1
|
Ghahramani Almanghadim H, Karimi B, Valizadeh S, Ghaedi K. Biological functions and affected signaling pathways by Long Non-Coding RNAs in the immune system. Noncoding RNA Res 2025; 10:70-90. [PMID: 39315339 PMCID: PMC11417496 DOI: 10.1016/j.ncrna.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/14/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Recently, the various regulative functions of long non-coding RNAs (LncRNAs) have been well determined. Recently, the vital role of LncRNAs as gene regulators has been identified in the immune system, especially in the inflammatory response. All cells of the immune system are governed by a complex and ever-changing gene expression program that is regulated through both transcriptional and post-transcriptional processes. LncRNAs regulate gene expression within the cell nucleus by influencing transcription or through post-transcriptional processes that affect the splicing, stability, or translation of messenger RNAs (mRNAs). Recent studies in immunology have revealed substantial alterations in the expression of lncRNAs during the activation of the innate immune system as well as the development, differentiation, and activation of T cells. These lncRNAs regulate key aspects of immune function, including the manufacturing of inflammatory molecules, cellular distinction, and cell movement. They do this by modulating protein-protein interactions or through base pairing with RNA and DNA. Here we review the current understanding of the mechanism of action of lncRNAs as novel immune-related regulators and their impact on physiological and pathological processes related to the immune system, including autoimmune diseases. We also highlight the emerging pattern of gene expression control in important research areas at the intersection between immunology and lncRNA biology.
Collapse
Affiliation(s)
| | - Bahareh Karimi
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Sepehr Valizadeh
- Department of Internal Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
NfκB signaling dynamics and their target genes differ between mouse blood cell types and induce distinct cell behavior. Blood 2022; 140:99-111. [PMID: 35468185 DOI: 10.1182/blood.2021012918] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 03/16/2022] [Indexed: 11/20/2022] Open
Abstract
Cells can use signaling pathway activity over time (i.e., dynamics) to control cell fates. However, little is known about the potential existence and function of signaling dynamics in primary hematopoietic stem and progenitor cells (HSPCs). Here, we use time-lapse imaging and tracking of single murine HSPCs from GFP-p65/H2BmCherry reporter mice to quantify their nuclear factor κB (NfκB) activity dynamics in response to TNFα and IL1β. We find response dynamics to be heterogeneous between individual cells, with cell type specific dynamics distributions. Transcriptome sequencing of single cells physically isolated after live dynamics quantification shows activation of different target gene programs in cells with different dynamics. Finally, artificial induction of oscillatory NfκB activity causes changes in GMP behavior. Thus, HSPC behavior can be influenced by signaling dynamics, which are tightly regulated during hematopoietic differentiation and enable cell type specific responses to the same signaling inputs.
Collapse
|
3
|
The potency of lncRNA MALAT1/miR-155/CTLA4 axis in altering Th1/Th2 balance of asthma. Biosci Rep 2021; 40:221794. [PMID: 31909418 PMCID: PMC7024843 DOI: 10.1042/bsr20190397] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives: The present study examined if the metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)/miR-155/CTLA-4 axis was involved in modifying Th1/Th2 balance, a critical indicator for asthma progression. Methods: Altogether 772 asthma patients and 441 healthy controls were recruited, and their blood samples were collected to determine expressional levels of MALAT1, miR-155, CTLA-4, T-bet, GATA3, Th1-type cytokines and Th2-type cytokines. The CD4+ T cells were administered with pcDNA3.1-MALAT1, si-MALAT1, miR-155 mimic and miR-155 inhibitor to assess their effects on cytokine release. The luciferase reporter gene assay was also adopted to evaluate the sponging relationships between MALAT1 and miR-155, as well as between miR-155 and CTLA-4. Results: Over-expressed MALAT1 and under-expressed miR-155 were more frequently detected among asthma patients who showed traits of reduced forced expiratory failure volume in 1 s (FEV1), FEV1/forced vital capacity (FVC) and FEV1% of predicted (P<0.05). Moreover, MALAT1 expression was negatively expressed with the Th1/Th2 and T-bet/GATA3 ratios, yet miR-155 expression displayed a positively correlation with the ratios (P<0.05). Additionally, the IFN-γ, IL-2 and T-bet levels were reduced under the influence of pcDNA3.1-MALAT1 and miR-155 inhibitor, while levels of IL-4, IL-10 and GATA3 were raised under identical settings (P<0.05). Furthermore, MALAT1 constrained expression of miR-155 within CD4+ T cells by sponging it, and CTLA-4 could interfere with the effects of MALAT1 and miR-155 on Th1/Th2 balance and T-bet/Gata3 ratio (P<0.05). Conclusion: MALAT1 sponging miR-155 was involved with regulation of Th1/Th2 balance within CD4+ T cells, which might aid to develop therapies for amelioration of asthmatic inflammation.
Collapse
|
4
|
LncRNAs and Immunity: Coding the Immune System with Noncoding Oligonucleotides. Int J Mol Sci 2021; 22:ijms22041741. [PMID: 33572313 PMCID: PMC7916124 DOI: 10.3390/ijms22041741] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) represent key regulators of gene transcription during the inflammatory response. Recent findings showed lncRNAs to be dysregulated in human diseases, such as inflammatory bowel disease, diabetes, allergies, asthma, and cancer. These noncoding RNAs are crucial for immune mechanism, as they are involved in differentiation, cell migration and in the production of inflammatory mediators through regulating protein–protein interactions or their ability to assemble with RNA and DNA. The last interaction can occur in cis or trans and is responsible for all the possible lncRNAs biological effects. Our proposal is to provide an overview on lncRNAs roles and functions related to immunity and immune mediated diseases, since these elucidations could be beneficial to untangle the complex bond between them.
Collapse
|
5
|
Buda P, Chyb M, Smorczewska-Kiljan A, Wieteska-Klimczak A, Paczesna A, Kowalczyk-Domagała M, Okarska-Napierała M, Sobalska-Kwapis M, Grochowalski Ł, Słomka M, Sitek A, Ksia Żyk J, Strapagiel D. Association Between rs12037447, rs146732504, rs151078858, rs55723436, and rs6094136 Polymorphisms and Kawasaki Disease in the Population of Polish Children. Front Pediatr 2021; 9:624798. [PMID: 33692975 PMCID: PMC7937642 DOI: 10.3389/fped.2021.624798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Kawasaki disease (KD) is an acute self-limited febrile vasculitis that mainly affects young children. Coronary artery involvement is the most serious complication in children with KD. It is currently the leading cause of acquired cardiac disease in children from developed countries. Literature data indicate a significant role of genetic susceptibility to KD. Objective: The aim of this study was to perform the first Genome-Wide Association Study (GWAS) in a population of Polish children with KD and identify susceptible genes involved in the pathogenesis of KD. Materials and Methods: The blood samples of Kawasaki disease patients (n = 119) were collected between 2016 and 2020, isolated and stored at the Department of Pediatrics, Nutrition and Metabolic Diseases, Children's Memorial Health Institute in Warsaw. The control group was based on Polish donors (n = 6,071) registered as the POPULOUS collection at the Biobank Lab of The Department of Molecular Biophysics in University of Lodz. DNA samples were genotyped for 558,231 Single Nucleotide Polymorphisms (SNPs) using the 24 × 1 Infinium HTS Human Core Exome microarrays according to the protocol provided by the manufacturer. In order to discover and verify genetic risk-factors for KD, association analysis was carried out using PLINK 1.9. Results: Of all 164,395 variants, 5 were shown to occur statistically (padjusted < 0.05) more frequent in Kawasaki disease patients than in controls. Those are: rs12037447 in non-coding sequence (padjusted = 8.329 × 10-4, OR = 8.697, 95% CI; 3.629-20.84) and rs146732504 in KIF25 (padjusted = 0.007354, OR = 11.42, 95% CI; 3.79-34.43), rs151078858 in PTPRJ (padjusted = 0.04513, OR = 8.116, 95% CI; 3.134-21.01), rs55723436 in SPECC1L (padjusted = 0.04596, OR = 5.596, 95% CI; 2.669-11.74), rs6094136 in RPN2 (padjusted = 0.04755, OR = 10.08, 95% CI; 3.385-30.01) genes. Conclusion: Polymorphisms of genes KIF25, PTRPJ, SPECC1L, RNP2 may be linked with the incidence of Kawasaki disease in Polish children.
Collapse
Affiliation(s)
- Piotr Buda
- Department of Pediatrics, Nutrition, and Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland
| | - Maciej Chyb
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Anna Smorczewska-Kiljan
- Department of Pediatrics, Nutrition, and Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Wieteska-Klimczak
- Department of Pediatrics, Nutrition, and Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland
| | - Agata Paczesna
- Department of Cardiology, The Children's Memorial Health Institute, Warsaw, Poland
| | | | | | - Marta Sobalska-Kwapis
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland.,BBMRI.pl Consortium, Wrocław, Poland
| | - Łukasz Grochowalski
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Marcin Słomka
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland.,BBMRI.pl Consortium, Wrocław, Poland
| | - Aneta Sitek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Janusz Ksia Żyk
- Department of Pediatrics, Nutrition, and Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland
| | - Dominik Strapagiel
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland.,BBMRI.pl Consortium, Wrocław, Poland
| |
Collapse
|
6
|
Schut CH, Farzan A, Fraser RS, Ainslie-Garcia MH, Friendship RM, Lillie BN. Identification of single-nucleotide variants associated with susceptibility to Salmonella in pigs using a genome-wide association approach. BMC Vet Res 2020; 16:138. [PMID: 32414370 PMCID: PMC7227190 DOI: 10.1186/s12917-020-02344-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Salmonella enterica serovars are a major cause of foodborne illness and have a substantial impact on global human health. In Canada, Salmonella is commonly found on swine farms and the increasing concern about drug use and antimicrobial resistance associated with Salmonella has promoted research into alternative control methods, including selecting for pig genotypes associated with resistance to Salmonella. The objective of this study was to identify single-nucleotide variants in the pig genome associated with Salmonella susceptibility using a genome-wide association approach. Repeated blood and fecal samples were collected from 809 pigs in 14 groups on farms and tonsils and lymph nodes were collected at slaughter. Sera were analyzed for Salmonella IgG antibodies by ELISA and feces and tissues were cultured for Salmonella. Pig DNA was genotyped using a custom 54 K single-nucleotide variant oligo array and logistic mixed-models used to identify SNVs associated with IgG seropositivity, shedding, and tissue colonization. RESULTS Variants in/near PTPRJ (p = 0.0000066), ST6GALNAC3 (p = 0.0000099), and DCDC2C (n = 3, p < 0.0000086) were associated with susceptibility to Salmonella, while variants near AKAP12 (n = 3, p < 0.0000358) and in RALGAPA2 (p = 0.0000760) may be associated with susceptibility. CONCLUSIONS Further study of the variants and genes identified may improve our understanding of neutrophil recruitment, intracellular killing of bacteria, and/or susceptibility to Salmonella and may help future efforts to reduce Salmonella on-farm through genetic approaches.
Collapse
Affiliation(s)
- Corinne H Schut
- Department of Pathobiology, University of Guelph, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada
| | - Abdolvahab Farzan
- Department of Pathobiology, University of Guelph, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada
- Department of Population Medicine, University of Guelph, Guelph, Ontario, Canada
| | - Russell S Fraser
- Department of Pathobiology, University of Guelph, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada
- Present address: Department of Pathology and Microbiology, Atlantic Veterinary College, University of PEI, Charlottetown, Prince Edward Island, Canada
| | | | - Robert M Friendship
- Department of Population Medicine, University of Guelph, Guelph, Ontario, Canada
| | - Brandon N Lillie
- Department of Pathobiology, University of Guelph, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
7
|
Pei W, Zhang Y, Li X, Luo M, Chen T, Zhang M, Zhong M, Lv K. LncRNA AK085865 depletion ameliorates asthmatic airway inflammation by modulating macrophage polarization. Int Immunopharmacol 2020; 83:106450. [PMID: 32247269 DOI: 10.1016/j.intimp.2020.106450] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 01/19/2023]
Abstract
Accumulating evidence indicates that regulators of macrophages polarization may play a key role in the development of allergic asthma (AA). However, the exact role of long non-coding RNAs (lncRNAs) in regulating in macrophages polarization in the pathogenesis of dermatophagoides farinae protein 1(Der f1)-induced AA is not fully understood. The purpose of this study was to determine the function of lncRNA AK085865 in regulating macrophages in AA. Here we report that lncRNA AK085865 served as a critical regulator of macrophages polarization and reduced the pathological progress of asthmatic airway inflammation. In response to the challenge of Der f1, AK085865-/- mice displayed attenuated allergic airway inflammation, including decreased eosinophil in BALF and reduced production of IgE, which were associated with decreased mucous glands and goblet cell hyperplasia. In addition, Der f1-treated AK085865-/- mice show fewer M2 macrophages when compared with WT asthmatic mice. After adopting bone marrow-derived macrophages (BMDM, M0) from WT mice, Der f1-treated AK085865-/- mice also revealed a light inflammatory reactions. We further observed that the percentage of type II innate immune lymphoid cells (ILC2s) decreased in AK085865-/- asthmatic mice. Moreover, M2 macrophages helped promote the differentiation of ILC2s, probably through the exosomal pathway secreted by M2 macrophages. Taken together, these findings reveal that AK085865 depletion can ameliorate asthmatic airway inflammation by modulating macrophage polarization and M2 macrophages can promote the differentiation of innate lymphoid cells progenitor (ILCP) into ILC2s.
Collapse
Affiliation(s)
- Weiya Pei
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Yingying Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Department of Laboratory Medicine (Wannan Medical College), Wuhu, PR China
| | - Xueqin Li
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Mengsha Luo
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Tianbing Chen
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Mengying Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Min Zhong
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes (Wannan Medical College), Wuhu, PR China; Central Laboratory, The First Affiliated Hospital of Wannan Medical College, Wuhu, PR China.
| |
Collapse
|
8
|
Takahashi K, Kim RH, Pasic L, He L, Nagasaka S, Katagiri D, May T, Shimizu A, Harris RC, Mernaugh RL, Takahashi T. Agonistic anti-CD148 monoclonal antibody attenuates diabetic nephropathy in mice. Am J Physiol Renal Physiol 2020; 318:F647-F659. [PMID: 31984788 DOI: 10.1152/ajprenal.00288.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CD148 is a transmembrane protein tyrosine phosphatase (PTP) that is expressed in the renal vasculature, including the glomerulus. Previous studies have shown that CD148 plays a role in the negative regulation of growth factor signals (including epidermal growth factor and vascular endothelial growth factor), suppressing cell proliferation and transformation. However, the role of CD148 in kidney disease remains unknown. Here, we generated an agonistic anti-CD148 antibody and evaluated its effects in murine diabetic nephropathy (DN). Monoclonal antibodies (mAbs) against the mouse CD148 ectodomain sequence were generated by immunizing CD148 knockout (CD148KO) mice. The mAbs that increased CD148 activity were selected by biological (proliferation) and biochemical (PTP activity) assays. The mAb (18E1) that showed strong agonistic activity was injected (10 mg/kg ip) in streptozotocin-induced wild-type and CD148KO diabetic mice for 6 wk, and the renal phenotype was then assessed. The effects of 18E1 mAb in podocyte growth factor signals were also assessed in culture. Compared with control IgG, 18E1 mAb significantly decreased albuminuria and mesangial expansion without altering hyperglycemia and blood pressure in wild-type diabetic mice. Immunohistochemical evaluation showed that 18E1 mAb significantly prevented the reduction of podocyte number and nephrin expression and decreased glomerular fibronectin expression and renal macrophage infiltration. The 18E1 mAb showed no effects in CD148KO diabetic mice. Furthermore, we demonstrated that 18E1 mAb reduces podocyte epidermal growth factor receptor signals in culture and in diabetic mice. These findings suggest that agonistic anti-CD148 mAb attenuates DN in mice, in part by reducing epidermal growth factor receptor signals in podocytes. This antibody may be used for the treatment of early DN.
Collapse
Affiliation(s)
- Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rachel H Kim
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lejla Pasic
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Lilly He
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shinya Nagasaka
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Daisuke Katagiri
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tracy May
- Vanderbilt University Antibody and Protein Resource, Nashville, Tennessee
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
9
|
Yue D, Zhao J, Chen H, Guo M, Chen C, Zhou Y, Xu L. MicroRNA-7, synergizes with RORα, negatively controls the pathology of brain tissue inflammation. J Neuroinflammation 2020; 17:28. [PMID: 31959187 PMCID: PMC6970296 DOI: 10.1186/s12974-020-1710-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/13/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Accumulating evidence has documented that microRNA-7 (miR-7) plays an important role in the pathology of various diseases. However, the potential role of miR-7 in brain tissue inflammation (BTI) remains unclear. METHODS We detected the expression of miR-7 in LPS-induced murine BTI model and observed the possible effects of miR-7 deficiency on the pathology of BTI. To elucidate the mechanism, the target gene of miR-7 was screened out by Gene chip assay and its potential roles in BTI were evaluated by Western blot, immunofluorescence, and RNAi assay, respectively. RESULTS MiR-7 was upregulated in brain tissue in BTI mice and its deficiency could significantly aggravate the pathology of brain tissue. Moreover, RORα, a new target molecule of miR-7, was upregulated in brain tissue from miR-7 deficiency BTI mice. Of note, downregulation of RORα could remarkably exacerbate the pathology of brain tissue and elevate the transduction of NF-κB and ERK1/2 signaling pathways in brain tissue from miR-7 deficiency BTI mice. Furthermore, RORα and miR-7 were dominantly co-expressed in neurons of BTI mice. Finally, RORα synergized with miR-7 to control the inflammatory reaction of neuronal cells in response to LPS stimulation. CONCLUSIONS MiR-7 expression is upregulated in BTI model. Moreover, miR-7 synergizes with its target gene RORα to control the inflammation reaction of neurons, thereby orchestrating the pathology of BTI.
Collapse
Affiliation(s)
- Dongxu Yue
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Huizi Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Ya Zhou
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China.,Department of Medical Physics, Zunyi Medical University, Zunyi, 563099, Guizhou, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, 563099, Guizhou, China. .,Department of Immunology, Zunyi Medical University, Zunyi, 563099, Guizhou, China.
| |
Collapse
|
10
|
Joslyn RC, Forero A, Green R, Parker SE, Savan R. Long Noncoding RNA Signatures Induced by Toll-Like Receptor 7 and Type I Interferon Signaling in Activated Human Plasmacytoid Dendritic Cells. J Interferon Cytokine Res 2019; 38:388-405. [PMID: 30230983 DOI: 10.1089/jir.2018.0086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) exhibit highly lineage-specific expression and act through diverse mechanisms to exert control over a wide range of cellular processes. lncRNAs can function as potent modulators of innate immune responses through control of transcriptional and posttranscriptional regulation of mRNA expression and processing. Recent studies have demonstrated that lncRNAs participate in the regulation of antiviral responses and autoimmune disease. Despite their emerging role as immune mediators, the mechanisms that govern lncRNA expression and function have only begun to be characterized. In this study, we explore the role of lncRNAs in human plasmacytoid dendritic cells (pDCs), which are critical sentinel sensors of viral infection and contribute to the development of autoimmune disease. Using genome-wide sequencing approaches, we dissect the contributions of Toll-like receptor 7 (TLR7) and type I interferon (IFN-I) in the regulation of coding and noncoding RNA expression in CAL-1 pDCs treated with R848 or IFNβ. Functional enrichment analysis reveals both the unique and synergistic roles of TLR7 and IFN-I signaling in the orchestration of pDC function. These observations were consistent with primary cell immune responses elicited by detection of viral infection. We identified and characterized the conditional TLR7- and IFN-I-dependent regulation of 588 lncRNAs. Dysregulation of these lncRNAs could significantly alter pDC maturation, IFN-I and inflammatory cytokine production, antigen presentation, costimulation or tolerance cues, turnover, or localization, all consequential events during viral infection or IFN-I-driven autoimmune diseases such as systemic lupus erythematosus. These findings demonstrate the differential responsiveness of lncRNAs to unique immune stimuli, uncover regulatory mechanisms of lncRNA expression, and reveal a novel and tractable platform for the study of lncRNA expression and function.
Collapse
Affiliation(s)
- Rochelle C Joslyn
- 1 Department of Immunology and University of Washington , Seattle, Washington
| | - Adriana Forero
- 1 Department of Immunology and University of Washington , Seattle, Washington
| | - Richard Green
- 1 Department of Immunology and University of Washington , Seattle, Washington.,2 Center for Innate Immunity and Immune Disease, University of Washington , Seattle, Washington
| | - Stephen E Parker
- 1 Department of Immunology and University of Washington , Seattle, Washington
| | - Ram Savan
- 1 Department of Immunology and University of Washington , Seattle, Washington.,2 Center for Innate Immunity and Immune Disease, University of Washington , Seattle, Washington
| |
Collapse
|
11
|
Lu H, Deng S, Zheng M, Hu K. iTRAQ plasma proteomics analysis for candidate biomarkers of type 2 incipient diabetic nephropathy. Clin Proteomics 2019; 16:33. [PMID: 31384238 PMCID: PMC6668123 DOI: 10.1186/s12014-019-9253-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Background Diabetic nephropathy is the most frequent cause of end-stage renal disease worldwide. Identification of biomarkers for diabetic nephropathy for early diagnosis may be the key to avoiding damage from this condition. Methods Proteomic iTRAQ technology was first used to identify differentially expressed plasma proteins in type 2 incipient diabetic nephropathy (IDN) using a Q-Exactive mass spectrometer. Results Compared with controls, 57 proteins (32 upregulated and 25 downregulated proteins) were identified. Furthermore, the gelsolin, collectin-11, PTPRJ, and AKAP-7 proteins were confirmed by Western blots as candidate biomarkers for type 2 IDN through ROC analysis. Conclusions These findings offer a theoretical basis for the early treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Hongmei Lu
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Shaodong Deng
- 1The Second Clinical Medical College, Guangdong Medical University, Dongguan, 523808 China
| | - Minghui Zheng
- 2Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120 China
| | - Kunhua Hu
- 3Proteomics Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| |
Collapse
|
12
|
Hadjicharalambous MR, Lindsay MA. Long Non-Coding RNAs and the Innate Immune Response. Noncoding RNA 2019; 5:ncrna5020034. [PMID: 31010202 PMCID: PMC6630897 DOI: 10.3390/ncrna5020034] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Innate immunity provides the initial defence against infection and it is now clear that long non-coding RNAs (lncRNAs) are important regulators of this response. Following activation of the innate response, we commonly see rapid induction of these lncRNAs and this is often mediated via the pro-inflammatory transcription factor, nuclear factor-κB (NF-κB). Knockdown studies have shown that lncRNAs tend to act in trans to regulate the expression of multiple inflammatory mediators and other responses. Mechanistically, many lncRNAs have demonstrated acting through heterogeneous nuclear ribonucleoproteins, complexes that are implicated chromatin re-modelling, transcription process and translation. In addition, these lncRNAs have also been shown to interact with multiple other proteins involved in the regulation of chromatin re-modelling, as well as those proteins involved in intracellular immune signalling, which include NF-κB. In this review, we will describe the evidence that supports this emerging role of lncRNA in the innate immune response.
Collapse
Affiliation(s)
| | - Mark A Lindsay
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
13
|
Rougeot J, Torraca V, Zakrzewska A, Kanwal Z, Jansen HJ, Sommer F, Spaink HP, Meijer AH. RNAseq Profiling of Leukocyte Populations in Zebrafish Larvae Reveals a cxcl11 Chemokine Gene as a Marker of Macrophage Polarization During Mycobacterial Infection. Front Immunol 2019; 10:832. [PMID: 31110502 PMCID: PMC6499218 DOI: 10.3389/fimmu.2019.00832] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/29/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are phagocytic cells from the innate immune system, which forms the first line of host defense against invading pathogens. These highly dynamic immune cells can adopt specific functional phenotypes, with the pro-inflammatory M1 and anti-inflammatory M2 polarization states as the two extremes. Recently, the process of macrophage polarization during inflammation has been visualized by real time imaging in larvae of the zebrafish. This model organism has also become widely used to study macrophage responses to microbial pathogens. To support the increasing use of zebrafish in macrophage biology, we set out to determine the complete transcriptome of zebrafish larval macrophages. We studied the specificity of the macrophage signature compared with other larval immune cells and the macrophage-specific expression changes upon infection. We made use of the well-established mpeg1, mpx, and lck fluorescent reporter lines to sort and sequence the transcriptome of larval macrophages, neutrophils, and lymphoid progenitor cells, respectively. Our results provide a complete dataset of genes expressed in these different immune cell types and highlight their similarities and differences. Major differences between the macrophage and neutrophil signatures were found within the families of proteinases. Furthermore, expression of genes involved in antigen presentation and processing was specifically detected in macrophages, while lymphoid progenitors showed expression of genes involved in macrophage activation. Comparison with datasets of in vitro polarized human macrophages revealed that zebrafish macrophages express a strongly homologous gene set, comprising both M1 and M2 markers. Furthermore, transcriptome analysis of low numbers of macrophages infected by the intracellular pathogen Mycobacterium marinum revealed that infected macrophages change their transcriptomic response by downregulation of M2-associated genes and overexpression of specific M1-associated genes. Among the infection-induced genes, a homolog of the human CXCL11 chemokine gene, cxcl11aa, stood out as the most strongly overexpressed M1 marker. Upregulation of cxcl11aa in Mycobacterium-infected macrophages was found to require the function of Myd88, a critical adaptor molecule in the Toll-like and interleukin 1 receptor pathways that are central to pathogen recognition and activation of the innate immune response. Altogether, our data provide a valuable data mining resource to support infection and inflammation research in the zebrafish model.
Collapse
Affiliation(s)
- Julien Rougeot
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Vincenzo Torraca
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Ania Zakrzewska
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Zakia Kanwal
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | | - Frida Sommer
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Herman P Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
14
|
Wang Y, Song X, Li Z, Liu B. Long non-coding RNAs in coronary atherosclerosis. Life Sci 2018; 211:189-197. [PMID: 30195033 DOI: 10.1016/j.lfs.2018.08.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/26/2022]
Abstract
Coronary atherosclerosis (CAS), a leading cause of cardiovascular disease, is a major cause of death worldwide. CAS is a chronic disease in the aorta that can be caused by dyslipidemia, abnormal glucose metabolism, endothelial cell dysfunction, vascular smooth muscle cell (VSMC) or fibrous connective tissue hyperplasia, immune inflammatory reactions, and many other factors. The pathogenesis of CAS is not fully understood, as it is a complex lesion complicated by multiple factors. Damage-response theories have put forward endothelial cell (EC) injury as the initiating factor for CAS; the addition of lipid metabolism disorders may enhance monocyte adhesion, increase the proliferation and migration of fibroblasts and VSMCs, and accelerate the development of CAS. Furthermore, inflammatory and immune responses can create a vicious cycle of endothelial injury, which also plays key roles in the formation of CAS. Therefore, in order to elucidate the mechanisms controlling CAS, it is important to study the etiology of vascular cell dysfunction, abnormal energy and metabolism disorders, and immune and inflammatory reactions. Non-coding RNAs play regulatory roles in the pathogenesis of CAS, especially long non-coding RNAs (lncRNAs); lncRNAs have recently become a major focus for cardiovascular disease mechanisms, as they play numerous roles in the progression of CAS. Therefore, in this review, we discuss the role of lncRNAs in the pathogenesis of coronary CAS, and their role in the prevention and treatment of coronary CAS.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xianjing Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
15
|
Peng B, Liu A, Yu X, Xu E, Dai J, Li M, Yang Q. Silencing of lncRNA AFAP1-AS1 suppressed lung cancer development by regulatory mechanism in cis and trans. Oncotarget 2017; 8:93608-93623. [PMID: 29212176 PMCID: PMC5706822 DOI: 10.18632/oncotarget.20549] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/04/2017] [Indexed: 12/18/2022] Open
Abstract
Although the long noncoding RNA AFAP1-AS1 has been shown to be involved in various types of cancer, its involvement in lung cancer remains poorly understood. In the current study, we found that AFAP1-AS1 was substantially over expressed in lung cancer tissues and cell lines. In addition, AFAP1-AS1 expression level was proven to be associated with the malignant features of lung cancer. Knockdown of AFAP1-AS1 significantly suppressed cell proliferation by increasing cell apoptosis and G0/G1 phase retardation of cell cycle in lung cancer cells. Furthermore, AFAP1-AS1 knockdown could suppress tumor growth of lung cancer in BALB/c nude mice. We also identified that AFAP1-AS1 silencing could influence the expression of AFAP1 and KRT1 on mRNA and protein level by cis and trans regulatory mechanism. Moreover, the oncogenic activities of AFAP1-AS1 on cell proliferation are partially mediated by KRT1. In summary, these findings demonstrate that AFAP1-AS1 plays an essential role in promoting lung cancer development in vitro and vivo. It indicated that AFAP1-AS1 is a promising prognostic predictor for patients with lung cancer.
Collapse
Affiliation(s)
- Baoying Peng
- The State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Yuexiu District, Guangzhou 510120, PR China.,The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| | - Anfei Liu
- The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| | - Xuanwei Yu
- The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| | - Enwu Xu
- Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China
| | - Jiabin Dai
- The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| | - Mengcheng Li
- The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| | - Qiaoyuan Yang
- The State Key Lab of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Yuexiu District, Guangzhou 510120, PR China.,The Institute for Chemical Carcinogenesis, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou 511436, China
| |
Collapse
|
16
|
Zhang XF, Tu R, Li K, Ye P, Cui X. Tumor Suppressor PTPRJ Is a Target of miR-155 in Colorectal Cancer. J Cell Biochem 2017; 118:3391-3400. [PMID: 28316102 DOI: 10.1002/jcb.25995] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/16/2017] [Indexed: 01/02/2023]
Abstract
PTPRJ is known for its antiproliferative role. Loss of heterozygosity (LOH) of PTPRJ has frequently been observed in various human cancers including colorectal cancer (CRC), lung cancer, and breast cancer. However, the function and mechanism of PTPRJ in CRC are not well understood. At the present study, we show that ectopic expression of PTPRJ inhibits cell growth, migration, and invasiveness in CRC cell line HCT116. Moreover, PTPRJ inhibits the tumorigenecity of HCT116 in a xenograft tumor model. MiR-155, the well-known oncomiR in CRC, is identified as an upstream factor of PTPRJ. MiR-155 directly binds to the 3' untranslated region of PTPRJ mRNA and suppresses the mRNA and protein levels of PTPRJ. Furthermore, the growth-promoting and AKT signaling activation effect of miR-155 was abrogated by PTPRJ overexpression, and vice versa. Our study reveals the crucial role of miR-155/PTPRJ/AKT axis in proliferation and migration of CRC cells and suggests a therapeutic potential of PTPRJ. J. Cell. Biochem. 118: 3391-3400, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiao-Fei Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Rongfu Tu
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Keke Li
- College of Life Sciences, Wuhan University, Wuhan 430070, P. R. China
| | - Pengxiang Ye
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| | - Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430072, P. R. China
| |
Collapse
|
17
|
Mumtaz PT, Bhat SA, Ahmad SM, Dar MA, Ahmed R, Urwat U, Ayaz A, Shrivastava D, Shah RA, Ganai NA. LncRNAs and immunity: watchdogs for host pathogen interactions. Biol Proced Online 2017; 19:3. [PMID: 28465674 PMCID: PMC5406993 DOI: 10.1186/s12575-017-0052-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 04/06/2017] [Indexed: 01/23/2023] Open
Abstract
Immune responses combat various infectious agents by inducing inflammatory responses, antimicrobial pathways and adaptive immunity. The polygenic responses to these external stimuli are temporally and coordinately regulated. Specific lncRNAs are induced to modulate innate and adaptive immune responses which can function through various target interactions like RNA-DNA, RNA-RNA, and RNA-protein interaction and hence affect the immunogenic regulation at various stages of gene expression. LncRNA are found to be present in various immune cells like monocytes, macrophages, dendritic cells, neutrophils, T cells and B cells. They have been shown to be involved in many biological processes, including the regulation of the expression of genes, the dosage compensation and genomics imprinting, but the knowledge how lncRNAs are regulated and how they alter cell differentiation/function is still obscure. Further dysregulation of lncRNA has been seen in many diseases, but as yet very less research has been carried out to understand the role of lncRNAs in regulation during host-pathogens interactions. In this review, we summarize the functional developments and mechanism of action of lncRNAs, in immunity and defense of host against pathogens.
Collapse
Affiliation(s)
- Peerzada Tajamul Mumtaz
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
- School of Life Sciences Jaipur National University, Jaipur, Rajasthan India
| | - Shakil Ahmad Bhat
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Syed Mudasir Ahmad
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Mashooq Ahmad Dar
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Raashid Ahmed
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Uneeb Urwat
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Aadil Ayaz
- Division of Animal Breeding and Genetics, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Divya Shrivastava
- School of Life Sciences Jaipur National University, Jaipur, Rajasthan India
| | - Riaz Ahmad Shah
- Division of Biotechnology, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| | - Nazir Ahmad Ganai
- Division of Animal Breeding and Genetics, Faculty of Veterinary Sciences and Animal Husbandry, SKUAST-K, Shuhama, Srinagar, Jammu and Kashmir India
| |
Collapse
|
18
|
Schneble N, Müller J, Kliche S, Bauer R, Wetzker R, Böhmer FD, Wang ZQ, Müller JP. The protein-tyrosine phosphatase DEP-1 promotes migration and phagocytic activity of microglial cells in part through negative regulation of fyn tyrosine kinase. Glia 2016; 65:416-428. [DOI: 10.1002/glia.23100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Nadine Schneble
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
- Leibniz Institute on Aging; Beutenberstraße 11 Jena Germany
| | - Julia Müller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
| | - Stefanie Kliche
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke-University; Leipziger Str. 44 Magdeburg Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
| | - Reinhard Wetzker
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
| | - Frank-D. Böhmer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging; Beutenberstraße 11 Jena Germany
| | - Jörg P. Müller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital; Hans-Knöll-Straße 2 Jena Germany
| |
Collapse
|
19
|
Li H, Zhu H, Ge J. Long Noncoding RNA: Recent Updates in Atherosclerosis. Int J Biol Sci 2016; 12:898-910. [PMID: 27314829 PMCID: PMC4910607 DOI: 10.7150/ijbs.14430] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/20/2016] [Indexed: 12/27/2022] Open
Abstract
Long noncoding RNAs belong to a class of noncoding RNAs longer than 200 nucleotides with the epigenetic regulation potential. As a novel molecular regulator, lncRNAs are often dysregulated in various pathological conditions and display multiple functions in a wide range of biological processes. Given that recent studies have indicated that lncRNAs are involved in atherosclerosis-related smooth muscle cell, endothelial cell, macrophage and lipid metabolism regulation, it is pertinent to understand the potential function of lncRNAs in atherosclerosis development. This review will highlight the recent updates of lncRNAs in atherogenesis and also discuss their potential roles as novel therapeutic targets.
Collapse
Affiliation(s)
- Hao Li
- 1. Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongming Zhu
- 2. Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Junbo Ge
- 1. Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.; 3. Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Murphy MB, Medvedev AE. Long noncoding RNAs as regulators of Toll-like receptor signaling and innate immunity. J Leukoc Biol 2016; 99:839-50. [PMID: 26965636 PMCID: PMC6608019 DOI: 10.1189/jlb.2ru1215-575r] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/18/2016] [Accepted: 02/23/2016] [Indexed: 12/30/2022] Open
Abstract
Sensing of microbial pathogens and endogenous "alarmins" by macrophages and dendritic cells is reliant on pattern recognition receptors, including membrane-associated TLRs, cytosolic nucleotide-binding and oligomerization domain leucine-rich repeat-containing receptors, retinoic acid-inducible gene I-like receptors, and absent in melanoma 2-like receptors. Engagement of TLRs elicits signaling pathways that activate inflammatory genes whose expression is regulated by chromatin-modifying complexes and transcription factors. Long noncoding RNAs have emerged as new regulators of inflammatory mediators in the immune system. They are expressed in macrophages, dendritic cells, neutrophils, NK cells, and T- and B-lymphocytes and are involved in immune cell differentiation and activation. Long noncoding RNAs act via repression or activation of transcription factors, modulation of stability of mRNA and microRNA, regulation of ribosome entry and translation of mRNAs, and controlling components of the epigenetic machinery. In this review, we focus on recent advances in deciphering the mechanisms by which long noncoding RNAs regulate TLR-driven responses in macrophages and dendritic cells and discuss the involvement of long noncoding RNAs in endotoxin tolerance, autoimmune, and inflammatory diseases. The dissection of the role of long noncoding RNAs will improve our understanding of the mechanisms of regulation of inflammation and may provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Michael B Murphy
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Andrei E Medvedev
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
21
|
Jain S, Thakkar N, Chhatai J, Pal Bhadra M, Bhadra U. Long non-coding RNA: Functional agent for disease traits. RNA Biol 2016; 14:522-535. [PMID: 27229269 DOI: 10.1080/15476286.2016.1172756] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In recent years, long non-coding RNAs (lncRNAs) have attracted the attention of researchers with their involvement in all facets of life. LncRNAs are transcripts of more than 200 nucleotides which lack defined protein coding potential. Although they do not code for proteins, a large number of them are involved in regulating gene expression and translation. The presence of numerous lncRNAs in the human genome has prompted us to investigate the contribution of these molecules to human biology and medicine. In this review, we present the potential role of lncRNAs interlinked to different human diseases and genetic disorders. We also describe their role in cellular differentiation and aging and discuss their potential importance as biomarkers and as therapeutic agents.
Collapse
Affiliation(s)
- Sriyans Jain
- a Functional Genomics and Gene Silencing Group , CSIR- Center for Cellular and Molecular Biology , Hyderabad , India
| | - Nirav Thakkar
- a Functional Genomics and Gene Silencing Group , CSIR- Center for Cellular and Molecular Biology , Hyderabad , India
| | - Jagamohan Chhatai
- a Functional Genomics and Gene Silencing Group , CSIR- Center for Cellular and Molecular Biology , Hyderabad , India
| | - Manika Pal Bhadra
- b Centre for Chemical Biology , Indian Institute for Chemical Technology , Hyderabad , India
| | - Utpal Bhadra
- a Functional Genomics and Gene Silencing Group , CSIR- Center for Cellular and Molecular Biology , Hyderabad , India
| |
Collapse
|
22
|
Effects of Guchang Capsule on Dextran Sulphate Sodium-Induced Experimental Ulcerative Colitis in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3150651. [PMID: 27313642 PMCID: PMC4894999 DOI: 10.1155/2016/3150651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/01/2016] [Accepted: 05/05/2016] [Indexed: 12/22/2022]
Abstract
Guchang capsule (GC) is a Chinese materia medica standardized product extracted from 15 Chinese traditional medical herbs and it has been clinically used in the treatment of intestinal disease. In this study, in order to extend the research of GC in intestinal disease, we were aiming to evaluate potential effects of GC on dextran sulphate sodium- (DSS-) induced murine experimental colitis and to elucidate the underlying mechanisms. GC treatment attenuated DSS-induced body weight loss and reduced the mortality. Moreover, GC treatment prevented DSS-induced colonic pathological damage; meanwhile it inhibited proinflammatory cytokines production in colon tissues. In vitro, GC significantly reduced LPS-induced proinflammatory cytokines production via inhibiting the activation of NF-κB in macrophage cells, and the expressions of several long noncoding RNAs (lncRNAs) which were reported in regulating NF-κB signaling pathway were obviously affected by adding GC into culture medium. In conclusion, our data suggested that administration of GC exhibits therapeutic effects on DSS-induced colitis partially through regulating the expression of NF-κB related lncRNAs in infiltrating immune cells.
Collapse
|
23
|
Zhou T, Ding JW, Wang XA, Zheng XX. Long noncoding RNAs and atherosclerosis. Atherosclerosis 2016; 248:51-61. [PMID: 26987066 DOI: 10.1016/j.atherosclerosis.2016.02.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is universally recognized as a chronic lipid-induced inflammation of the vessel wall in response to dyslipidemia and haemodynamic stress involving dysfunction and activation of resident vascular cells as well as infiltration of leukocytes. As members of nonprotein-coding RNAs, the long noncoding RNAs (lncRNAs) are implicated in various biological processes. Accumulating evidences suggest that lncRNAs regulate the function of vascular wall, activation of macrophages, lipid metabolism and immune response. Here, we review the effects of lncRNAs on the progress of atherosclerosis.
Collapse
Affiliation(s)
- Tian Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Jia-wang Ding
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China.
| | - Xin-an Wang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| | - Xia-xia Zheng
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, Hubei Province, China; Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, Hubei Province, China
| |
Collapse
|
24
|
Abstract
Precise and dynamic regulation of gene expression is a key feature of immunity. In recent years, rapid advances in transcriptome profiling analysis have led to recognize long non-coding RNAs (lncRNAs) as an additional layer of gene regulation context. In the immune system, lncRNAs are found to be widely expressed in immune cells including monocytes, macrophages, dendritic cells (DC), neutrophils, T cells and B cells during their development, differentiation and activation. However, the functional importance of immune-related lncRNAs is just emerging to be characterized. In this review, we discuss the up-to-date knowledge of lncRNAs in immune regulation.
Collapse
Affiliation(s)
- Hua Geng
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xiao-Di Tan
- Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Corresponding author. Center for Intestinal and Liver Inflammation Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Avenue, Box 217, Chicago, IL 60611, USA. Tel.: +1 (773) 755 6380; fax: +1 (773) 755 6581.
| |
Collapse
|
25
|
Transcriptome Analysis of Long Noncoding RNAs in Toll-Like Receptor 3-Activated Mesenchymal Stem Cells. Stem Cells Int 2015; 2016:6205485. [PMID: 26681952 PMCID: PMC4670881 DOI: 10.1155/2016/6205485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/21/2015] [Accepted: 06/11/2015] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess great immunomodulatory capacity which lays the foundation for their therapeutic effects in a variety of diseases. Recently, toll-like receptors (TLR) have been shown to modulate MSC functions; however, the underlying molecular mechanisms are poorly understood. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are an important class of regulators involved in a wide range of biological processes. To explore the potential involvement of lncRNAs in TLR stimulated MSCs, we performed a comprehensive lncRNA and mRNA profiling through microarray. 10.2% of lncRNAs (1733 out of 16967) and 15.1% of mRNA transcripts (1760 out of 11632) were significantly differentially expressed (absolute fold-change ≥5
, P value ≤0.05) in TLR3 stimulated MSCs. Furthermore, we characterized the differentially expressed lncRNAs through their classes and length distribution and correlated them with differentially expressed mRNA. Here, we are the first to determine genome-wide lncRNAs expression patterns in TLR3 stimulated MSCs by microarray and this work could provide a comprehensive framework of the transcriptome landscapes of TLR3 stimulated MSCs.
Collapse
|
26
|
Marques-Rocha JL, Samblas M, Milagro FI, Bressan J, Martínez JA, Marti A. Noncoding RNAs, cytokines, and inflammation-related diseases. FASEB J 2015; 29:3595-611. [PMID: 26065857 DOI: 10.1096/fj.14-260323] [Citation(s) in RCA: 360] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 06/02/2015] [Indexed: 12/15/2022]
Abstract
Chronic inflammation is involved in the onset and development of many diseases, including obesity, atherosclerosis, type 2 diabetes, osteoarthritis, autoimmune and degenerative diseases, asthma, periodontitis, and cirrhosis. The inflammation process is mediated by chemokines, cytokines, and different inflammatory cells. Although the molecules and mechanisms that regulate this primary defense mechanism are not fully understood, recent findings offer a putative role of noncoding RNAs, especially microRNAs (miRNAs), in the progression and management of the inflammatory response. These noncoding RNAs are crucial for the stability and maintenance of gene expression patterns that characterize some cell types, tissues, and biologic responses. Several miRNAs, such as miR-126, miR-132, miR-146, miR-155, and miR-221, have emerged as important transcriptional regulators of some inflammation-related mediators. Additionally, little is known about the involvement of long noncoding RNAs, long intergenic noncoding RNAs, and circular RNAs in inflammation-mediated processes and the homeostatic imbalance associated with metabolic disorders. These noncoding RNAs are emerging as biomarkers with diagnosis value, in prognosis protocols, or in the personalized treatment of inflammation-related alterations. In this context, this review summarizes findings in the field, highlighting those noncoding RNAs that regulate inflammation, with emphasis on recognized mediators such as TNF-α, IL-1, IL-6, IL-18, intercellular adhesion molecule 1, VCAM-1, and plasminogen activator inhibitor 1. The down-regulation or antagonism of the noncoding RNAs and the administration of exogenous miRNAs could be, in the near future, a promising therapeutic strategy in the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- José Luiz Marques-Rocha
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Mirian Samblas
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Fermin I Milagro
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Josefina Bressan
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - J Alfredo Martínez
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Amelia Marti
- *Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Nutrition, Food Science, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Physiopathology of Obesity, Carlos III Institute, Madrid, Spain; and Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
27
|
The Emerging Functions of Long Noncoding RNA in Immune Cells: Autoimmune Diseases. J Immunol Res 2015; 2015:848790. [PMID: 26090502 PMCID: PMC4451983 DOI: 10.1155/2015/848790] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/19/2015] [Indexed: 12/20/2022] Open
Abstract
The long noncoding RNAs (lncRNAs) are RNA transcripts more than 200 nucleotides in length, which do not encode proteins. The lncRNAs are emerging as an important regulator of biological process, such as chromatin remodeling, gene transcription, protein transport, and trafficking through diverse mechanisms. The lncRNAs play crucial role in various multigenetics human diseases including cancers and neurological diseases and currently its role in autoimmune diseases is attracting many researchers. Recent studies have reported that differentiation and activation of immune cells, T cells, B cells, macrophages, and NK cells have correlation with lncRNAs, which have also an essential role in autoimmune diseases such as rheumatoid arthritis and SLE. Therefore, elucidation of the roles of lncRNAs in autoimmunity could be beneficial to understand the pathogenesis of autoimmune diseases. In this review article we attempt to highlight the recent progress regarding lncRNAs studies and summarize its role in autoimmune diseases.
Collapse
|
28
|
Mao AP, Shen J, Zuo Z. Expression and regulation of long noncoding RNAs in TLR4 signaling in mouse macrophages. BMC Genomics 2015; 16:45. [PMID: 25652569 PMCID: PMC4320810 DOI: 10.1186/s12864-015-1270-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 01/22/2015] [Indexed: 12/20/2022] Open
Abstract
Background Though long non-coding RNAs (lncRNAs) are emerging as critical regulators of immune responses, whether they are involved in LPS-activated TLR4 signaling pathway and how is their expression regulated in mouse macrophages are still unexplored. Results By repurposing expression microarray probes, we identified 994 lncRNAs in bone marrow-derived macrophages (BMDMs) and classified them to enhancer-like lncRNAs (elncRNAs) and promoter-associated lncRNAs (plncRNAs) according to chromatin signatures defined by relative levels of H3K4me1 and H3K4me3. Fifteen elncRNAs and 12 plncRNAs are differentially expressed upon LPS stimulation. The expression change of lncRNAs and their neighboring protein-coding genes are significantly correlated. Also, the regulation of both elncRNAs and plncRNAs expression is associated with H3K4me3 and H3K27Ac. Crucially, many identified LPS-regulated lncRNAs, such as lncRNA-Nfkb2 and lncRNA-Rel, locate near to immune response protein-coding genes. The majority of LPS-regulated lncRNAs had at least one binding site among the transcription factors p65, IRF3, JunB and cJun. Conclusions We established an integrative microarray analysis pipeline for profiling lncRNAs. Also, our results suggest that lncRNAs can be important regulators of LPS-induced innate immune response in BMDMs. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1270-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ai-Ping Mao
- Department of Pathology, Committee on Immunology, University of Chicago, Chicago, Illinois, the United States.
| | - Jun Shen
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao-Tong University, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai, China.
| | - Zhixiang Zuo
- Department of Medicine, University of Chicago, 900 East 57th street, Chicago, IL, 60637, USA.
| |
Collapse
|
29
|
Regulated expression of PTPRJ by COX-2/PGE2 axis in endothelial cells. PLoS One 2014; 9:e114996. [PMID: 25532119 PMCID: PMC4274085 DOI: 10.1371/journal.pone.0114996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 11/17/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This study was designed to examine a novel role of COX-2/PGE2 signaling as a regulator of PTPRJ expression in endothelial cells. METHODS A bioinformatics analysis of a whole genome array was carried out to search for regulators of PTPRJ expression in endothelial cells. PTPRJ expression was also measured in endothelial cells derived from a balloon injury-induced neointimal hyperplasia model in male New Zealand Rabbits. Changes in PTPRJ expression in HUVEC cells was examined by RT-PCR and western blotting after transfection of COX-2 plasmids or treatment with varying concentrations of a COX-2 inhibitor. RESULTS A significant correlation was identified between COX-2 and PTPRJ in GSE39264 (Pearson correlation coefficient = -0.87; n = 22; P < 0.01, two-tailed). PTPRJ expression was reduced during the progression of neointimal hyperplasia after balloon injury, which correlated with an increase in COX-2 expression. In HUVECs, after transfection with 1 µg/ml, 0.5 µg/ml, or 0.25 µg/ml COX-2 plasmids, PTPRJ protein expression was reduced to 0.60- (± 0.08), 0.75- (± 0.09), and 0.88- (± 0.04) fold, respectively, while mRNA expression was reduced to 0.15- (± 0.03), 0.26- (± 0.05), and 0.47- (± 0.09) fold, respectively. After treatment of HUVECs with 10 µmol/L or 20 µmol/L celecoxib, the reduction in PTPRJ expression induced by COX-2 over-expression was not only rescued but in fact increased by 2.05-fold (± 0.28) and 3.34-fold (± 0.37), respectively, compared with control. CONCLUSIONS Our results suggest that COX-2/PGE2 signaling may function as a negative regulator of PTPRJ expression in endothelial cells both in vivo and vitro.
Collapse
|
30
|
Heward JA, Lindsay MA. Long non-coding RNAs in the regulation of the immune response. Trends Immunol 2014; 35:408-19. [PMID: 25113636 PMCID: PMC7106471 DOI: 10.1016/j.it.2014.07.005] [Citation(s) in RCA: 326] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 07/13/2014] [Accepted: 07/16/2014] [Indexed: 11/23/2022]
Abstract
Widespread changes in lncRNA expresssion are associated with the immune response. lncRNAs regulate the inflammatory response following activation of innate immunity. lncRNAs regulate T cell differentiation and migration. The action of long non-coding RNAs is mediated via diverse mechanisms.
It is increasingly clear that long non-coding RNAs (lncRNAs) regulate a variety biological responses, and that they do so by a diverse range of mechanisms. In the field of immunology, recent publications have shown widespread changes in the expression of lncRNAs during the activation of the innate immune response and T cell development, differentiation, and activation. These lncRNAs control important aspects of immunity such as production of inflammatory mediators, differentiation, and cell migration through regulating protein–protein interactions or via their ability to basepair with RNA and DNA. We review the current understanding of the mechanism of action of these immune-related lncRNAs, discuss their impact on physiological and pathological processes, and highlight important areas of inquiry at the intersection between immunology and lncRNA biology.
Collapse
Affiliation(s)
- James A Heward
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Mark A Lindsay
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, UK.
| |
Collapse
|
31
|
Wang Y, Pang WJ, Wei N, Xiong Y, Wu WJ, Zhao CZ, Shen QW, Yang GS. Identification, stability and expression of Sirt1 antisense long non-coding RNA. Gene 2014; 539:117-24. [PMID: 24480449 DOI: 10.1016/j.gene.2014.01.037] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/31/2013] [Accepted: 01/10/2014] [Indexed: 01/23/2023]
Abstract
Natural antisense transcripts (NATs) exist ubiquitously as pivotal molecules to regulate coding gene expression. Sirtuin 1 (Sirt1) is a NAD-dependent deacetylase which is involved in myogenesis. However, whether Sirt1 transcribes NAT during C2C12 differentiation is still unknown. In this study, we identified a Sirt1 NAT which was designated as Sirt1 antisense long non-coding RNA (AS lncRNA) by sequencing and bioinformatic analysis. The level of Sirt1 AS lncRNA was greater in spleen but less in muscle tissue. The expression of both Sirt1 mRNA and Sirt1 AS lncRNA decreased during C2C12 myogenic differentiation, whereas the levels of miR-34a, which targets Sirt1, increased gradually. We further found that the half-life of Sirt1 AS lncRNA was 10h, but that of Sirt1 mRNA was 6h in C2C12 cells treated with 2 μg/ml Actinomycin D. Therefore, compared with Sirt1 mRNA, Sirt1 AS lncRNA was more stable. Overexpression of Sirt1 AS lncRNA increased the levels of Sirt1 protein, whereas overexpression of Sirt1 AS lncRNA mutant did not affect the level of Sirt1 protein in C2C12 cells. Moreover, downregulation of Sirt1 mRNA caused by miR-34a was counteracted by Sirt1 AS lncRNA in C2C12 cells. Taken together, we identified a novel NAT of Sirt1 which implicated in myogenesis through regulating Sirt1 expression.
Collapse
Affiliation(s)
- Yu Wang
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China.
| | - Wei-Jun Pang
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China.
| | - Ning Wei
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| | - Yan Xiong
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| | - Wen-Jing Wu
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| | - Cun-Zhen Zhao
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| | - Qing-Wu Shen
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| | - Gong-She Yang
- College of Animal Science and Technology, Northwest A&F University, Laboratory of Animal Fat Deposition & Muscle Development, Yangling Shaanxi 712100, China
| |
Collapse
|