they up-regulated Ccl2, miR-142, miR-375 and Tlr4. In particular, minocycline initially enhanced IL-1β, IL-6, IL-22, GM-CSF and IL-4 colonic production and monocyte recruitment to the intestine, subsequently increasing Ly6C- MHCII+ macrophages, Tregs and type 2 intestinal immune responses. CONCLUSIONS AND IMPLICATIONS Immunomodulatory tetracyclines potentiate protective immune pathways leading to mucosal healing and resolution, representing a promising drug reposition strategy for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- J Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - A Rodríguez-Nogales
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - F Algieri
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - T Vezza
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - L Hidalgo-Garcia
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - M Garrido-Barros
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - M P Utrilla
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - F Garcia
- Clinical Microbiology Service, Hospital Universitario San Cecilio, ibs. GRANADA, Red de, Investigación en SIDA, Granada, Spain
| | - N Chueca
- Clinical Microbiology Service, Hospital Universitario San Cecilio, ibs. GRANADA, Red de, Investigación en SIDA, Granada, Spain
| | - M E Rodriguez-Cabezas
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - N Garrido-Mesa
- School of Health, Sport and Bioscience, University of East London, London, UK
| | - J Gálvez
- CIBER-EHD, Department of Pharmacology, ibs. GRANADA, Center for Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
261
|
O'Rourke M, Trenkmann M, Connolly M, Fearon U, Murphy CC. Novel gene targets for miRNA146a and miRNA155 in anterior uveitis. Br J Ophthalmol 2018; 103:279-285. [PMID: 30297336 DOI: 10.1136/bjophthalmol-2018-312885] [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: 07/13/2018] [Revised: 08/30/2018] [Accepted: 09/15/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND/AIMS Anterior uveitis (AU) is the most common form of intraocular inflammation. MicroRNAs (miRNA) are small, non-coding RNAs functioning as post-transcriptional repressors of gene expression. Knowledge of miRNAs can implicate specific genes and pathogenic signalling pathways in disease. This study examines miRNA expression, function and target genes in AU pathogenesis. METHODS AU and healthy control (HC) peripheral blood mononuclear cells (PBMC) were initially screened for expression of five miRNAs by real-time PCR. Regulation of the aberrantly expressed miRNAs by TLR1/2, TLR3, TLR4, IL1β and TNFα was quantified by real-time PCR and paired cytokine outputs measured by ELISA. Functional effects of miRNA overexpression using transfected THP1 cells examined IL6, IL8, IL10 and IL1β cytokine outputs by ELISA. Target genes were identified using TargetScan online computational algorithm and relevant targets verified by cloning of the 3'UTR and luciferase reporter gene assays. RESULTS Increased expression of miRNA146a (p<0.01), miRNA155 (p<0.05) and miRNA125a5p (p<0.01) was demonstrated in AU PBMC compared with HC. miRNA155 was increased following TLR1/2 (p<0.05) and TLR4 (p<0.05) stimulation and miRNA146a increased in response to IL1β (p<0.05). In a proinflammatory environment, miRNA155 overexpression in THP1 cells yielded increased cytokine output whereas miRNA146a overexpression showed decreased cytokine output. CD80, PRKCE and VASN were confirmed as novel targets for miRNA146a and SMAD2, TYRP1 and FBXO22 for miRNA155. CONCLUSION This study identifies overexpression of proinflammatory miRNA155, regulatory miRNA146a and miRNA125a-5p in AU. CD80, PRKCE and VASN are novel miRNA146a targets and SMAD2, TYRP1 and FBXO22 are novel targets for miRNA155.
Collapse
Affiliation(s)
- Micheal O'Rourke
- Department of Ophthalmology, Royal College of Surgeons in Ireland, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
| | - Michelle Trenkmann
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, University College, Dublin, Ireland
| | - Mary Connolly
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, University College, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Conor C Murphy
- Department of Ophthalmology, Royal College of Surgeons in Ireland, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
| |
Collapse
|
262
|
Surbhi, Borniger JC, Russart KLG, Zhang N, Magalang UJ, Nelson RJ. miR-155 deletion modulates lipopolysaccharide-induced sleep in female mice. Chronobiol Int 2018; 36:188-202. [PMID: 30299169 DOI: 10.1080/07420528.2018.1525617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immune signaling is known to regulate sleep. miR-155 is a microRNA that regulates immune responses. We hypothesized that miR-155 would alter sleep regulation. Thus, we investigated the potential effects of miR-155 deletion on sleep-wake behavior in adult female homozygous miR-155 knockout (miR-155KO) mice and littermate controls (WT). Mice were implanted with biotelemetry units and EEG/EMG biopotentials were recorded continuously for three baseline days. miR-155KO mice had decreased bouts of NREM and REM sleep compared with WT mice, but no differences were observed in the length of sleep bouts or total time spent in sleep-wake states. Locomotor activity and subcutaneous temperature did not differ between WT and miR-155KO mice. Following baseline recordings, mice were sleep-deprived during the first six hours of the rest phase (light phase; ZT 0-6) followed by an 18 h recovery period. There were no differences between groups in sleep rebound (% sleep and NREM δ power) after sleep deprivation. Following recovery from sleep deprivation, mice were challenged with a somnogen (viz., lipopolysaccharide (LPS)) one hour prior to the initiation of the dark (active) phase. Biopotentials were continuously recorded for the following 24 h, and miR-155KO mice displayed increased wakefulness and decreased NREM sleep during the dark phase following LPS injection. Additionally, miR-155KO mice had reduced EEG slow-wave responses (0.5-4 Hz) compared to WT mice. Together, our findings indicate that miR-155 deletion attenuates the somnogenic and EEG delta-enhancing effects of LPS. Abbreviations: ANOVA: analysis of variance; EEG: electroencephalogram; EMG: electromyogram; h: hour; IL-1: interleukin-1; IL-6: interleukin-6; IP: intra-peritoneal; LPS: lipopolysaccharide; miR/miRNA: microRNA; miR-155KO: miR-155 knockout; NREM: non-rapid eye movement; REM: rapid eye movement; TNF: tumor necrosis factor; SWS: slow-wave sleep; WT: wild-type.
Collapse
Affiliation(s)
- Surbhi
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA
| | - Jeremy C Borniger
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA
| | - Kathryn L G Russart
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA
| | - Ning Zhang
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA
| | - Ulysses J Magalang
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA.,b Department of Medicine , The Ohio State University Wexner Medical Center , Columbus , OH USA
| | - Randy J Nelson
- a Department of Neuroscience , The Ohio State University Wexner Medical Center , Columbus , OH USA
| |
Collapse
|
263
|
Awad H, Bratasz A, Nuovo G, Burry R, Meng X, Kelani H, Brown M, Ramadan ME, Williams J, Bouhliqah L, Popovich PG, Guan Z, Mcallister C, Corcoran SE, Kaspar B, Michele Basso D, Otero JJ, Kirsch C, Davis IC, Croce CM, Michaille JJ, Tili E. MiR-155 deletion reduces ischemia-induced paralysis in an aortic aneurysm repair mouse model: Utility of immunohistochemistry and histopathology in understanding etiology of spinal cord paralysis. Ann Diagn Pathol 2018; 36:12-20. [PMID: 29966831 PMCID: PMC6208131 DOI: 10.1016/j.anndiagpath.2018.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022]
Abstract
Spinal cord paralysis is relatively common after surgical repair of thoraco-abdominal aortic aneurysm (TAAA) and its etiology is unknown. The present study was designed to examine the histopathology of the disease and investigate whether miR-155 ablation would reduce spinal cord ischemic damage and delayed hindlimb paralysis induced by aortic cross-clamping (ACC) in our mouse model. The loss of locomotor function in ACC-paralyzed mice correlated with the presence of extensive gray matter damage and central cord edema, with minimal white matter histopathology. qRTPCR and Western blotting showed that the spinal cords of wild-type ACC mice that escaped paralysis showed lower miR-155 expression and higher levels of transcripts encoding Mfsd2a, which is implicated in the maintenance of blood-brain barrier integrity. In situ based testing demonstrated that increased miR-155 detection in neurons was highly correlated with the gray matter damage and the loss of one of its targets, Mfsd2a, could serve as a good biomarker of the endothelial cell damage. In vitro, we demonstrated that miR-155 targeted Mfsd2a in endothelial cells and motoneurons and increased endothelial cell permeability. Finally, miR-155 ablation slowed the progression of central cord edema, and reduced the incidence of paralysis by 40%. In sum, the surgical pathology findings clearly indicated that the epicenter of the ischemic-induced paralysis was the gray matter and that endothelial cell damage correlated to Mfsd2a loss is a good biomarker of the disease. MiR-155 targeting therefore offers new therapeutic opportunity for edema caused by traumatic spinal cord injury and diagnostic pathologists, by using immunohistochemistry, can clarify if this mechanism also is important in other ischemic diseases of the CNS, including stroke.
Collapse
Affiliation(s)
- Hamdy Awad
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Bratasz
- Small Animal Imaging Center Shared Resource, Wexner Medical Center, OSU, USA
| | - Gerard Nuovo
- Present address: Phylogeny, Inc., Powell, OH 43065-7295, USA.
| | - Richard Burry
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaomei Meng
- Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Hesham Kelani
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Melissa Brown
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Mohamed E Ramadan
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jim Williams
- Present address: Phylogeny, Inc., Powell, OH 43065-7295, USA
| | - Lamia Bouhliqah
- Department of ENT, Wexner Medical Center, OSU, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Zhen Guan
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Cynthia Mcallister
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Sarah E Corcoran
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Brian Kaspar
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - D Michele Basso
- School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - José J Otero
- Department of Pathology, Wexner Medical Center, OSU, Columbus, OH 43210, USA
| | - Claudia Kirsch
- Department of Radiology, NSUH, 300 Community Drive, Manhasset, NY 11030, USA
| | - Ian C Davis
- Department of Veterinary Biosciences, College of Veterinary Medicine, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Carlo Maria Croce
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Jean-Jacques Michaille
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA; BioPerox-IL, UB-INSERM IFR #100, Université de Bourgogne-Franche Comté, Faculté Gabriel, 6 Bd. Gabriel, 21000 Dijon, France
| | - Esmerina Tili
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA
| |
Collapse
|
264
|
Liu D, Cao S, Zhou Y, Xiong Y. Recent advances in endotoxin tolerance. J Cell Biochem 2018; 120:56-70. [PMID: 30246452 DOI: 10.1002/jcb.27547] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
Endotoxin tolerance is defined as a reduced capacity of a cell to respond endotoxin (lipopolysaccharide, LPS) challenge after an initial encounter with endotoxin in advance. The body becomes tolerant to subsequent challenge with a lethal dose of endotoxin and cytokines release and cell/tissue damage induced by inflammatory reaction are significantly reduced in the state of endotoxin tolerance. The main characteristics of endotoxin tolerance are downregulation of inflammatory mediators such as tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and C-X-C motif chemokine 10 (CXCL10) and upregulation of anti-inflammatory cytokines such as IL-10 and transforming growth factor β (TGF-β). Therefore, endotoxin tolerance is often regarded as the regulatory mechanism of the host against excessive inflammation. Endotoxin tolerance is a complex pathophysiological process and involved in multiple cellular signal pathways, receptor alterations, and biological molecules. However, the exact mechanism remains elusive up to date. To better understand the underlying cellular and molecular mechanisms of endotoxin tolerance, it is crucial to investigate the comprehensive cellular signal pathways, signaling proteins, cell surface molecules, proinflammatory and anti-inflammatory cytokines, and other mediators. Endotoxin tolerance plays an important role in reducing the mortality of sepsis, endotoxin shock, and other endotoxin-related diseases. Recent reports indicated that endotoxin tolerance is also related to other diseases such as cystic fibrosis, acute coronary syndrome, liver ischemia-reperfusion injury, and cancer. The aim of this review is to discuss the recent advances in endotoxin tolerance mainly based on the cellular and molecular mechanisms by outline the current state of the knowledge of the involvement of the toll-like receptor 4 (TLR4) signaling pathways, negative regulate factor, microRNAs, apoptosis, chromatin modification, and gene reprogramming of immune cells in endotoxin tolerance. We hope to provide a new idea and scientific basis for the rational treatment of endotoxin-related diseases such as endotoxemia, sepsis, and endotoxin shock clinically.
Collapse
Affiliation(s)
- Dan Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yejiang Zhou
- Gastrointestinal Surgery, Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Yuxia Xiong
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
265
|
Ma B, Yang Y, Li Z, Zhao D, Zhang W, Jiang Y, Xue D. Modular bioinformatics analysis demonstrates that a Toll‑like receptor signaling pathway is involved in the regulation of macrophage polarization. Mol Med Rep 2018; 18:4313-4320. [PMID: 30221738 PMCID: PMC6172368 DOI: 10.3892/mmr.2018.9486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022] Open
Abstract
In recent years, an increasing number of studies on the roles of macrophages in tumors, immune responses and metabolism have been published, in which macrophage polarization has been an extensively discussed topic. In the present study, differentially expressed genes in various types of macrophages were analyzed using the Gene Expression Omnibus database. Cluster analysis of differentially expressed genes was conducted, and a protein-protein interaction (PPI) network was constructed. Finally, modular analysis and functional enrichment analysis revealed that a Toll-like receptor (TLR) signaling pathway is involved in the regulation of macrophage polarization. Furthermore, the high-degree proteins in the PPI network that are involved in the molecular regulation of macrophage polarization are closely associated with proteins of the TLR signaling pathway. These results suggested that the TLR signaling pathways may be a principal direction of future research on the regulation of macrophage polarization.
Collapse
Affiliation(s)
- Biao Ma
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhituo Li
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dali Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Weihui Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yanfeng Jiang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
266
|
Giri BR, Mahato RI, Cheng G. Roles of microRNAs in T cell immunity: Implications for strategy development against infectious diseases. Med Res Rev 2018; 39:706-732. [PMID: 30272819 DOI: 10.1002/med.21539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 12/19/2022]
Abstract
T cell immunity plays a vital role in pathogen infections. MicroRNA (miRNAs) are small, single-stranded noncoding RNAs that regulate T cell immunity by targeting key transcriptional factors, signaling proteins, and cytokines associated with T cell activation, differentiation, and function. The dysregulation of miRNA expression in T cells may lead to specific immune responses and can provide new therapeutic opportunities against various infectious diseases. Here, we summarize recent studies that focus on the roles of miRNAs in T cell immunity and highlight miRNA functions in prevalent infectious diseases. Additionally, we also provide insights into the functions of extracellular vesicle miRNAs and attempt to delineate the mechanism of miRNA sorting into extracellular vesicles and their immunomodulatory functions. Moreover, methodologies and strategies for miRNA delivery against infectious diseases are summarized. Finally, potential strategies for miRNA-based therapies are proposed.
Collapse
Affiliation(s)
- Bikash R Giri
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Guofeng Cheng
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
267
|
Kollarova J, Cenk E, Schmutz C, Marko D. The mycotoxin alternariol suppresses lipopolysaccharide-induced inflammation in THP-1 derived macrophages targeting the NF-κB signalling pathway. Arch Toxicol 2018; 92:3347-3358. [PMID: 30175388 PMCID: PMC6208963 DOI: 10.1007/s00204-018-2299-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
Alternariol (AOH) is a secondary metabolite formed by black mold of the genus Alternaria alternata. Due to limited hazard and occurrence data, AOH is still considered as an “emerging mycotoxin” and, as such, not monitored and regulated yet. Recent studies indicate immunosuppressive effects in vitro by altering the expression of CD molecules and proinflammatory cytokines, which are indispensable in mounting an innate immune response. However, the mode of action by which AOH exerts its immunosuppressive effects has not been unraveled yet. The present study aimed to characterise the impact of AOH on the nuclear factor kappa B (NF-κB) pathway, the expression of NF-κB target cytokines and involved regulatory microRNAs (miRNAs). In THP-1 derived macrophages, AOH (1–20 µM) was found to suppress lipopolysaccharide (LPS)-induced NF-κB pathway activation, decrease secretion of the proinflammatory cytokines IL-8, IL-6, TNF-α and to induce secretion of the anti-inflammatory IL-10. Thereby, a distinct pattern of cytokine mRNA levels was monitored, varying between short- and long-term exposure. Concomitantly, AOH (2–20 µM) affected the transcription levels of miR-146a and miR-155 in LPS-stimulated THP-1 derived macrophages dose-dependently by down- and upregulation, respectively. In contrast, transcription of miR-16 and miR-125b, two other immune-related miRNAs, was not modulated. In the absence of a LPS stimulus, AOH (20 µM) did not affect basal NF-κB activity, but increased IL-10 transcription. Collectively, our results indicate, that AOH itself does not induce a proinflammatory immune response in human macrophages; however, in an inflamed environment it possesses the ability to repress inflammation by targeting the NF-κB signalling pathway and regulatory miRNAs.
Collapse
Affiliation(s)
- Jessica Kollarova
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Waehringerstr. 38, 1090, Vienna, Austria
| | - Ebru Cenk
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Waehringerstr. 38, 1090, Vienna, Austria
| | - Cornelia Schmutz
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Waehringerstr. 38, 1090, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Waehringerstr. 38, 1090, Vienna, Austria.
| |
Collapse
|
268
|
Abstract
A rapidly growing body of evidence supports the premise that neuroinflammation plays an important role in initiating and sustaining seizures in a range of pediatric epilepsies. Clinical and experimental evidence indicate that neuroinflammation is both an outcome and a contributor to seizures. In this manner, seizures that arise from an initial insult (e.g. infection, trauma, genetic mutation) contribute to an inflammatory response that subsequently promotes recurrent seizures. This cyclical relationship between seizures and neuroinflammation has been described as a 'vicious cycle.' Studies of human tissue resected for surgical treatment of refractory epilepsy have reported activated inflammatory and immune signaling pathways, while animal models have been used to demonstrate that key inflammatory mediators lead to increased seizure susceptibility. Further characterization of the molecular mechanisms involved in this cycle may ultimately enable the development of new therapeutic approaches for the treatment of epilepsy. In this brief review we focus on key inflammatory mediators that have become prominent in recent literature of epilepsy, including newly characterized microRNAs and their potential role in neuroinflammatory signaling.
Collapse
Affiliation(s)
- Shruti Bagla
- Division of Hematology/Oncology, Department of Pediatrics, Room 3L22, Children's Hospital of Michigan, 3901 Beaubien Blvd, Detroit, MI 48201, USA
| | - Alan A Dombkowski
- Division of Clinical Pharmacology and Toxicology, Department of Pediatrics, Room 3L22, Children's Hospital of Michigan, 3901 Beaubien Blvd, Detroit, MI 48201, USA
| |
Collapse
|
269
|
Harquail J, LeBlanc N, Landry C, Crapoulet N, Robichaud GA. Pax-5 Inhibits NF-κB Activity in Breast Cancer Cells Through IKKε and miRNA-155 Effectors. J Mammary Gland Biol Neoplasia 2018; 23:177-187. [PMID: 30032344 DOI: 10.1007/s10911-018-9404-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/05/2018] [Indexed: 12/17/2022] Open
Abstract
Pax-5, an essential transcription factor in B cell development, is aberrantly expressed in various B cell cancer lesions and solid tumors such as breast carcinoma. We have recently shown that Pax-5 regulates NF-κB activity which lead to the modulation of breast cancer phenotypic features (EMT-MET). NF-κB is known as a central mediator in inflammation, stress response as well as being a gatekeeper of pro-tumorigenic activity. However, little is known as to how Pax-5 affects this modulation. We thus turned our attention to microRNAs as potential regulatory effectors. In this study, we set out to elucidate the regulatory network between differential Pax-5 expression and NF-κB activity which dictate breast cancer malignancy. Through next-generation sequencing (NGS) of breast cancer cells conditionally expressing Pax-5, we profile significantly upregulated microRNAs; including microRNA-155, a known regulator of pathological processes and suppressor of malignant growth. Through the conditional expression of microRNA-155 in breast cancer models, we identify and validate IKKε (IKBKE) as a downstream target and an essential effector of Pax-5-mediated suppression of NF-κB signaling. Using rescue experiments, we also confirm that Pax-5 modulates NF-κB activity via IKKε downregulation. Interestingly, we also show that microRNA-155, in turn, supresses Pax-5 expression, indicative of an auto-regulatory feedback loop. Altogether, we demonstrate that Pax-5 inhibits NF-κB signalling through the regulation of microRNA-155 and its downstream target IKKε. The elucidation of this signaling network is relevant as Pax-5 and NF-κB are potent transcriptional regulators of breast cancer aggressivity. In addition, IKKε is relevant oncogene aberrantly expressed in 30% of breast carcinomas. Further insight into the regulatory pathways of breast cancer progression will eventually identify strategic therapeutic and prognostic targets to improve cancer patient outcome.
Collapse
Affiliation(s)
- Jason Harquail
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, E1C 8X3, Canada
| | - Nicolas LeBlanc
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, E1C 8X3, Canada
| | - Carine Landry
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, E1C 8X3, Canada
| | - Nicolas Crapoulet
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, E1C 8X3, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1A 3E9, Canada.
- Atlantic Cancer Research Institute, Moncton, NB, E1C 8X3, Canada.
| |
Collapse
|
270
|
Hassouna SS, Tayel MY, ElKaffash DM, Abdelhady AM, Elsayed EH. MicroRNA155 Expression in Relation to BDCAF Scored Behçet's Disease in an Egyptian Patients' Sample. Open Rheumatol J 2018; 12:115-122. [PMID: 30197703 PMCID: PMC6110073 DOI: 10.2174/1874312901812010115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/19/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023] Open
Abstract
Objective: To discover the possibility of using microRNA155 (miRNA155) expression level as a biomarker of Behçet's Disease (BD) activity or remission. Methods: Thirty BD patients’ white blood cells (WBCs) miRNA155 expression was measured and compared to WBCs miRNA155 expression in 15 healthy subjects. Assessment of disease activity was done using Behçet's Disease Current Activity Form (BDCAF). Results: miRNA155 expression significantly decreases with the increase of BD activity scored by BDCAF. Conclusion: Increased miRNA155 may be used as a biomarker of BD remission and thus in the disease follow up. There could be a prospect of treating the disease via microRNA 155 effect enhancement.
Collapse
Affiliation(s)
- Sally S Hassouna
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Manal Y Tayel
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Dalal M ElKaffash
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ahmed M Abdelhady
- Department of Opthalmology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman H Elsayed
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
271
|
MiR-146a induction by cyanobacterial lipopolysaccharide antagonist (CyP) mediates endotoxin cross-tolerance. Sci Rep 2018; 8:11367. [PMID: 30054544 PMCID: PMC6063882 DOI: 10.1038/s41598-018-29820-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/17/2018] [Indexed: 01/03/2023] Open
Abstract
Endotoxin tolerance is a phenomenon characterized by a reduced capacity of monocytes and macrophages to respond to repeated stimulation with lipopolysaccharide (LPS) which has been suggested to represent a way of controlling the intensity and duration of innate immune response. During endotoxin tolerance, monocytes undergo functional re-programming primarily by epigenetic regulation. Recently, micro-RNA (miR)-146a has been demonstrated to be the major player of the negative regulation of the pro-inflammatory response, affecting TNF-α production. In this study, we have employed CyP, a cyanobacterial LPS antagonist acting on TLR4-MD2 complex, for priming human monocytes and evaluating their response to a subsequent challenge with E. coli LPS. Results show that CyP is able to induce cross-tolerance to E. coli LPS by inhibiting TNF-α production. The mechanism of action is mediated by a specific induction of miR-146a and reduction of IRAK1 and TRAF6 expressions in human monocytes by CyP priming. Up-regulation of miR-146a by CyP alone, affects subsequent cell response in term of TNF-α production even when monocytes are incubated with other TLR ligands, as lipoteichoic acid (LTA), thus confirming miR-146a as a critical player mediating TNF-α regulation during cross-tolerance with CyP.
Collapse
|
272
|
Liu B, Shyr Y, Cai J, Liu Q. Interplay between miRNAs and host genes and their role in cancer. Brief Funct Genomics 2018; 18:255-266. [PMID: 30785618 PMCID: PMC6609535 DOI: 10.1093/bfgp/elz002] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/21/2018] [Accepted: 01/23/2019] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding functional RNAs that post-transcriptionally regulate gene expression. They play essential roles in nearly all biological processes including cell development and differentiation, DNA damage repair, cell death as well as intercellular communication. They are highly involved in cancer, acting as tumor suppressors and/or promoters to modulate cell proliferation, epithelial-mesenchymal transition and tumor invasion and metastasis. Recent studies have shown that more than half of miRNAs are located within protein-coding or non-coding genes. Intragenic miRNAs and their host genes either share the promoter or have independent transcription. Meanwhile, miRNAs work as partners or antagonists of their host genes by fine-tuning their target genes functionally associated with host genes. This review outlined the complicated relationship between intragenic miRNAs and host genes. Focusing on miRNAs known as oncogenes or tumor suppressors in specific cancer types, it studied co-expression relationships between these miRNAs and host genes in the cancer types using TCGA data sets, which validated previous findings and revealed common, tumor-specific and even subtype-specific patterns. These observations will help understand the function of intragenic miRNAs and further develop miRNA therapeutics in cancer.
Collapse
Affiliation(s)
- Baohong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jianping Cai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
273
|
Korotkov A, Broekaart DWM, van Scheppingen J, Anink JJ, Baayen JC, Idema S, Gorter JA, Aronica E, van Vliet EA. Increased expression of matrix metalloproteinase 3 can be attenuated by inhibition of microRNA-155 in cultured human astrocytes. J Neuroinflammation 2018; 15:211. [PMID: 30031401 PMCID: PMC6054845 DOI: 10.1186/s12974-018-1245-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Background Temporal lobe epilepsy (TLE) is a chronic neurological disease, in which about 30% of patients cannot be treated adequately with anti-epileptic drugs. Brain inflammation and remodeling of the extracellular matrix (ECM) seem to play a major role in TLE. Matrix metalloproteinases (MMPs) are proteolytic enzymes largely responsible for the remodeling of the ECM. The inhibition of MMPs has been suggested as a novel therapy for epilepsy; however, available MMP inhibitors lack specificity and cause serious side effects. We studied whether MMPs could be modulated via microRNAs (miRNAs). Several miRNAs mediate inflammatory responses in the brain, which are known to control MMP expression. The aim of this study was to investigate whether an increased expression of MMPs after interleukin-1β (IL-1β) stimulation can be attenuated by inhibition of the inflammation-associated miR-155. Methods We investigated the expression of MMP2, MMP3, MMP9, and MMP14 in cultured human fetal astrocytes after stimulation with the pro-inflammatory cytokine IL-1β. The cells were transfected with miR-155 antagomiR, and the effect on MMP3 expression was investigated using real-time quantitative PCR and Western blotting. Furthermore, we characterized MMP3 and miR-155 expression in brain tissue of TLE patients with hippocampal sclerosis (TLE-HS) and during epileptogenesis in a rat TLE model. Results Inhibition of miR-155 by the antagomiR attenuated MMP3 overexpression after IL-1β stimulation in astrocytes. Increased expression of MMP3 and miR-155 was also evident in the hippocampus of TLE-HS patients and throughout epileptogenesis in the rat TLE model. Conclusions Our experiments showed that MMP3 is dynamically regulated by seizures as shown by increased expression in TLE tissue and during different phases of epileptogenesis in the rat TLE model. MMP3 can be induced by the pro-inflammatory cytokine IL-1β and is regulated by miR-155, suggesting a possible strategy to prevent epilepsy via reduction of inflammation. Electronic supplementary material The online version of this article (10.1186/s12974-018-1245-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anatoly Korotkov
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Diede W M Broekaart
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jasper J Anink
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Johannes C Baayen
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Sander Idema
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Erwin A van Vliet
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands. .,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.
| |
Collapse
|
274
|
Li TR, Jia YJ, Wang Q, Shao XQ, Zhang P, Lv RJ. Correlation between tumor necrosis factor alpha mRNA and microRNA-155 expression in rat models and patients with temporal lobe epilepsy. Brain Res 2018; 1700:56-65. [PMID: 30006293 DOI: 10.1016/j.brainres.2018.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/03/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023]
Abstract
Accumulative evidence demonstrates that there is an inseparable connection between inflammation and temporal lobe epilepsy (TLE). Some recent studies have found that the multifunctional microRNA-155 (miR-155) is a key regulator in controlling the neuroinflammatory response of TLE rodent animals and patients. The aim of the present study was to investigate the dynamic expression pattern of tumor necrosis factor alpha (TNF-α) as a pro-inflammatory cytokine and miR-155 as a posttranscriptional inflammation-related miRNA in the hippocampus of TLE rat models and patients. We performed real-time quantitative PCR (qRT-PCR) on the rat hippocampus 2 h, 7 days, 21 days and 60 days following kainic acid-induced status epilepticus (SE) and on hippocampi obtained from TLE patients and normal controls. To further characterize the relationship between TNF-α and miR-155, we examined the effect of antagonizing miR-155 on TNF-α secretion using its antagomir. Here, we found that TNF-α secretion and miR-155 expression levels were correlated after SE. The expression of TNF-α reached peak levels in the acute phase (2h post-SE) of seizure and then gradually decreased; however, it rose again in the chronic phase (60 days post-SE). miR-155 expression started to increase 2 h post-SE, reached peak levels in the latent phase (7 days post-SE) of seizure and then gradually decreased. The variation in the trend of miR-155 lagged behind that of TNF-α. In patients with TLE, the expression levels of both TNF-α and miR-155 were also significantly increased. Furthermore, antagonizing miR-155 inhibited the production of TNF-α in the hippocampal tissues of TLE rat models. Our findings demonstrate a critical role for miR-155 in the physiological regulation of the TNF-α pro-inflammatory response and elucidate the role of neuroinflammation in the pathogenesis of TLE. Therefore, regulation of the miR-155/TNF-α axis may be a new therapeutic target for TLE.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, 88 Road of JianKang, WeiHui, Xinxiang 453100, PR China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing 100050, PR China
| | - Yan-Jie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 East Road of JianShe, Erqi District, Zhengzhou 450052, PR China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing 100050, PR China
| | - Xiao-Qiu Shao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing 100050, PR China
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, 88 Road of JianKang, WeiHui, Xinxiang 453100, PR China
| | - Rui-Juan Lv
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases, 6 TianTanXiLi, Dongcheng District, Beijing 100050, PR China.
| |
Collapse
|
275
|
Michaille JJ, Piurowski V, Rigot B, Kelani H, Fortman EC, Tili E. MiR-663, a MicroRNA Linked with Inflammation and Cancer That Is under the Influence of Resveratrol. MEDICINES 2018; 5:medicines5030074. [PMID: 29987196 PMCID: PMC6163211 DOI: 10.3390/medicines5030074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 12/25/2022]
Abstract
Resveratrol (trans-3,5,4′-trihydroxystilbene, RSV) is a non-flavonoid dietary polyphenol with antioxidant, anti-inflammatory and anti-cancer properties that is primarily found in red berries. While RSV displays many beneficial effects in vitro, its actual effects in vivo or in animal models remain passionately debated. Recent publications suggest that RSV pleiotropic effects could arise from its capability to regulate the expression and activity of microRNAs, short regulators themselves capable of regulating up to several hundreds of target genes. In particular, RSV increases microRNA miR-663 expression in different human cell lines, suggesting that at least some of its multiple beneficial properties are through the modulation of expression of this microRNA. Indeed, the expression of microRNA miR-663 is reduced in certain cancers where miR-663 is considered to act as a tumor suppressor gene, as well as in other pathologies such as cardiovascular disorders. Target of miR-663 include genes involved in tumor initiation and/or progression as well as genes involved in pathologies associated with chronic inflammation. Here, we review the direct and indirect effects of RSV on the expression of miR-663 and its target transcripts, with emphasise on TGFβ1, and their expected health benefits, and argue that elucidating the molecular effects of different classes of natural compounds on the expression of microRNAs should help to identify new therapeutic targets and design new treatments.
Collapse
Affiliation(s)
- Jean-Jacques Michaille
- BioPerox-IL, UB-INSERM IFR #100, Faculté Gabriel, Université de Bourgogne-Franche Comté, 21000 Dijon, France.
| | - Victoria Piurowski
- Department of Biology, Franklin College of Arts and Sciences, University of Georgia, Athes, GA 30602, USA.
| | - Brooke Rigot
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Hesham Kelani
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Emily C Fortman
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Esmerina Tili
- Department of Cancer Biology and Genetics, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
276
|
Liang X, Zheng S, Cui J, Yu D, Yang G, Zhou L, Wang B, Cai L, Li W. Alterations of MicroRNA Expression in the Liver, Heart, and Testis of Mice Upon Exposure to Repeated Low-Dose Radiation. Dose Response 2018; 16:1559325818799561. [PMID: 30263020 PMCID: PMC6153535 DOI: 10.1177/1559325818799561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRs), which regulate target gene expression at the post-transcriptional level, play a crucial role in inducing biological effects upon high-dose ionizing radiation. Yet, the miR expression profiles in response to repeated low-dose radiation (LDR) in vivo have not been elucidated. This study investigated the response profiles of 11 miRs with functions involved in metabolism, DNA damage and repair, inflammation, and fibrosis in mouse liver, heart, and testis upon repeated LDR exposure for 4 months. The expression profiles were evaluated using stem-loop quantitative reverse transcription polymerase chain reaction immediately and at 2 months after LDR exposure. The expression profiles varied significantly at both time points. At the organ level, the heart was the most affected, followed by the liver and testis, in which significant miR upregulation related to DNA damage response was found. Metabolism-related miRs decreased in the liver and increased in the testis. The current results showed immediate and long-lasting alterations in the miR expression profiles in response to repeated LDR in different organs.
Collapse
Affiliation(s)
- Xinyue Liang
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
- Pediatric Research Institute, Department of Pediatrics of the
University of Louisville, Louisville, KY, USA
| | - Shirong Zheng
- Pediatric Research Institute, Department of Pediatrics of the
University of Louisville, Louisville, KY, USA
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
| | - Dehai Yu
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
| | - Guozi Yang
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
| | - Brain Wang
- Department of Radiation Oncology, The University of Louisville,
Louisville, KY, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics of the
University of Louisville, Louisville, KY, USA
- Department of Radiation Oncology, The University of Louisville,
Louisville, KY, USA
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun,
China
| |
Collapse
|
277
|
Wang W, Bian H, Li F, Li X, Zhang D, Sun S, Song S, Zhu Q, Ren W, Qin C, Qi J. HBeAg induces the expression of macrophage miR-155 to accelerate liver injury via promoting production of inflammatory cytokines. Cell Mol Life Sci 2018; 75:2627-2641. [PMID: 29349567 PMCID: PMC11105519 DOI: 10.1007/s00018-018-2753-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/11/2018] [Accepted: 01/16/2018] [Indexed: 12/14/2022]
Abstract
Activation of Kupffer cells (KCs) induced that inflammatory cytokine production plays a central role in the pathogenesis of HBV infection. The previous studies from our and other laboratory demonstrated miRNAs can regulate TLR-inducing inflammatory responses to macrophage. However, the involvement of miRNAs in HBV-associated antigen-induced macrophage activation is still not thoroughly understood. Here, we evaluated the effects and mechanisms of miR-155 in HBV-associated antigen-induced macrophage activation. First, co-culture assay of HepG2 or HepG2.2.15 cells and RAW264.7 macrophages showed that HepG2.2.15 cells could significantly promote macrophages to produce inflammatory cytokines. Furthermore, we, respectively, stimulated RAW264.7 macrophages, mouse primary peritoneal macrophages, or healthy human peripheral blood monocytes with HBV-associated antigens, including HBcAg, HBeAg, and HBsAg, and found that only HBeAg could steadily enhance the production of inflammatory cytokines in these cells. Subsequently, miRNAs sequencing presented the up- or down-regulated expression of multiple miRNAs in HBeAg-stimulated RAW264.7 cells. In addition, we verified the expression of miR-155 and its precursors BIC gene with q-PCR in the system of co-culture or HBeAg-stimulated macrophages. Meanwhile, the increased miR-155 expression was positively correlation with serum ALT, AST, and HBeAg levels in AHB patients. Although MAPK, PI3K, and NF-κB signal pathways were all activated during HBeAg treatment, only PI3K and NF-κB pathways were involved in miR-155 expression induced by HBeAg stimulation. Consistently, miR-155 over-expression inhibited production of inflammatory cytokines, which could be reversed by knocking down miR-155. Moreover, we demonstrated that miR-155 regulated HBeAg-induced cytokine production by targeting BCL-6, SHIP-1, and SOCS-1. In conclusion, our data revealed that HBeAg augments the expression of miR-155 in macrophages via PI3K and NF-κB signal pathway and the increased miR-155 promotes HBeAg-induced inflammatory cytokine production by inhibiting the expression of BCL-6, SHIP-1, and SOCS-1.
Collapse
Affiliation(s)
- Wenwen Wang
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Hongjun Bian
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Feifei Li
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Xiao Li
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Di Zhang
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Shuohuan Sun
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Shouyang Song
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Qiang Zhu
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Wanhua Ren
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Chengyong Qin
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China
| | - Jianni Qi
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China.
- Shandong Provincial Engineering and Technological Research Center for Liver Diseases Prevention and Control, Jinan, 250021, Shandong, China.
| |
Collapse
|
278
|
Yang Y, Alderman C, Sehlaoui A, Xiao Y, Wang W. MicroRNAs as Immunotherapy Targets for Treating Gastroenterological Cancers. Can J Gastroenterol Hepatol 2018; 2018:9740357. [PMID: 30046565 PMCID: PMC6038585 DOI: 10.1155/2018/9740357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/02/2018] [Indexed: 01/17/2023] Open
Abstract
Gastroenterological cancers are the most common cancers categorized by systems and are estimated to comprise 18.4% of all cancers in the United States in 2017. Gastroenterological cancers are estimated to contribute 26.2% of cancer-related death in 2017. Gastroenterological cancers are characterized by late diagnosis, metastasis, high recurrence, and being refractory to current therapies. Since the current targeted therapies provide limited benefit to the overall response and survival, there is an urgent need for developing novel therapeutic strategy to improve the outcome of gastroenterological cancers. Immunotherapy has been developed and underwent clinical trials, but displayed limited therapeutic benefit. Since aberrant expressions of miRNAs are found in gastroenterological cancers and miRNAs have been shown to regulate antitumor immunity, the combination therapy combining the traditional antibody-based immunotherapy and novel miRNA-based immunotherapy is promising for achieving clinical success. This review summarizes the current knowledge about the miRNAs and long noncoding RNAs that exhibit immunoregulatory roles in gastroenterological cancers and precancerous diseases of digestive system, as well as the miRNA-based clinical trials for gastroenterological cancers. This review also analyzes the ongoing challenge of identifying appropriate therapy candidates for complex and dynamic tumor microenvironment, ensuring efficient and targeted delivery to specific cancer tissues, and developing strategy for avoiding off-target effect.
Collapse
Affiliation(s)
- Yixin Yang
- College of Natural, Applied and Health Sciences, Kean University, 100 Morris Avenue, Union, NJ 07083, USA
| | - Christopher Alderman
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Ayoub Sehlaoui
- Department of Biological Sciences, Emporia State University, 1 Kellogg Circle, Emporia, KS 66801, USA
| | - Yuan Xiao
- Department of Biological Sciences, Emporia State University, 1 Kellogg Circle, Emporia, KS 66801, USA
| | - Wei Wang
- Department of Thoracic Surgery III, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
| |
Collapse
|
279
|
Tahamtan A, Teymoori-Rad M, Nakstad B, Salimi V. Anti-Inflammatory MicroRNAs and Their Potential for Inflammatory Diseases Treatment. Front Immunol 2018; 9:1377. [PMID: 29988529 PMCID: PMC6026627 DOI: 10.3389/fimmu.2018.01377] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/04/2018] [Indexed: 12/27/2022] Open
Abstract
Inflammation is a complicated biological and pathophysiological cascade of responses to infections and injuries, and inflammatory mechanisms are closely related to many diseases. The magnitude, the complicated network of pro- and anti-inflammatory factors, and the direction of the inflammatory response can impact on the development and progression of various disorders. The currently available treatment strategies often target the symptoms and not the causes of inflammatory disease and may often be ineffective. Since the onset and termination of inflammation are crucial to prevent tissue damage, a range of mechanisms has evolved in nature to regulate the process including negative and positive feedback loops. In this regard, microRNAs (miRNAs) have emerged as key gene regulators to control inflammation, and it is speculated that they are fine-tune signaling regulators to allow for proper resolution and prevent uncontrolled progress of inflammatory reactions. In this review, we discuss recent findings related to significant roles of miRNAs in immune regulation, especially the potential utility of these molecules as novel anti-inflammatory agents to treat inflammatory diseases. Furthermore, we discuss the possibilities of using miRNAs as drugs in the form of miRNA mimics or miRNA antagonists.
Collapse
Affiliation(s)
- Alireza Tahamtan
- Infectious Diseases Research Centre, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Britt Nakstad
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
280
|
Peltier D, Reddy P. Non-Coding RNA Mediated Regulation of Allogeneic T Cell Responses After Hematopoietic Transplantation. Front Immunol 2018; 9:1110. [PMID: 29963039 PMCID: PMC6013767 DOI: 10.3389/fimmu.2018.01110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/03/2018] [Indexed: 12/21/2022] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is an effective therapy for several malignant and non-malignant disorders. The precise control of allogeneic T cells is critical for successful outcomes after BMT. The mechanisms governing desirable (graft-versus-leukemia) versus undesirable (graft-versus-host disease) allogeneic responses remain incompletely understood. Non-coding RNAs (ncRNA) are controllers of gene expression that fine-tune cellular responses. Multiple microRNAs (miRNAs), a type of ncRNA, have recently been shown to influence allogeneic T cell responses in both murine models and clinically. Here, we review the role of various miRNAs that regulate T cell responses, either positively or negatively, to allo-stimulation and highlight their potential relevance as biomarkers and as therapeutic targets for improving outcomes after allogeneic BMT.
Collapse
Affiliation(s)
- Daniel Peltier
- Division of Hematology and Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
281
|
Wang D, Tang M, Zong P, Liu H, Zhang T, Liu Y, Zhao Y. MiRNA-155 Regulates the Th17/Treg Ratio by Targeting SOCS1 in Severe Acute Pancreatitis. Front Physiol 2018; 9:686. [PMID: 29937734 PMCID: PMC6002743 DOI: 10.3389/fphys.2018.00686] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/17/2018] [Indexed: 12/11/2022] Open
Abstract
Acute pancreatitis (AP) is a serious condition associated with intestinal barrier disruption or inflammation of the pancreatic tissue. Specific microRNAs are involved in the pathogenesis of AP, during which IL-17-producing CD4+ T helper (Th17) cells accumulate in the pancreas. In this study, significantly increased levels of miR-155 were detected in clinical samples from patients with AP, and overexpression of miR-155 correlated with severe AP (SAP). To identify the effect of miR-155 on T cell differentiation, we isolated CD4+ T lymphocytes and in vitro experiments showed that inhibition of miR-155 significantly reversed the stress-induced increase in the Th17/Treg ratio. The results also showed that miR-155 increased the Th17-mediated inflammatory response by targeting SOCS1. The interaction between miR-155 and the 3′-UTR of SOCS1 was confirmed by a dual luciferase reporter assay and RT-PCR. Experimental AP of varying severity was induced in BALB/c mice by caerulein hyperstimulation and miR-155 expression was found to increase with disease progression. Inhibition of miR-155 expression significantly improved the pathology of the pancreas. We also observed downregulation of expression of inflammatory factors, IL-17, SOCS1 and phosphorylated STAT1 after miR-155 inhibition. In summary, miR-155 regulates the Th17/Treg ratio by targeting SOCS1, most probably via direct binding to its 3′-UTR region, indicating that this microRNA may be a potential biomarker and/or therapeutic target for AP.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Maochun Tang
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Pengfei Zong
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Hua Liu
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Ting Zhang
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yu Liu
- The Community Health Service Center of Nanxiang Town, Shanghai, China
| | - Yan Zhao
- Department of Gastroenterology, Tenth People's Hospital of Tongji University, Shanghai, China
| |
Collapse
|
282
|
The Kat in the HAT: The Histone Acetyl Transferase Kat6b (MYST4) Is Downregulated in Murine Macrophages in Response to LPS. Mediators Inflamm 2018; 2018:7852742. [PMID: 29977151 PMCID: PMC6011073 DOI: 10.1155/2018/7852742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
Epigenetic modulators, including histone methylases, demethylases, and deacetylases, have been implicated previously in the regulation of classical and alternative macrophage activation pathways. In this study, we show that the histone acetyl transferase (HAT) Kat6B (MYST4) is strongly suppressed (>80%) in macrophages by lipopolysaccharide (LPS) (M1 activation), while Kat6A, its partner in the MOZ/MORF complex, is reciprocally upregulated. This pattern of expression is not altered by LPS together with the adenosine receptor agonist NECA (M2d activation). This is despite the observation that miR-487b, a putative regulator of Kat6B expression, is mildly stimulated by LPS, but strongly suppressed by LPS/NECA. Other members of the MYST family of HATs (Kat5, Kat7, and Kat8) are unaffected by LPS treatment. Using the pLightswitch 3′UTR reporter plasmid, the miR-487b binding site in the Kat6b 3′UTR was found to play a role in the LPS-mediated suppression of Kat6B expression, but other as-yet unidentified factors are also involved. As Kat6B is a HAT that has the potential to modulate gene expression by its effects on chromatin accessibility, we are continuing our studies into the potential roles of this epigenetic modulator in macrophage activation pathways.
Collapse
|
283
|
Wang X, Ye X, Ji J, Wang J, Xu B, Zhang Q, Ming J, Liu X. MicroRNA‑155 targets myosin light chain kinase to inhibit the migration of human bone marrow‑derived mesenchymal stem cells. Int J Mol Med 2018; 42:1585-1592. [PMID: 29901087 DOI: 10.3892/ijmm.2018.3718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 05/31/2018] [Indexed: 11/06/2022] Open
Abstract
Toll‑like receptors (TLRs) are expressed in human bone marrow‑derived mesenchymal stromal cells (BM‑MSCs). The activation of TLRs is important in the proliferation, differ-entiation, migration and hematopoiesis‑supporting functions of BM‑MSCs. MicroRNAs (miRNAs) are involved in various biological functions by mediating mRNA degradation or inhibiting the translation of target genes. Our previous study confirmed that TLRs regulate the migration ability of BM‑MSCs. It was also identified that multiple miRNAs were regulated by TLRs. In view of this, it was hypothesized that TLR‑regulated miRNAs may be important in regulating the migration of BM‑MSCs. The migration ability of BM‑MSCs was evaluated following transfection of the cells with the mimics or antagonists of miRNA (miR)‑27b, miR‑146a, miR‑155 and miR‑154. miR‑155 significantly inhibited cell migration. Myosin light chain kinase (MYLK) was identified as the direct target of miR‑155 in BM‑MSCs, which was further investigated using the luciferase reporter assay. However, miR‑155 did not affect the expression of upstream proteins of the RhoA pathway controlling the activity of MYLK, suggesting that miR‑155 directly suppressed the expression of MYLK without affecting the RhoA pathway. These results may facilitate the development and clinical use of BM‑MSCs in terms of their migration.
Collapse
Affiliation(s)
- Xingbing Wang
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Xu Ye
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Jingjuan Ji
- Reproductive Medicine Center, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Jian Wang
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Bo Xu
- Reproductive Medicine Center, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Qian Zhang
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Jing Ming
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Xin Liu
- Department of Hematology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
284
|
Buschmann D, Kirchner B, Hermann S, Märte M, Wurmser C, Brandes F, Kotschote S, Bonin M, Steinlein OK, Pfaffl MW, Schelling G, Reithmair M. Evaluation of serum extracellular vesicle isolation methods for profiling miRNAs by next-generation sequencing. J Extracell Vesicles 2018; 7:1481321. [PMID: 29887978 PMCID: PMC5990937 DOI: 10.1080/20013078.2018.1481321] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/18/2018] [Indexed: 01/08/2023] Open
Abstract
Extracellular vesicles (EVs) are intercellular communicators with key functions in physiological and pathological processes and have recently garnered interest because of their diagnostic and therapeutic potential. The past decade has brought about the development and commercialization of a wide array of methods to isolate EVs from serum. Which subpopulations of EVs are captured strongly depends on the isolation method, which in turn determines how suitable resulting samples are for various downstream applications. To help clinicians and scientists choose the most appropriate approach for their experiments, isolation methods need to be comparatively characterized. Few attempts have been made to comprehensively analyse vesicular microRNAs (miRNAs) in patient biofluids for biomarker studies. To address this discrepancy, we set out to benchmark the performance of several isolation principles for serum EVs in healthy individuals and critically ill patients. Here, we compared five different methods of EV isolation in combination with two RNA extraction methods regarding their suitability for biomarker discovery-focused miRNA sequencing as well as biological characteristics of captured vesicles. Our findings reveal striking method-specific differences in both the properties of isolated vesicles and the ability of associated miRNAs to serve in biomarker research. While isolation by precipitation and membrane affinity was highly suitable for miRNA-based biomarker discovery, methods based on size-exclusion chromatography failed to separate patients from healthy volunteers. Isolated vesicles differed in size, quantity, purity and composition, indicating that each method captured distinctive populations of EVs as well as additional contaminants. Even though the focus of this work was on transcriptomic profiling of EV-miRNAs, our insights also apply to additional areas of research. We provide guidance for navigating the multitude of EV isolation methods available today and help researchers and clinicians make an informed choice about which strategy to use for experiments involving critically ill patients.
Collapse
Affiliation(s)
- Dominik Buschmann
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany.,Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,Dr. von Hauner Children's Hospital, LMU Munich, Munich, Germany
| | - Stefanie Hermann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Melanie Märte
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Christine Wurmser
- Chair of Animal Breeding, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Florian Brandes
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | | | - Ortrud K Steinlein
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Michael W Pfaffl
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustav Schelling
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Marlene Reithmair
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
285
|
Ottaviani L, Sansonetti M, da Costa Martins PA. Myocardial cell-to-cell communication via microRNAs. Noncoding RNA Res 2018; 3:144-153. [PMID: 30175287 PMCID: PMC6114265 DOI: 10.1016/j.ncrna.2018.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/30/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Lara Ottaviani
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Marida Sansonetti
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
286
|
Marques-Rocha JL, Garcia-Lacarte M, Samblas M, Bressan J, Martínez JA, Milagro FI. Regulatory roles of miR-155 and let-7b on the expression of inflammation-related genes in THP-1 cells: effects of fatty acids. J Physiol Biochem 2018; 74:579-589. [PMID: 29790117 DOI: 10.1007/s13105-018-0629-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
Abstract
The main aim of this investigation was to study the regulatory roles of let-7b and miR-155-3p on the expression of inflammation-associated genes in monocytes, macrophages, and lipopolysaccharide (LPS)-activated macrophages (AcM). A second goal was to analyze the potential modulatory roles of different fatty acids, including oleic, palmitic, eicosapentaenoic (EPA), and docosahexaenoic (DHA), on the expression of these miRNAs in the three cell types. This hypothesis was tested in human acute monocytic leukemia cells (THP-1), which were differentiated into macrophages with 2-O-tetradecanoylphorbol-13-acetate (TPA) and further activated with LPS for 24 h. Monocytes, macrophages, and AcM were transfected with a negative control, or mimics for miR-155-3p and miR-let-7b-5p. The expression of both miRNAs and some proinflammatory genes was analyzed by qRT-PCR. Interestingly, let-7b mimic reduced the expression of IL6 and TNF in monocytes, and SERPINE1 expression in LPS-activated macrophages. However, IL6, TNF, and SERPINE1 were upregulated in macrophages by let-7b mimic. IL6 expression was higher in the three types of cells after transfecting with miR-155-3p mimic. Similarly, expression of SERPINE1 was increased by miR-155-3p mimic in monocytes and macrophages. However, TLR4 was downregulated by miR-155-3p in monocytes and macrophages. Regarding the effects of the different fatty acids, oleic acid increased the expression of let-7b in macrophages and AcM and also increased the expression of miR-155 in monocytes when compared with DHA but not when compared with non-treated cells. Overall, these results suggest anti- and proinflammatory roles of let-7b and miR-155-3p in THP-1 cells, respectively, although these outcomes are strongly dependent on the cell type. Noteworthy, oleic acid might exert beneficial anti-inflammatory effects in immune cells (i.e., non-activated and LPS-activated macrophages) by upregulating the expression of let-7b.
Collapse
Affiliation(s)
- J L Marques-Rocha
- Department of Integrated Education of Health, Federal University of Espirito Santo, Vitória, Brazil
| | - M Garcia-Lacarte
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain
| | - M Samblas
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain
| | - J Bressan
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Brazil
| | - J A Martínez
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,IMDEA Food, Madrid, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain. .,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
287
|
Nejad C, Stunden HJ, Gantier MP. A guide to miRNAs in inflammation and innate immune responses. FEBS J 2018; 285:3695-3716. [DOI: 10.1111/febs.14482] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/08/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Charlotte Nejad
- Centre for Innate Immunity and Infectious Diseases Hudson Institute of Medical Research Clayton Australia
- Department of Molecular and Translational Science Monash University Clayton Australia
| | - H. James Stunden
- Institute of Innate Immunity Biomedical Center University Hospitals Bonn Bonn Germany
| | - Michael P. Gantier
- Centre for Innate Immunity and Infectious Diseases Hudson Institute of Medical Research Clayton Australia
- Department of Molecular and Translational Science Monash University Clayton Australia
| |
Collapse
|
288
|
Park J, Jeong S, Park K, Yang K, Shin S. Expression profile of microRNAs following bone marrow-derived mesenchymal stem cell treatment in lipopolysaccharide-induced acute lung injury. Exp Ther Med 2018; 15:5495-5502. [PMID: 29904430 DOI: 10.3892/etm.2018.6118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/23/2018] [Indexed: 12/11/2022] Open
Abstract
Immunomodulatory or immunosuppressive properties of bone marrow-derived mesenchymal stem cells (BM-MSCs) facilitate the treatment of acute respiratory distress syndrome and acute lung injury (ALI). Dysregulated miRNA (miRNA or miR) expression associated with the effects of BM-MSCs was assessed in a rat model of lipopolysaccharide (LPS)-induced ALI. The present study performed biochemical tests to assess five analytes, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate, blood urea nitrogen (BUN), and creatinine (CREA). Total cell count was assessed and the percentage of bronchoalveolar lavage neutrophil content was also examined. The results Histopathological examination of rat upper lobe lung tissue was then used to estimate lung injury score (LIS). The levels of AST, lactate, BUN and creatinine (excluding ALT), released into the circulation upon injury, were significantly lower in ALI rats treated with BM-MSCs than in ALI rats alone (P<0.05). BM-MSC rats exhibited a significantly decreased bronchoalveolar lavage neutrophil percentage and LIS compared with that of LPS treated rats alone (P<0.05). In addition, the miRNA expression profile was determined following treatment with BM-MSCs via microarray analysis. A total of 95/690 miRNAs were differentially expressed following the treatment of BM-MSCs in rats with ALI. Among the 95 miRNAs, 66 were upregulated and 29 were downregulated; 9 miRNAs were significantly upregulated (miR-1843-3p, miR-323-3p, miR-183-5p, miR-182 and miR-196b-3p) or downregulated (miR-547-3p, miR-301b-5p, miR-503-3p and miR-142-3p). A total of 3 miRNAs were inversely expressed in ALI treated with BM-MSCs compared with untreated ALI. Of these 3 miRNAs, the expression of miR-142-3p and miR-503-3p was upregulated in the LPS groups and downregulated in the BM-MSC groups. miR-196b-3p was downregulated in the LPS group and upregulated in the BM-MSC groups. miRNAs have a role in cell proliferation, immune response, inflammation and apoptosis, which may be associated with the therapeutic effects of BM-MSCs in ALI. In summary, BM-MSCs improved multi-organ damage and attenuated lung injury. Different miRNA profiles were expressed following BM-MSC treatment of ALI. These dysregulated miRNAs participated in BM-MSC-mediated immunomodulation of ALI.
Collapse
Affiliation(s)
- Joonhong Park
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sikyoung Jeong
- Department of Emergency Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kicheol Park
- Clinical Research Institute, Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon 34943, Republic of Korea
| | - Keumjin Yang
- Clinical Research Institute, Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon 34943, Republic of Korea
| | - Soyoung Shin
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
289
|
Burgos-Aceves MA, Cohen A, Smith Y, Faggio C. A potential microRNA regulation of immune-related genes in invertebrate haemocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 621:302-307. [PMID: 29190554 DOI: 10.1016/j.scitotenv.2017.11.285] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 06/07/2023]
Abstract
Bivalve mollusks have been employed as sentinel organisms in environmental health programs due to their sedentary lifestyle, filter-feeding behavior and their ability to accumulate pathogens or toxin molecules inside tissues. Endocrine disrupting chemicals (EDCs) can be up taken and bioaccumulated, and due to sensibility of mollusks to these EDCs, being able to cause immune alterations. Recently, microRNAs (miRNAs) were shown to be involved in modulation and buffering developmental processes against the effects of environmental alterations and pathogenic microorganisms. Moreover, it is suggested that this miRNAs are incorporated into the estrogen-controlled immune network, regulating mechanism of immune gene expression at the posttranscriptional level, modulating immune responses as phagocytosis, redox reaction and apoptosis in bivalve haemocytes. Thus, miRNAs can be used as biomarkers that specifically elucidate immunotoxic effects caused by exogenous biotic or abiotic factors, and can act as useful tools in integrated monitoring environmental health programs. In this review, we aim to describe the investigations that have been carried out on miRNAs in bivalve mollusks, especially those associated with immune responses against infectious agents and xenobiotic exposure.
Collapse
Affiliation(s)
- Mario Alberto Burgos-Aceves
- Centro de Investigaciones Biológicas del Noroeste, S.C., Mar Bermejo 195, Col. Playa Palo de Sta. Rita, La Paz, BCS 23096, Mexico
| | - Amit Cohen
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Yoav Smith
- Genomic Data Analysis Unit, The Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 91120, Israel
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres, 31, 98166 Messina, Italy.
| |
Collapse
|
290
|
Momen-Heravi F, Bala S. miRNA regulation of innate immunity. J Leukoc Biol 2018; 103:1205-1217. [PMID: 29656417 DOI: 10.1002/jlb.3mir1117-459r] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/15/2018] [Accepted: 02/25/2018] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNA and are pivotal posttranscriptional regulators of both innate and adaptive immunity. They act by regulating the expression of multiple immune genes, thus, are the important elements to the complex immune regulatory network. Deregulated expression of specific miRNAs can lead to potential autoimmunity, immune tolerance, hyper-inflammatory phenotype, and cancer initiation and progression. In this review, we discuss the contributory pathways and mechanisms by which several miRNAs influence the development of innate immunity and fine-tune immune response. Moreover, we discuss the consequence of deregulated miRNAs and their pathogenic implications.
Collapse
Affiliation(s)
- Fatemeh Momen-Heravi
- Division of Periodontics, Section of Oral and Diagnostic Sciences, Columbia University College of Dental Medicine, New York, New York, USA
| | - Shashi Bala
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
291
|
Narayan N, Bracken CP, Ekert PG. MicroRNA-155 expression and function in AML: An evolving paradigm. Exp Hematol 2018; 62:1-6. [PMID: 29601851 DOI: 10.1016/j.exphem.2018.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) arises when immature myeloid blast cells acquire multiple, recurrent genetic and epigenetic changes that result in dysregulated proliferation. Acute leukemia is the most common form of pediatric cancer, with AML accounting for ~20% of all leukemias in children. The genomic aberrations that drive AML inhibit myeloid differentiation and activate signal transduction pathways that drive proliferation. MicroRNAs, a class of small (~22 nucleotide) noncoding RNAs that posttranscriptionally suppress the expression of specifically targeted transcripts, are also frequently dysregulated in AML, which may prove useful for the purposes of disease classification, prognosis, and future therapeutic approaches. MicroRNA expression profiles are associated with patient prognosis and responses to standard chemotherapy, including predicting therapy resistance in AML. miR-155 is the primary focus of this review because it has been repeatedly associated with poorer survival across multiple cohorts of adult and pediatric AML. We discuss some novel features of miR-155 expression in AML, in particular how the levels of expression can critically influence function. Understanding the role of microRNAs in AML and the ways in which microRNA expression influences AML biology is one means to develop novel and more targeted therapies.
Collapse
Affiliation(s)
- Nisha Narayan
- Murdoch Childrens Research Institute, Parkville, 3052, Australia
| | - Cameron P Bracken
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Paul G Ekert
- Murdoch Childrens Research Institute, Parkville, 3052, Australia.
| |
Collapse
|
292
|
D’Amore S, Härdfeldt J, Cariello M, Graziano G, Copetti M, Di Tullio G, Piglionica M, Scialpi N, Sabbà C, Palasciano G, Vacca M, Moschetta A. Identification of miR-9-5p as direct regulator of ABCA1 and HDL-driven reverse cholesterol transport in circulating CD14+ cells of patients with metabolic syndrome. Cardiovasc Res 2018; 114:1154-1164. [DOI: 10.1093/cvr/cvy077] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/22/2018] [Indexed: 01/01/2023] Open
Abstract
Abstract
Aims
Metabolic syndrome (MS) is a cluster of cardio-metabolic risk factors associated with atherosclerosis and low-grade inflammation. Using unbiased expression screenings in peripheral blood mononuclear cells, we depict here a novel expression chart of 678 genes and 84 microRNAs (miRNAs) controlling inflammatory, immune and metabolic responses. In order to further elucidate the link between inflammation and the HDL cholesterol pathway in MS, we focussed on the regulation of the ATP-binding cassette transporter A1 (ABCA1), a key player in cholesterol efflux (CE).
Methods and results
ABCA1 mRNA levels are suppressed in CD14+ cells of MS patients and are negatively correlated to body mass index (BMI), insulin-resistance (HOMA-IR) and cardiovascular risk, and positively to HDL cholesterol and CE. miRNA target in silico prediction identified a putative modulatory role of ABCA1 for the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) target miR-9-5p, whose expression pattern was up-regulated in CD14+ cells of MS patients, positively correlated to BMI, HOMA-IR, and triglycerides, and negatively to ABCA1 mRNA levels, HDL cholesterol and CE. Ectopic gain and loss of miR-9-5p function in macrophages modulated ABCA1 mRNA and protein levels, ABCA1 miRNA 3’-untranslated region target sequence reporter assay, and CE into HDL, thus confirming ABCA1 as a target of miR-9-5p.
Conclusions
We identified the NF-κB target miR-9-5p as a negative regulator of ABCA1 adding a novel target pathway in the relationship between inflammation and HDL-driven reverse cholesterol transport for prevention or treatment of atherosclerosis in MS.
Collapse
Affiliation(s)
- Simona D’Amore
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
- National Cancer Research Center, IRCCS Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco, 65, 70124 Bari, Italy
| | - Jennifer Härdfeldt
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
- INBB, National Institute for Biostructures and Biosystems, Viale delle Medaglie d'Oro 305, 00136 Rome, Italy
| | - Marica Cariello
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
- INBB, National Institute for Biostructures and Biosystems, Viale delle Medaglie d'Oro 305, 00136 Rome, Italy
| | - Giusi Graziano
- National Cancer Research Center, IRCCS Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco, 65, 70124 Bari, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics IRCCS ‘Casa Sollievo della Sofferenza’, Viale Cappuccini 1, 71013 San Giovanni Rotondo (FG), Italy
| | - Giuseppe Di Tullio
- De Matteis Laboratory, Telethon Institute of Genetics and Medicine, Piazza Aldo Moro 34, 80078 Pozzuoli (NA), Italy
| | - Marilidia Piglionica
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Natasha Scialpi
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Carlo Sabbà
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Giuseppe Palasciano
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Michele Vacca
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy
- National Cancer Research Center, IRCCS Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco, 65, 70124 Bari, Italy
| |
Collapse
|
293
|
Jingjing Z, Nan Z, Wei W, Qinghe G, Weijuan W, Peng W, Xiangpeng W. MicroRNA-24 Modulates Staphylococcus aureus-Induced Macrophage Polarization by Suppressing CHI3L1. Inflammation 2018; 40:995-1005. [PMID: 28303416 DOI: 10.1007/s10753-017-0543-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages play a crucial role in host innate anti-Staphylococcus aureus defense, which is tightly regulated by multiple factors, including microRNAs. A recent study showed that miR-24 plays an important role in macrophage polarization. Here, we investigated the biological function of miR-24 in S. aureus-stimulated macrophages. The results revealed that miR-24 expression was significantly decreased in both human and mouse macrophage cell lines with S. aureus stimulation in a time-dependent manner. Moreover, miR-24 overexpression significantly decreased the production of M1 phenotype markers, such as IL-6, iNOS, TNF-α, CD86, and CD80, whereas it increased the production of M2 markers, such as Arg1, CCL17, CCL22, CD163, and CD206, in S. aureus-stimulated macrophages. Conversely, knockdown of miR-24 promoted M1 macrophage polarization but diminished M2 macrophage polarization in S. aureus-stimulated macrophages. Furthermore, CHI3L1 was predicted as a target gene of miR-24 using bioinformatics software and identified by luciferase reporter assay. Additionally, miR-24 overexpression inhibited CHI3L1 expression and downregulated the downstream MAPK pathway in S. aureus-stimulated macrophages. Finally, CHI3L1 overexpression rescued macrophage polarization and MAPK pathway inhibition induced by miR-24 mimic transfection in S. aureus-stimulated macrophages. In conclusion, the data suggest that miR-24 serves as a molecular regulator in S. aureus-induced macrophage polarization through targeting of CHI3L1 and regulation of the MAPK pathway, which may provide a promising therapeutic target for S. aureus-related infections and inflammatory diseases.
Collapse
Affiliation(s)
- Zhang Jingjing
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China
| | - Zhang Nan
- Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wu Wei
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Guo Qinghe
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China
| | - Wang Weijuan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wang Peng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, China
| | - Wang Xiangpeng
- Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, No. 601 Jinsui Road, Hongqi District, Xinxiang, 453003, Henan Province, China.
| |
Collapse
|
294
|
ElSharawy A, Röder C, Becker T, Habermann JK, Schreiber S, Rosenstiel P, Kalthoff H. Concentration of circulating miRNA-containing particles in serum enhances miRNA detection and reflects CRC tissue-related deregulations. Oncotarget 2018; 7:75353-75365. [PMID: 27683108 PMCID: PMC5342746 DOI: 10.18632/oncotarget.12205] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 09/12/2016] [Indexed: 01/02/2023] Open
Abstract
The emerging potential of miRNAs as biomarkers for cancer detection demands parallel evaluation of strategies for reliable identification of disease-related signatures from easily accessible and pertinent body compartments. Here, we addressed whether efficient concentration of circulating miRNA-carrying particles is a rationale for miRNA biomarker discovery. We systematically compared miRNA signatures in 93 RNA preparations from three serum entities (whole serum, particle-concentrated, and particle-depleted fractions) and corresponding tissue samples from patients with colorectal cancer (CRC) as a model disease. Significant differences between whole sera and particle-concentrated serum fractions of CRC patients emerged for 45 of 742 tested miRNAs. Twenty-eight of these 45 miRNAs were differentially expressed between particle-concentrated serum fractions of metastatic CRC- and healthy individuals. Over half of these candidates (15 of 28) showed deregulations only in concentrated serum fractions, but not in whole sera, compared to the respective controls.Our results also provided evidence of a consistent downregulation of miR-486 and miR-92a, and further showed a possible "strand-specific" deregulation of extracellular miRNAs in CRC. More importantly, most of the identified miRNAs in the enriched sera reflected the patterns of the corresponding tumor tissues and showed links to cancer-related inflammation. Further investigation of seven serum pools revealed a subset of potential extracellular miRNA candidates to be implicated in both neoplastic and inflammatory bowel disease.Our findings demonstrate that enrichment and sensitive detection of miRNA carriers is a promising approach to detect CRC-related pathological changes in liquid biopsies, and has potential for clinical diagnostics.
Collapse
Affiliation(s)
- Abdou ElSharawy
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany.,Faculty of Sciences, Division of Biochemistry, Department of Chemistry, Damietta University, New Damietta City, Egypt
| | - Christian Röder
- Institute for Experimental Cancer Research, Christian-Albrechts-University, Kiel, Germany
| | - Thomas Becker
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Jens K Habermann
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany.,Clinic for Internal Medicine I, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
295
|
Corridoni D, Chapman T, Ambrose T, Simmons A. Emerging Mechanisms of Innate Immunity and Their Translational Potential in Inflammatory Bowel Disease. Front Med (Lausanne) 2018. [PMID: 29515999 PMCID: PMC5825991 DOI: 10.3389/fmed.2018.00032] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Activation of the innate immune system through pattern-recognition receptor (PRR) signaling plays a pivotal role in the early induction of host defense following exposure to pathogens. Loss of intestinal innate immune regulation leading aberrant immune responses has been implicated in the pathogenesis of inflammatory bowel disease (IBD). The precise role of PRRs in gut inflammation is not well understood, but considering their role as bacterial sensors and their genetic association with IBD, they likely contribute to dysregulated immune responses to the commensal microbiota. The purpose of this review is to evaluate the emerging functions of PRRs including their functional cross-talk, how they respond to mitochondrial damage, induce mitophagy or autophagy, and influence adaptive immune responses by interacting with the antigen presentation machinery. The review also summarizes some of the recent attempts to harness these pathways for therapeutic approaches in intestinal inflammation.
Collapse
Affiliation(s)
- Daniele Corridoni
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Thomas Chapman
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Tim Ambrose
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Alison Simmons
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
296
|
Wang MJ, Xu YY, Huang RY, Chen XM, Chen HM, Han L, Yan YH, Lu CJ. Role of an imbalanced miRNAs axis in pathogenesis of psoriasis: novel perspectives based on review of the literature. Oncotarget 2018; 8:5498-5507. [PMID: 27729619 PMCID: PMC5354926 DOI: 10.18632/oncotarget.12534] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/03/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Specific profile of microRNAs (miRNAs, miR) expressed in psoriasis has been identified in the past few years, while the studies on roles and molecular mechanisms of these miRNAs are still on the way. In our previous study, four specific miRNAs (miR-31, miR-203, hsa-miR-99a and miR-125b) were found to be specifically altered in psoriatic lesions.We therefore conducted a systematic literature review in this study to reveal the role of these miRNAs in the pathogenesis of psoriasis in order to inform future research. METHODS The related articles indexed in PubMed (MEDLINE) database were searched and analyzed. We identified eligible studies related to the mechanism research of miR-31, miR-203, hsa-miR-99a and miR-125b in psoriasis or psoriatic lesional skin from inception up to July 2016. The experts in the field of miRNAs and Psoriasis were involved in analysis process. RESULT Both miR-31 and miR-203 are dramatically upregulated in psoriatic lesions. The former plays the pro-proliferative, pro-differentiative and pro-inflammatory roles and the latter holds the potentials for anti-proliferation, pro-inflammation and pro-differentiation in psoriatic keratinocytes. Conversely, both hsa-miR-99a and miR-125b are significantly downregulated in psoriatic skin. These two miRNAs are able to inhibit proliferation while promote differentiation of psoriatic keratinocytes, and miR-125b can also suppress inflammation in psoriatic lesions. By analyzing the contexts related to these miRNAs, we found that each of them does not act alone but rather work in concert with other miRNAs. The imbalance between miR-31/miR-203and hsa-miR-99a/miR-125b may contribute to the intense proliferation and abnormal differentiation of psoriatic keratinocytes, which is a characteristic of pathogenesis of psoriasis. CONCLUSION An imbalanced miRNAs axis was for the first time outlined. Apparently, upregulation of miR-31/miR-203 and downregulation of hsa-miR-99a/miR-125b work together in concert to facilitate the development of psoriasis pathogenesis. Further work in this field holds the potentials to open a new way to study psoriasis.
Collapse
Affiliation(s)
- Mao-Jie Wang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Yong-Yue Xu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Run-Yue Huang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Section of Metabolic Diseases Research, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Xiu-Min Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Hai-Ming Chen
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Ling Han
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Yu-Hong Yan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Chuan-Jian Lu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| |
Collapse
|
297
|
Harrison SR, Scambler T, Oubussad L, Wong C, Wittmann M, McDermott MF, Savic S. Inositol-Requiring Enzyme 1-Mediated Downregulation of MicroRNA (miR)-146a and miR-155 in Primary Dermal Fibroblasts across Three TNFRSF1A Mutations Results in Hyperresponsiveness to Lipopolysaccharide. Front Immunol 2018; 9:173. [PMID: 29467762 PMCID: PMC5808292 DOI: 10.3389/fimmu.2018.00173] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/19/2018] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor (TNF)-receptor-associated periodic fever syndrome (TRAPS) is a rare monogenic autoinflammatory disorder characterized by mutations in the TNFRSF1A gene, causing TNF-receptor 1 (TNFR1) misfolding, increased cellular stress, activation of the unfolded protein response (UPR), and hyperresponsiveness to lipopolysaccharide (LPS). Both microRNA (miR)-146a and miR-155 provide negative feedback for LPS-toll-like receptor 2/4 signaling and cytokine production, through regulation of nuclear factor kappa B (NF-κB). In this study, we hypothesized that proinflammatory cytokine signaling in TRAPS downregulates these two miRs, resulting in LPS-induced hyperresponsiveness in TRAPS dermal fibroblasts (DFs), irrespective of the underlying genetic mutation. Primary DF were isolated from skin biopsies of TRAPS patients and healthy controls (HC). TNFR1 cell surface expression was measured using immunofluorescence. DF were stimulated with LPS, interleukin (IL)-1β, thapsigargin, or TNF, with and without inositol-requiring enzyme 1 (IRE1) inhibitor (4u8C), following which miR-146a and miR-155 expression was measured by RT-qPCR. IL-1β, IL-6, and TNF secretion was measured by enzyme-linked immunosorbent assays, and baseline expression of 384 different miRs was assessed using microfluidics assays. TNFR1 was found to be expressed on the surface of HC DF but expression was deficient in all samples with TRAPS-associated mutations. HC DF showed significant dose-dependent increases in both miR-146a and miR-155 expression levels in response to LPS; however, TRAPS DF failed to upregulate either miR-146a or miR-155 under the same conditions. This lack of miR-146a and miR-155 upregulation was associated with increased proinflammatory cytokine production in TRAPS DF in response to LPS challenge, which was abrogated by 4u8C. Incubation of HC DF with IL-1β led to downregulation of miR-146a and miR-155 expression, which was dependent on IRE1 enzyme. We observed global dysregulation of hundreds of other miRs at baseline in the TRAPS DF. In summary, these data suggest a mechanism whereby IL-1β, produced in response to activation of the UPR in TRAPS DF, downregulates miR-146a and miR-155, by inducing IRE1-dependent cleavage of both these miRs, thereby impairing negative regulation of NF-κB and increasing proinflammatory cytokine production.
Collapse
Affiliation(s)
- Stephanie R Harrison
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom
| | - Thomas Scambler
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom
| | - Lylia Oubussad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom
| | - Chi Wong
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom
| | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom.,Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom.,National Institute for Health Research-Leeds Biomedical Research Centre, Leeds, United Kingdom
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Leeds, United Kingdom.,National Institute for Health Research-Leeds Biomedical Research Centre, Leeds, United Kingdom.,Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
298
|
Rothschild DE, McDaniel DK, Ringel-Scaia VM, Allen IC. Modulating inflammation through the negative regulation of NF-κB signaling. J Leukoc Biol 2018; 103:10.1002/JLB.3MIR0817-346RRR. [PMID: 29389019 PMCID: PMC6135699 DOI: 10.1002/jlb.3mir0817-346rrr] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/16/2022] Open
Abstract
Immune system activation is essential to thwart the invasion of pathogens and respond appropriately to tissue damage. However, uncontrolled inflammation can result in extensive collateral damage underlying a diverse range of auto-inflammatory, hyper-inflammatory, and neoplastic diseases. The NF-κB signaling pathway lies at the heart of the immune system and functions as a master regulator of gene transcription. Thus, this signaling cascade is heavily targeted by mechanisms designed to attenuate overzealous inflammation and promote resolution. Mechanisms associated with the negative regulation of NF-κB signaling are currently under intense investigation and have yet to be fully elucidated. Here, we provide an overview of mechanisms that negatively regulate NF-κB signaling through either attenuation of signal transduction, inhibition of posttranscriptional signaling, or interference with posttranslational modifications of key pathway components. While the regulators discussed for each group are far from comprehensive, they exemplify common mechanistic approaches that inhibit this critical biochemical signaling cascade. Despite their diversity, a commonality among these regulators is their selection of specific targets at key inflection points in the pathway, such as TNF-receptor-associated factor family members or essential kinases. A better understanding of these negative regulatory mechanisms will be essential to gain greater insight related to the maintenance of immune system homeostasis and inflammation resolution. These processes are vital elements of disease pathology and have important implications for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Daniel E. Rothschild
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Dylan K. McDaniel
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
| | - Veronica M. Ringel-Scaia
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg VA 24061
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA 24061
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016
| |
Collapse
|
299
|
Wang H, Liu L, Liu X, Zhang M, Li X. Correlation between miRNAs and target genes in response to Campylobacter jejuni inoculation in chicken. Poult Sci 2018; 97:485-493. [DOI: 10.3382/ps/pex343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/14/2017] [Indexed: 12/19/2022] Open
|
300
|
Xie Y, Sun M, Xia Y, Shu R. An RNA-seq screen of P. gingivalis LPS treated human gingival fibroblasts. Arch Oral Biol 2018; 88:77-84. [PMID: 29407755 DOI: 10.1016/j.archoralbio.2018.01.002] [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: 03/02/2017] [Revised: 12/10/2017] [Accepted: 01/07/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND In gingival tissues, lipopolysaccharide (LPS) from Porphyromonas gingivalis (P. gingivalis) is the most critical stimulator for inducing inflammatory response. Human gingival fibroblasts (HGFs) are the major constituents of gingival connective tissues. The aim of this study was to investigate P. gingivalis LPS induced whole transcriptional profile in HGFs and the potential crosstalk between microRNAs (miRNAs) and inflammatory cytokines. METHODS RNA-seq was performed on HGFs with and without P. gingivalis LPS treatment. The gene expression of selected inflammatory cytokines and miRNAs induced by LPS at different time points was evaluated by quantitative RT-PCR. The protein expression of chemokines was further confirmed by ELISA. RESULTS Interestingly, most of the significantly changed genes (198/204) were up-regulated at 4 h after 10 μg/ml LPS stimulation, including inflammatory cytokines and miRNAs. Confirmed by quantitative RT-PCR, the mRNA levels of IL-1β, IL-6 and IL-8 showed single up-regulation peak (4 h/6 h) after 1 μg/ml and 10 μg/ml LPS treatment. Similarly, 1 μg/ml LPS induced single up-regulation peak (8 h) of miRNA-146a, -146b and -155 expression. However, 10 μg/ml LPS induced the increased expression of miRNA-146a and -155 at both early stage (2 h/4 h) and late stage (24 h). CONCLUSION Taken together, we investigated P. gingivalis LPS induced whole transcriptional profile, and the different behaviors of miRNA expression induced by different doses of LPS in HGFs.
Collapse
Affiliation(s)
- Yufeng Xie
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Mengjun Sun
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yiru Xia
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Rong Shu
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|