51
|
Nahalka J. The role of the protein-RNA recognition code in neurodegeneration. Cell Mol Life Sci 2019; 76:2043-2058. [PMID: 30980111 PMCID: PMC11105320 DOI: 10.1007/s00018-019-03096-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
MicroRNAs are small endogenous RNAs that pair and bind to sites on mRNAs to direct post-transcriptional repression. However, there is a possibility that microRNAs directly influence protein structure and activity, and this influence can be termed post-translational riboregulation. This conceptual review explores the literature on neurodegenerative disorders. Research on the association between neurodegeneration and RNA-repeat toxicity provides data that support a protein-RNA recognition code. For example, this code explains why hnRNP H and SFPQ proteins, which are involved in amyotrophic lateral sclerosis, are sequestered by the (GGGGCC)n repeat sequence. Similarly, it explains why MNBL proteins and (CTG)n repeats in RNA, which are involved in myotonic dystrophy, are sequestered into RNA foci. Using this code, proteins involved in diseases can be identified. A simple protein BLAST search of the human genome for amino acid repeats that correspond to the nucleotide repeats reveals new proteins among already known proteins that are involved in diseases. For example, the (CAG)n repeat sequence, when transcribed into possible peptide sequences, leads to the identification of PTCD3, Rem2, MESP2, SYPL2, WDR33, COL23A1, and others. After confirming this approach on RNA repeats, in the next step, the code was used in the opposite manner. Proteins that are involved in diseases were compared with microRNAs involved in those diseases. For example, a reasonable correspondence of microRNA 9 and 107 with amyloid-β-peptide (Aβ42) was identified. In the last step, a miRBase search for micro-nucleotides, obtained by transcription of a prion amino acid sequence, revealed new microRNAs and microRNAs that have previously been identified as involved in prion diseases. This concept provides a useful key for designing RNA or peptide probes.
Collapse
Affiliation(s)
- Jozef Nahalka
- Institute of Chemistry, Centre for Glycomics, Slovak Academy of Sciences, Dubravska cesta 9, 84538, Bratislava, Slovak Republic.
- Institute of Chemistry, Centre of Excellence for White-green Biotechnology, Slovak Academy of Sciences, Trieda Andreja Hlinku 2, 94976, Nitra, Slovak Republic.
| |
Collapse
|
52
|
Microarray profiling of lung long non-coding RNAs and mRNAs in lipopolysaccharide-induced acute lung injury mouse model. Biosci Rep 2019; 39:BSR20181634. [PMID: 30979832 PMCID: PMC6488857 DOI: 10.1042/bsr20181634] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in various biological processes as well as many respiratory diseases, while the role of lncRNAs in acute lung injury (ALI) remains unclear. The present study aimed to profile the expression of lung lncRNAs and mRNAs in lipopolysaccharide (LPS)-induced ALI mouse model. C57BL/6 mice were exposed to LPS or phosphate-buffered saline for 24 h, and lncRNAs and mRNAs were profiled by Arraystar mouse LncRNA Array V3.0. Bioinformatics analysis gene ontology including (GO) and pathway analysis and cell study in vitro was used to investigate potential mechanisms. Based on the microarray results, 2632 lncRNAs and 2352 mRNAs were differentially expressed between ALI and control mice. The microarray results were confirmed by the quantitative real-time PCR (qRT-PCR) results of ten randomized selected lncRNAs. GO analysis showed that the altered mRNAs were mainly related to the processes of immune system, immune response and defense response. Pathway analysis suggests that tumor necrosis factor (TNF) signaling pathway, NOD-like receptor pathway, and cytokine-cytokine receptor interaction may be involved in ALI. LncRNA-mRNA co-expression network analysis indicated that one individual lncRNA may interact with several mRNAs, and one individual mRNA may also interact with several lncRNAs. Small interfering RNA (siRNA) for ENSMUST00000170214.1, - ENSMUST00000016031.13 significantly inhibited LPS-induced TNF-α and interleukin (IL)-1β production in murine RAW264.7 macrophages. Our results found significant changes of lncRNAs and mRNAs in the lungs of LPS-induced ALI mouse model, and intervention targeting lncRNAs may attenuate LPS-induced inflammation, which may help to elucidate the role of lncRNAs in the pathogenesis and treatment of ALI.
Collapse
|
53
|
Silva AM, Moura SR, Teixeira JH, Barbosa MA, Santos SG, Almeida MI. Long noncoding RNAs: a missing link in osteoporosis. Bone Res 2019; 7:10. [PMID: 30937214 PMCID: PMC6437190 DOI: 10.1038/s41413-019-0048-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is a systemic disease that results in loss of bone density and increased fracture risk, particularly in the vertebrae and the hip. This condition and associated morbidity and mortality increase with population ageing. Long noncoding (lnc) RNAs are transcripts longer than 200 nucleotides that are not translated into proteins, but play important regulatory roles in transcriptional and post-transcriptional regulation. Their contribution to disease onset and development is increasingly recognized. Herein, we present an integrative revision on the studies that implicate lncRNAs in osteoporosis and that support their potential use as therapeutic tools. Firstly, current evidence on lncRNAs involvement in cellular and molecular mechanisms linked to osteoporosis and its major complication, fragility fractures, is reviewed. We analyze evidence of their roles in osteogenesis, osteoclastogenesis, and bone fracture healing events from human and animal model studies. Secondly, the potential of lncRNAs alterations at genetic and transcriptomic level are discussed as osteoporosis risk factors and as new circulating biomarkers for diagnosis. Finally, we conclude debating the possibilities, persisting difficulties, and future prospects of using lncRNAs in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Andreia Machado Silva
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - Sara Reis Moura
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| | - José Henrique Teixeira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Mário Adolfo Barbosa
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Susana Gomes Santos
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Maria Inês Almeida
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, Porto, Portugal
| |
Collapse
|
54
|
Toll-like receptor 9 signaling promotes autophagy and apoptosis via divergent functions of the p38/JNK pathway in human salivary gland cells. Exp Cell Res 2019; 375:51-59. [PMID: 30610847 DOI: 10.1016/j.yexcr.2018.12.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 12/25/2022]
Abstract
Abnormal signaling transduction in salivary gland cells is associated with the pathogenesis of Sjögren's syndrome (SS). Previously, we identified aberrant expression of toll-like receptor 9 (TLR9) in gland cells of SS patients and mouse models. In this study, we investigated the role of TLR9 and its downstream p38/mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) signaling in mediating apoptosis and autophagy in human salivary gland (HSG) cells. We selected either CpG-Odn, a classical TLR9 activator, or lentivirus-packaged TLR9 full-length cDNA to activate TLR9 signaling transduction. Activation of TLR9 signaling induced phosphorylation of its downstream protein kinases, p38/MAPK and JNK, in a time-dependent manner, and decreased HSG cell viability. Western blotting of LC3B-II and p62 in both normal and autophagic flux-administered conditions revealed elevated autophagy upon TLR9 activation. Observing the cell cytoplasm through transmission electron microscopy and mRFP-GFP-LC3B-tagged fluorescence confirmed an increased number of autophagosomes and autolysosomes in TLR9-activated cells. Bax/Bcl-2 ratio calculations, caspase-3 activity assays and Hoechst nuclear staining were utilized to confirm the involvement of apoptosis in TLR9 signaling activation. Furthermore, we selected SB239063, a p38/MAPK signaling inhibitor, and SP600125, a JNK inhibitor, to identify the functions of p38/MAPK and JNK in TLR9-mediated signaling transduction. Multiple approaches, including Western blotting assays, fluorescence assessments and caspase-3 activity measurements, confirmed that inhibition of p38/MAPK signaling ameliorated both autophagy and apoptosis in TLR9-activated HSG cells, whereas inhibition of JNK signaling attenuated apoptosis but failed to modulate autophagy in the models mentioned above. Our results indicate a divergent function of p38/MAPK and JNK in TLR9-mediated autophagy and apoptosis in salivary gland cells.
Collapse
|
55
|
Dong J, Zheng S, Yang X, Song X. Cell proliferation in kidney carcinoma is inhibited by lncRNA GASL1. EUR J INFLAMM 2019. [DOI: 10.1177/2058739219854598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Long noncoding RNA (lncRNA) GASL1 was identified as a novel lncRNA, which plays an important role in the proliferation and apoptosis of cells. This study aimed to compare the expression of GASL1 mRNA in kidney cancer cells and normal cells and detect the biological role of GASL1 in kidney cancer cell line A498. Polymerase chain reaction (PCR) was performed to examine the expression of GASL1 mRNA in kidney cancer tissues, normal tissues, and the cell lines. GASL1 overexpression was achieved in kidney cancer cell lines A498 through transfection. MTT was used to detect the effects of GASL1 overexpression in A498 cells. GASL1 mRNA was significantly overexpressed in adjacent normal tissues compared with renal cell carcinoma. The expression of GASL1 is lower in kidney cancer cell lines than in normal kidney epithelium cell line HREpiC. Overexpression of GASL1 inhibits the proliferation of renal carcinoma cell lines. GASL1 mRNA was down-regulated in kidney cancer tissues and may play a role in kidney cancer cell proliferation.
Collapse
Affiliation(s)
- Jianping Dong
- Department of Uropoiesis Surgical, Shouguang People’s Hospital, Shouguang, China
| | - Shiping Zheng
- Department of Uropoiesis Surgical, Shouguang People’s Hospital, Shouguang, China
| | - Xiaoyan Yang
- Department of Uropoiesis Surgical, Shouguang People’s Hospital, Shouguang, China
| | - Xiuyan Song
- Department of Uropoiesis Surgical, Shouguang People’s Hospital, Shouguang, China
| |
Collapse
|
56
|
Circular RNAs as Potential Biomarkers and Therapeutic Targets for Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:177-191. [PMID: 30919338 DOI: 10.1007/978-3-030-12668-1_10] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Epidemiological studies provide evidence of a continuous rise in metabolic diseases throughout industrialized countries. Metabolic diseases are commonly associated with different abnormalities that hold a key role in the emergence and progression of frequent disorders including diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD), obesity, metabolic syndrome and cardiovascular diseases. The burden of metabolic diseases is believed to arise through complex interaction between genetic and epigenetic factors, lifestyle changes and environmental exposure to triggering stimuli. The diagnosis and treatment of metabolic disorders continue to be an overwhelming challenge. Thus, the development of novel biomarkers may enhance the accuracy of the diagnosis at an early stage of the disease and allow effective intervention. Over the past decade, progress has been made in exploring the potential role of noncoding RNAs (ncRNAs) in the regulation of gene networks involved in metabolic diseases. A growing body of evidence now suggests that aberrant expression of circular RNAs (circRNAs) is relevant to the occurrence and development of metabolic diseases. Accordingly, circRNAs are proposed as predictive biomarkers and potential therapeutic targets for these diseases. As the field of circRNAs is rapidly evolving and knowledge is increasing, the present paper provides current understanding of the regulatory roles of these RNA species mainly in the pathogenesis of DM, NAFLD and obesity. Furthermore, some of the limitations to the promise of circRNAs and perspectives on their future research are discussed.
Collapse
|
57
|
Chen J, Yang X, Gong D, Cui Y, Hu J, Li H, Liu P, Li C, Cheng X, Liu L, Chen H, Zu X. Long noncoding RNA X-inactive specific transcript as a prognostic factor in cancer patients: A meta-analysis based on retrospective studies. Medicine (Baltimore) 2019; 98:e14095. [PMID: 30653128 PMCID: PMC6370154 DOI: 10.1097/md.0000000000014095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/AIMS Emerging evidence showed the long noncoding RNA X-inactive specific transcript (lncRNA XIST) may play a crucial role in various cancers. However, its prognostic value in cancer patients remains controversial. Therefore, we performed an in-depth meta-analysis to investigate the potential clinical value of lncRNA XIST as a prognostic marker for cancer patients. METHODS A comprehensive literature search was performed from PubMed, Embase and the Cochrane Central Search Library by January 2018. Pooled hazard ratios (HRs) or odds ratios (ORs) with 95% confidence interval (95% Cl) were calculated to evaluate the prognosis as well as clinicopathological parameters of XIST, respectively. RESULTS A total of 18 retrospective studies with 1351 cancer patients were included. Current meta-analysis revealed that elevated lncRNA XIST expression was associated with poor overall survival (OS) (HR = 2.14, 95% CI = 1.26-3.64; P = .005) and disease free survival (DFS) (HR = 4.52, 95% CI = 1.42-14.37; P = .011). The clinicopathological parameters analysis demonstrated that increased XIST expression was significantly associated with tumor size (OR = 2.93, 95% CI = 2.24-3.84; P < .001), clinical stage (OR = 2.73, 95% CI = 1.62-4.58; P < .001) and lymph node metastasis (OR = 2.44, 95% CI = 1.74-3.42; P < .001). In addition, subgroup analysis based on cancer type revealed that lncRNA XIST expression correlated with distant metastasis in digestive cancer (OR = 2.90, 95% CI = 1.80-4.68; P < .001). CONCLUSION The current meta-analysis results indicated lncRNA XIST expression level could serve as a prognostic predictor and biomarker in multiple cancers.
Collapse
Affiliation(s)
- Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Xiong Yang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Dongkui Gong
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Yu Cui
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Chao Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Xu Cheng
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Longfei Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Hequn Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha
| |
Collapse
|
58
|
Xue Z, Cui C, Liao Z, Xia S, Zhang P, Qin J, Guo Q, Chen S, Fu Q, Yin Z, Ye Z, Tang Y, Shen N. Identification of LncRNA Linc00513 Containing Lupus-Associated Genetic Variants as a Novel Regulator of Interferon Signaling Pathway. Front Immunol 2018; 9:2967. [PMID: 30619325 PMCID: PMC6305415 DOI: 10.3389/fimmu.2018.02967] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by augmented type I interferon signaling. High-throughput technologies have identified plenty of SLE susceptibility single-nucleotide polymorphisms (SNPs) yet the exact roles of most of them are still unknown. Functional studies are principally focused on SNPs in the coding regions, with limited attention paid to the SNPs in non-coding regions. Long non-coding RNAs (lncRNAs) are important players in shaping the immune response and show relationship to autoimmune diseases. In order to reveal the role of SNPs located near SLE related lncRNAs, we performed a transcriptome profiling of SLE patients and identified linc00513 as a significantly over expressed lncRNA containing functional SLE susceptibility loci in the promoter region. The risk-associated G allele of rs205764 and A allele of rs547311 enhanced linc00513 promoter activity and related to increased expression of linc00513 in SLE. We also identified linc00513 to be a novel positive regulator of type I interferon pathway by promoting the phosphorylation of STAT1 and STAT2. Elevated linc00513 expression positively correlated with IFN score in SLE patients. Linc00513 expression was higher in active disease patients than those inactive ones. In conclusion, our data identify two functional promoter variants of linc00513 that contribute to increased level of linc00513 and confer susceptibility on SLE. The study provides new insights into the genetics of SLE and extends the role of lncRNAs in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Zhixin Xue
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chaojie Cui
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhuojun Liao
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shiwei Xia
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pingjing Zhang
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jialin Qin
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiang Guo
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sheng Chen
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiong Fu
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
59
|
Gao Y, Li S, Zhang Z, Yu X, Zheng J. The Role of Long Non-coding RNAs in the Pathogenesis of RA, SLE, and SS. Front Med (Lausanne) 2018; 5:193. [PMID: 30018955 PMCID: PMC6038710 DOI: 10.3389/fmed.2018.00193] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid diseases are a group of systemic autoimmune diseases which affect multiple organs with largely unknown etiology. In the past decade, long non-coding RNAs (lncRNAs) have emerged as important regulators of biological processes and contribute deeply to immune cell development and immune responses. Substantial evidences have been accumulated showing that LncRNAs involved in the pathogenesis of the rheumatoid diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS). In this review, we summarize literature combined with bioinformatics methods to analyze the unique and common lncRNAs patterns in rheumatoid diseases and try to reveal the important function of lncRNAs in RA, SLE and SS.
Collapse
Affiliation(s)
- Yunzhen Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Shasha Li
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Xinhua Yu
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, Members of the German Center for Lung Research, Borstel, Germany
| | - Junfeng Zheng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| |
Collapse
|
60
|
Hu G, Niu F, Humburg BA, Liao K, Bendi S, Callen S, Fox HS, Buch S. Molecular mechanisms of long noncoding RNAs and their role in disease pathogenesis. Oncotarget 2018; 9:18648-18663. [PMID: 29719633 PMCID: PMC5915100 DOI: 10.18632/oncotarget.24307] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/13/2018] [Indexed: 12/13/2022] Open
Abstract
LncRNAs are long non-coding regulatory RNAs that are longer than 200 nucleotides. One of the major functions of lncRNAs is the regulation of specific gene expression at multiple steps including, recruitment and expression of basal transcription machinery, post-transcriptional modifications and epigenetics [1]. Emerging evidence suggests that lncRNAs also play a critical role in maintaining tissue homeostasis during physiological and pathological conditions, lipid homeostasis, as well as epithelial and smooth muscle cell homeostasis, a topic that has been elegantly reviewed [2-5]. While aberrant expression of lncRNAs has been implicated in several disease conditions, there is paucity of information about their contribution to the etiology of diseases [6]. Several studies have compared the expression of lncRNAs under normal and cancerous conditions and found differential expression of several lncRNAs, suggesting thereby an involvement of lncRNAs in disease processes [7, 8]. Furthermore, the ability of lncRNAs to influence epigenetic changes also underlies their role in disease pathogenesis since epigenetic regulation is known to play a critical role in many human diseases [1]. LncRNAs thus are not only involved in homeostatic functioning but also play a vital role in the progression of many diseases, thereby underscoring their potential as novel therapeutic targets for the alleviation of a variety of human disease conditions.
Collapse
Affiliation(s)
- Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bree A. Humburg
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sunil Bendi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shannon Callen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard S. Fox
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
61
|
lncRNAs Regulate Innate Immune Responses and Their Roles in Macrophage Polarization. Mediators Inflamm 2018; 2018:8050956. [PMID: 29599646 PMCID: PMC5828099 DOI: 10.1155/2018/8050956] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is the first line of defense against microbial pathogens. The activated innate immune system plays important roles in eliciting antimicrobial defenses. Despite the benefits of innate immune responses, excessive inflammation will cause host damage. Thus, tight regulation of these processes is required for the maintenance of immune homeostasis. Recently, a new class of long noncoding RNAs (lncRNAs) has emerged as important regulators in many physiological and pathological processes. Dysregulated lncRNAs have been found to be associated with excessive or uncontrolled inflammation. In this brief review, we summarize the roles of functional lncRNAs in regulating innate immune responses. We also discuss the roles of lncRNAs in macrophage polarization, an important molecular event in the innate immune responses.
Collapse
|