1
|
Tiwari A, Hobbs BD, Sharma R, Li J, Kho AT, Amr S, Celedón JC, Weiss ST, Hersh CP, Tantisira KG, McGeachie MJ. Peripheral blood miRNAs are associated with airflow below threshold in children with asthma. Respir Res 2025; 26:38. [PMID: 39856653 PMCID: PMC11763123 DOI: 10.1186/s12931-025-03116-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are crucial post-transcriptional regulators involved in inflammatory diseases, such as asthma. Poor lung function and airflow issues in childhood are linked to the development of chronic obstructive pulmonary disease (COPD) in adulthood. METHODS We analyzed small RNA-Seq data from 365 peripheral whole blood samples from the Genetics of Asthma in Costa Rica Study (GACRS) for association with airflow levels measured by FEV1/FVC. Differentially expressed (DE) miRNAs were identified using DESeq2 in R, adjusting for covariates and applying a 10% false discovery rate (FDR). The analysis included 361 samples and 649 miRNAs. The two DE miRNAs were further tested for association with airflow obstruction in a study of adult former smokers with and without COPD. RESULTS We found 1 upregulated and 1 downregulated miRNA in participants with airflow below the threshold compared to those above it. In the adult study, the same miRNAs were upregulated and downregulated in individuals with FEV1/FVC < 0.7 versus those with FEV1/FVC > 0.7, showing suggestive statistical evidence. The target genes of these miRNAs were enriched for PI3K-Akt, Hippo, WNT, MAPK, and focal adhesion pathways. CONCLUSIONS Two differentially expressed miRNAs were associated with airflow levels in children with asthma and airflow obstruction in adults with COPD. This suggests that shared genetic regulatory systems may influence childhood airflow and contribute to adulthood airflow obstruction.
Collapse
Affiliation(s)
- Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Brian D Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rinku Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiang Li
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alvin T Kho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Sami Amr
- Translational Genomics Core, Mass General Brigham Personalized Medicine, Cambridge, MA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Channing Division of Network Medicine, Harvard Medical School, 181 Longwood Avenue, Room 539, Boston, MA, 02115, USA.
| |
Collapse
|
2
|
Taylor HJ, Hung YH, Narisu N, Erdos MR, Kanke M, Yan T, Grenko CM, Swift AJ, Bonnycastle LL, Sethupathy P, Collins FS, Taylor DL. Human pancreatic islet microRNAs implicated in diabetes and related traits by large-scale genetic analysis. Proc Natl Acad Sci U S A 2023; 120:e2206797120. [PMID: 36757889 PMCID: PMC9963967 DOI: 10.1073/pnas.2206797120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Genetic studies have identified ≥240 loci associated with the risk of type 2 diabetes (T2D), yet most of these loci lie in non-coding regions, masking the underlying molecular mechanisms. Recent studies investigating mRNA expression in human pancreatic islets have yielded important insights into the molecular drivers of normal islet function and T2D pathophysiology. However, similar studies investigating microRNA (miRNA) expression remain limited. Here, we present data from 63 individuals, the largest sequencing-based analysis of miRNA expression in human islets to date. We characterized the genetic regulation of miRNA expression by decomposing the expression of highly heritable miRNAs into cis- and trans-acting genetic components and mapping cis-acting loci associated with miRNA expression [miRNA-expression quantitative trait loci (eQTLs)]. We found i) 84 heritable miRNAs, primarily regulated by trans-acting genetic effects, and ii) 5 miRNA-eQTLs. We also used several different strategies to identify T2D-associated miRNAs. First, we colocalized miRNA-eQTLs with genetic loci associated with T2D and multiple glycemic traits, identifying one miRNA, miR-1908, that shares genetic signals for blood glucose and glycated hemoglobin (HbA1c). Next, we intersected miRNA seed regions and predicted target sites with credible set SNPs associated with T2D and glycemic traits and found 32 miRNAs that may have altered binding and function due to disrupted seed regions. Finally, we performed differential expression analysis and identified 14 miRNAs associated with T2D status-including miR-187-3p, miR-21-5p, miR-668, and miR-199b-5p-and 4 miRNAs associated with a polygenic score for HbA1c levels-miR-216a, miR-25, miR-30a-3p, and miR-30a-5p.
Collapse
Affiliation(s)
- Henry J. Taylor
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, CambridgeCB2 0BB, UK
- Heart and Lung Research Institute, University of Cambridge, CambridgeCB2 0BB, UK
| | - Yu-Han Hung
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Michael R. Erdos
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Matthew Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Caleb M. Grenko
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Amy J. Swift
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Lori L. Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Francis S. Collins
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - D. Leland Taylor
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| |
Collapse
|
3
|
Jakwerth CA, Kitzberger H, Pogorelov D, Müller A, Blank S, Schmidt-Weber CB, Zissler UM. Role of microRNAs in type 2 diseases and allergen-specific immunotherapy. FRONTIERS IN ALLERGY 2022; 3:993937. [PMID: 36172292 PMCID: PMC9512106 DOI: 10.3389/falgy.2022.993937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 12/07/2022] Open
Abstract
MicroRNAs (miRs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases as well as their potential as biomarkers in allergen-specific treatment options. Their function as post-transcriptional regulators, controlling various cellular processes, is of high importance since any single miR can target multiple mRNAs, often within the same signalling pathway. MiRs can alter dysregulated expression of certain cellular responses and contribute to or cause, but in some cases prevent or repress, the development of various diseases. In this review article, we describe current research on the role of specific miRs in regulating immune responses in epithelial cells and specialized immune cells in response to various stimuli, in allergic diseases, and regulation in the therapeutic approach of allergen-specific immunotherapy (AIT). Despite the fact that AIT has been used successfully as a causative treatment option since more than a century, very little is known about the mechanisms of regulation and its connections with microRNAs. In order to fill this gap, this review aims to provide an overview of the current knowledge.
Collapse
|
4
|
Wang C, Wang J, Zheng X, Zhang J, Zhang J, Qiao G, Liu H, Zhao H, Bai J, Zhang H, Zhang Z. Epigenetic regulation is involved in traffic-related PM 2.5 aggravating allergic airway inflammation in rats. Clin Immunol 2021; 234:108914. [PMID: 34954131 DOI: 10.1016/j.clim.2021.108914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/27/2021] [Accepted: 12/20/2021] [Indexed: 11/29/2022]
Abstract
Increasing fine particulate matter (PM2.5) and epigenetic modifications are closely associated with the pathogenesis of asthma, but the definite mechanism remains unclear. The traffic-related PM2.5 exposure aggravated pulmonary inflammation and changed the methylation level of interferon gamma (Ifng) and interleukin (Il)4 genes, and then altered levels of affiliated cytokines of IFN-γ and IL-4 in rats with allergic airway inflammation. It also increased the level of miR146a and decreased the level of miR31. In addition, transcription factors of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 6 (Stat6) rose; forkhead box P3 (Foxp3) and signal transducer and activator of transcription 4 (Stat4) lowered. The traffic-related PM2.5 altered epigenetic modifications in allergic airway inflammation of rats leading to inflammation exacerbation through impaired regulatory T (Treg) cells function and T-helper type 1 (Th1)/Th2 cells imbalance, which provided a new target for the treatment and control of asthma.
Collapse
Affiliation(s)
- Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jing Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Xin Zheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jiaqi Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jingwei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Guoguo Qiao
- Teaching Experiment Center, School of Public Health, Shanxi Medical University, China
| | - Haifang Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Huichao Zhao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China.
| |
Collapse
|
5
|
Quan R, Liang W, Li H, Ning Q, Shang D. Silencing of miR-10b-5p alleviates the mechanical stretch-induced proliferation of HASMCs. Tissue Cell 2021; 74:101700. [PMID: 34871825 DOI: 10.1016/j.tice.2021.101700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 11/25/2022]
Abstract
MicroRNAs (miRNAs) are important mediators to human airway smooth muscle cells (HASMCs) phenotype remodeling and airway diseases. MicroRNA-10b-5p (miR-10b-5p) has been extensively studied in different fields. This study set out to probe into the effect of miR-10b-5p in cyclic mechanical stretch-induced apoptosis in HASMCs. The results showed that after 15 % deformation, 0.5 s stretching and 0.5 s cyclic mechanical stretching relaxation (0.5 Hz) occurred to HASMCs, miR-10b-5p showed up-regulation without inducing significant apoptosis. Moreover, the mRNA and protein expressions of FLT1 were reduced. Then, dual-luciferase reporter assay verified that FLT1 was targeted by miR-10b-5p, and miR-10b-5p silencing increased FLT1 expression, leading to a prolonged arrest of stretch-treated HASMCs at the G1/S stage, and increased cell apoptosis compared with control group. Furthermore, the activity of Caspase-3 was reinforced, and the ratio of Bcl-2 to Bax was markedly reduced after miR-10b-5p silencing. The current study proved that expression levels of p-PI3K and p-Akt in stretch-treated HASMCs of the inhibition group were significantly inhibited in comparison to those of the controls. The effects of miR-10b-5p overexpression are opposite to that of inhibition of miR-10b-5p in stretched HASMCs. In conclusion, this study showed that miR-10b-5p silencing could weaken the hypertrophy of HASMCs. MiR-10b-5p negatively regulated FLT1 expression, but positively regulated the PI3K/Akt pathway in HASMCs. By referring to other previous studies, we concluded that miR-10b-5p might be a potent target in the treatment of airway diseases.
Collapse
Affiliation(s)
- Rongxi Quan
- Department of Intensive Care Unit, Affiliated Tumor Hospital of Xinjiang Medical University, China
| | - Wei Liang
- Department of Intensive Care Unit, Affiliated Tumor Hospital of Xinjiang Medical University, China
| | - Hong Li
- Department of Respiration, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qian Ning
- Department of Respiration, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Dong Shang
- Department of Intensive Care Unit, Affiliated Tumor Hospital of Xinjiang Medical University, China; Department of Respiration, The First Affiliated Hospital of Xi'an Jiaotong University, China.
| |
Collapse
|
6
|
Effects of single bouts of different endurance exercises with different intensities on microRNA biomarkers with and without blood flow restriction: a three-arm, randomized crossover trial. Eur J Appl Physiol 2021; 121:3243-3255. [PMID: 34435273 PMCID: PMC8505326 DOI: 10.1007/s00421-021-04786-2] [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: 01/27/2021] [Accepted: 08/10/2021] [Indexed: 10/27/2022]
Abstract
PURPOSE Physical activity is associated with altered levels of circulating microRNAs (ci-miRNAs). Changes in miRNA expression have great potential to modulate biological pathways of skeletal muscle hypertrophy and metabolism. This study was designed to determine whether the profile of ci-miRNAs is altered after different approaches of endurance exercise. METHODS Eighteen healthy volunteers (aged 24 ± 3 years) participated this three-arm, randomized-balanced crossover study. Each arm was a single bout of treadmill-based acute endurance exercise at (1) 100% of the individual anaerobic threshold (IANS), (2) at 80% of the IANS and (3) at 80% of the IANS with blood flow restriction (BFR). Load-associated outcomes (fatigue, feeling, heart rate, and exhaustion) as well as acute effects (circulating miRNA patterns and lactate) were determined. RESULTS All training interventions increased the lactate concentration (LC) and heart rate (HR) (p < 0.001). The high-intensity intervention (HI) resulted in a higher LC than both lower intensity protocols (p < 0.001). The low-intensity blood flow restriction (LI-BFR) protocol led to a higher HR and higher LC than the low-intensity (LI) protocol without BFR (p = 0.037 and p = 0.003). The level of miR-142-5p and miR-197-3p were up-regulated in both interventions without BFR (p < 0.05). After LI exercise, the expression of miR-342-3p was up-regulated (p = 0.038). In LI-BFR, the level of miR-342-3p and miR-424-5p was confirmed to be up-regulated (p < 0.05). Three miRNAs and LC show a significant negative correlation (miR-99a-5p, p = 0.011, r = - 0.343/miR-199a-3p, p = 0.045, r = - 0.274/miR-125b-5p, p = 0.026, r = - 0.302). Two partial correlations (intervention partialized) showed a systematic impact of the type of exercise (LI-BFR vs. HI) (miR-99a-59: r = - 0.280/miR-199a-3p: r = - 0.293). CONCLUSION MiRNA expression patterns differ according to type of activity. We concluded that not only the intensity of the exercise (LC) is decisive for the release of circulating miRNAs-as essential is the type of training and the oxygen supply.
Collapse
|
7
|
Que E, James KL, Coffey AR, Smallwood TL, Albright J, Huda MN, Pomp D, Sethupathy P, Bennett BJ. Genetic architecture modulates diet-induced hepatic mRNA and miRNA expression profiles in Diversity Outbred mice. Genetics 2021; 218:6321522. [PMID: 34849860 PMCID: PMC8757298 DOI: 10.1093/genetics/iyab068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 07/27/2020] [Indexed: 11/30/2022] Open
Abstract
Genetic approaches in model organisms have consistently demonstrated that molecular traits such as gene expression are under genetic regulation, similar to clinical traits. The resulting expression quantitative trait loci (eQTL) have revolutionized our understanding of genetic regulation and identified numerous candidate genes for clinically relevant traits. More recently, these analyses have been extended to other molecular traits such as protein abundance, metabolite levels, and miRNA expression. Here, we performed global hepatic eQTL and microRNA expression quantitative trait loci (mirQTL) analysis in a population of Diversity Outbred mice fed two different diets. We identified several key features of eQTL and mirQTL, namely differences in the mode of genetic regulation (cis or trans) between mRNA and miRNA. Approximately 50% of mirQTL are regulated by a trans-acting factor, compared to ∼25% of eQTL. We note differences in the heritability of mRNA and miRNA expression and variance explained by each eQTL or mirQTL. In general, cis-acting variants affecting mRNA or miRNA expression explain more phenotypic variance than trans-acting variants. Finally, we investigated the effect of diet on the genetic architecture of eQTL and mirQTL, highlighting the critical effects of environment on both eQTL and mirQTL. Overall, these data underscore the complex genetic regulation of two well-characterized RNA classes (mRNA and miRNA) that have critical roles in the regulation of clinical traits and disease susceptibility
Collapse
Affiliation(s)
- Excel Que
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA 95616, USA.,Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Kristen L James
- Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 28081, USA
| | - Tangi L Smallwood
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 28081, USA
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - M Nazmul Huda
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA 95616, USA.,Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Daniel Pomp
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Brian J Bennett
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, CA 95616, USA.,Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
8
|
Kim M, Jo H, Kwon Y, Jeong MS, Jung HS, Kim Y, Jeoung D. MiR-154-5p-MCP1 Axis Regulates Allergic Inflammation by Mediating Cellular Interactions. Front Immunol 2021; 12:663726. [PMID: 34135893 PMCID: PMC8201518 DOI: 10.3389/fimmu.2021.663726] [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] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/17/2021] [Indexed: 12/25/2022] Open
Abstract
In a previous study, we have demonstrated that p62, a selective receptor of autophagy, can regulate allergic inflammation. In the present study, microRNA array analysis showed that miR-154-5p was increased by antigen (DNP-HSA) in a p62-dependent manner in rat basophilic leukemia cells (RBL2H3). NF-kB directly increased the expression of miR-154-5p. miR-154-5p mediated in vivo allergic reactions, including passive cutaneous anaphylaxis and passive systemic anaphylaxis. Cytokine array analysis showed that antigen stimulation increased the expression of MCP1 in RBL2H3 cells in an miR-154-5p-dependent manner. Reactive oxygen species (ROS)-ERK-NF-kB signaling increased the expression of MCP1 in antigen-stimulated RBL2H3 cells. Recombinant MCP1 protein induced molecular features of allergic reactions both in vitro and in vivo. Anaphylaxis-promoted tumorigenic potential has been known to be accompanied by cellular interactions involving mast cells, and macrophages, and cancer cells. Our experiments employing culture medium, co-cultures, and recombinant MCP1 protein showed that miR-154 and MCP1 mediated these cellular interactions. MiR-154-5p and MCP1 were found to be present in exosomes of RBL2H3 cells. Exosomes from PSA-activated BALB/C mouse induced molecular features of passive cutaneous anaphylaxis in an miR-154-5p-dependent manner. Exosomes from antigen-stimulated RBL2H3 cells enhanced both tumorigenic and metastatic potentials of B16F1 melanoma cells in an miR-154-5p-dependent manner. Exosomes regulated both ROS level and ROS mediated cellular interactions during allergic inflammation. Our results indicate that the miR-154-5p-MCP1 axis might serve as a valuable target for the development of anti-allergy therapeutics.
Collapse
Affiliation(s)
- Misun Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Hyein Jo
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Myeong Seon Jeong
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea.,Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Youngmi Kim
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
9
|
Li J, Tiwari A, Mirzakhani H, Wang AL, Kho AT, McGeachie MJ, Litonjua AA, Weiss ST, Tantisira KG. Circulating MicroRNA: Incident Asthma Prediction and Vitamin D Effect Modification. J Pers Med 2021; 11:307. [PMID: 33923455 PMCID: PMC8073146 DOI: 10.3390/jpm11040307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 12/23/2022] Open
Abstract
Of children with recurrent wheezing in early childhood, approximately half go on to develop asthma. MicroRNAs have been described as excellent non-invasive biomarkers due to their prognostic utility. We hypothesized that circulating microRNAs can predict incident asthma and that that prediction might be modified by vitamin D. We selected 75 participants with recurrent wheezing at 3 years old from the Vitamin D Antenatal Asthma Reduction Trial (VDAART). Plasma samples were collected at age 3 and sequenced for small RNA-Seq. The read counts were normalized and filtered by depth and coverage. Logistic regression was employed to associate miRNAs at age 3 with asthma status at age 5. While the overall effect of miRNA on asthma occurrence was weak, we identified 38 miRNAs with a significant interaction effect with vitamin D and 32 miRNAs with a significant main effect in the high vitamin D treatment group in VDAART. We validated the VDAART results in Project Viva for both the main effect and interaction effect. Meta-analysis was performed on both cohorts to obtain the combined effect and a logistic regression model was used to predict incident asthma at age 7 in Project Viva. Of the 23 overlapped miRNAs in the stratified and interaction analysis above, 9 miRNAs were replicated in Project Viva with strong effect size and remained in the meta-analysis of the two populations. The target genes of the 9 miRNAs were enriched for asthma-related Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways. Using logistic regression, microRNA hsa-miR-574-5p had a good prognostic ability for incident asthma prognosis with an area under the receiver operating characteristic (AUROC) of 0.83. In conclusion, miRNAs appear to be good biomarkers of incident asthma, but only when vitamin D level is considered.
Collapse
Affiliation(s)
- Jiang Li
- Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China;
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Alberta L. Wang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Alvin T. Kho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Michael J. McGeachie
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
| | - Kelan G. Tantisira
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (A.T.); (H.M.); (A.L.W.); (A.T.K.); (M.J.M.); (S.T.W.)
- Division of Pediatric Respiratory Medicine, Rady Children’s Hospital San Diego, University of California, San Diego, CA 92123, USA
| |
Collapse
|
10
|
Que E, James KL, Coffey AR, Smallwood TL, Albright J, Huda MN, Pomp D, Sethupathy P, Bennett BJ. Genetic Architecture Modulates Diet-Induced Hepatic mRNA and miRNA Expression Profiles in Diversity Outbred Mice. Genetics 2020; 216:241-259. [PMID: 32763908 PMCID: PMC7463293 DOI: 10.1534/genetics.120.303481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Genetic approaches in model organisms have consistently demonstrated that molecular traits such as gene expression are under genetic regulation, similar to clinical traits. The resulting expression quantitative trait loci (eQTL) have revolutionized our understanding of genetic regulation and identified numerous candidate genes for clinically relevant traits. More recently, these analyses have been extended to other molecular traits such as protein abundance, metabolite levels, and miRNA expression. Here, we performed global hepatic eQTL and microRNA expression quantitative trait loci (mirQTL) analysis in a population of Diversity Outbred mice fed two different diets. We identified several key features of eQTL and mirQTL, namely differences in the mode of genetic regulation (cis or trans) between mRNA and miRNA. Approximately 50% of mirQTL are regulated by a trans-acting factor, compared to ∼25% of eQTL. We note differences in the heritability of mRNA and miRNA expression and variance explained by each eQTL or mirQTL. In general, cis-acting variants affecting mRNA or miRNA expression explain more phenotypic variance than trans-acting variants. Lastly, we investigated the effect of diet on the genetic architecture of eQTL and mirQTL, highlighting the critical effects of environment on both eQTL and mirQTL. Overall, these data underscore the complex genetic regulation of two well-characterized RNA classes (mRNA and miRNA) that have critical roles in the regulation of clinical traits and disease susceptibility.
Collapse
Affiliation(s)
- Excel Que
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, California 95616
- Department of Nutrition, University of California, Davis, California
| | - Kristen L James
- Department of Nutrition, University of California, Davis, California
| | - Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina
| | - Tangi L Smallwood
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, North Carolina
| | - Jody Albright
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - M Nazmul Huda
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, California 95616
- Department of Nutrition, University of California, Davis, California
| | - Daniel Pomp
- Department of Genetics, University of North Carolina at Chapel Hill, North Carolina
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Brian J Bennett
- Western Human Nutrition Research Center, Agricultural Research Service, US Department of Agriculture, Davis, California 95616
- Department of Nutrition, University of California, Davis, California
| |
Collapse
|
11
|
Hosur V, Skelly DA, Francis C, Low BE, Kohar V, Burzenski LM, Amiji MM, Shultz LD, Wiles MV. Improved mouse models and advanced genetic and genomic technologies for the study of neutrophils. Drug Discov Today 2020; 25:1013-1025. [PMID: 32387410 DOI: 10.1016/j.drudis.2020.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022]
Abstract
Mice have been excellent surrogates for studying neutrophil biology and, furthermore, murine models of human disease have provided fundamental insights into the roles of human neutrophils in innate immunity. The emergence of novel humanized mice and high-diversity mouse populations offers the research community innovative and powerful platforms for better understanding, respectively, the mechanisms by which human neutrophils drive pathogenicity, and how genetic differences underpin the variation in neutrophil biology observed among humans. Here, we review key examples of these new resources. Additionally, we provide an overview of advanced genetic engineering tools available to further improve such murine model systems, of sophisticated neutrophil-profiling technologies, and of multifunctional nanoparticle (NP)-based neutrophil-targeting strategies.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA.
| | - Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Christopher Francis
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Benjamin E Low
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Vivek Kohar
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Lisa M Burzenski
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Leonard D Shultz
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| | - Michael V Wiles
- The Jackson Laboratory for Mammalian Genetics, 600 Main Street, Bar Harbor, ME 04609 USA
| |
Collapse
|
12
|
Qi C, Xu CJ, Koppelman GH. The role of epigenetics in the development of childhood asthma. Expert Rev Clin Immunol 2019; 15:1287-1302. [PMID: 31674254 DOI: 10.1080/1744666x.2020.1686977] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The development of childhood asthma is caused by a combination of genetic factors and environmental exposures. Epigenetics describes mechanisms of (heritable) regulation of gene expression that occur without changes in DNA sequence. Epigenetics is strongly related to aging, is cell-type specific, and includes DNA methylation, noncoding RNAs, and histone modifications.Areas covered: This review summarizes recent epigenetic studies of childhood asthma in humans, which mostly involve studies of DNA methylation published in the recent five years. Environmental exposures, in particular cigarette smoking, have significant impact on epigenetic changes, but few of these epigenetic signals are also associated with asthma. Several asthma-associated genetic variants relate to DNA methylation. Epigenetic signals can be better understood by studying their correlation with gene expression, which revealed higher presence and activation of blood eosinophils in asthma. Strong associations of nasal methylation signatures and atopic asthma were identified, which were replicable across different populations.Expert commentary: Epigenetic markers have been strongly associated with asthma, and might serve as biomarker of asthma. The causal and longitudinal relationships between epigenetics and disease, and between environmental exposures and epigenetic changes need to be further investigated. Efforts should be made to understand cell-type-specific epigenetic mechanisms in asthma.
Collapse
Affiliation(s)
- Cancan Qi
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cheng-Jian Xu
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Gastroenterology, Hepatology and Endocrinology, CiiM, Centre for individualised infection medicine, A joint venture between Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Gerard H Koppelman
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Brook AC, Jenkins RH, Clayton A, Kift-Morgan A, Raby AC, Shephard AP, Mariotti B, Cuff SM, Bazzoni F, Bowen T, Fraser DJ, Eberl M. Neutrophil-derived miR-223 as local biomarker of bacterial peritonitis. Sci Rep 2019; 9:10136. [PMID: 31300703 PMCID: PMC6625975 DOI: 10.1038/s41598-019-46585-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/01/2019] [Indexed: 02/08/2023] Open
Abstract
Infection remains a major cause of morbidity, mortality and technique failure in patients with end stage kidney failure who receive peritoneal dialysis (PD). Recent research suggests that the early inflammatory response at the site of infection carries diagnostically relevant information, suggesting that organ and pathogen-specific "immune fingerprints" may guide targeted treatment decisions and allow patient stratification and risk prediction at the point of care. Here, we recorded microRNA profiles in the PD effluent of patients presenting with symptoms of acute peritonitis and show that elevated peritoneal miR-223 and reduced miR-31 levels were useful predictors of bacterial infection. Cell culture experiments indicated that miR-223 was predominantly produced by infiltrating immune cells (neutrophils, monocytes), while miR-31 was mainly derived from the local tissue (mesothelial cells, fibroblasts). miR-223 was found to be functionally stabilised in PD effluent from peritonitis patients, with a proportion likely to be incorporated into neutrophil-derived exosomes. Our study demonstrates that microRNAs are useful biomarkers of bacterial infection in PD-related peritonitis and have the potential to contribute to disease-specific immune fingerprints. Exosome-encapsulated microRNAs may have a functional role in intercellular communication between immune cells responding to the infection and the local tissue, to help clear the infection, resolve the inflammation and restore homeostasis.
Collapse
Affiliation(s)
- Amy C Brook
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Robert H Jenkins
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, United Kingdom
| | - Aled Clayton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ann Kift-Morgan
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, United Kingdom
| | - Alex P Shephard
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Barbara Mariotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Simone M Cuff
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Flavia Bazzoni
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Timothy Bowen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, United Kingdom
| | - Donald J Fraser
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, United Kingdom
- Directorate of Nephrology and Transplantation, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom.
- Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
14
|
Kim M, Park Y, Kwon Y, Kim Y, Byun J, Jeong MS, Kim HU, Jung HS, Mun JY, Jeoung D. MiR-135-5p-p62 Axis Regulates Autophagic Flux, Tumorigenic Potential, and Cellular Interactions Mediated by Extracellular Vesicles During Allergic Inflammation. Front Immunol 2019; 10:738. [PMID: 31024564 PMCID: PMC6460569 DOI: 10.3389/fimmu.2019.00738] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/19/2019] [Indexed: 11/13/2022] Open
Abstract
The objective of this study was to investigate the relationship between autophagy and allergic inflammation. In vitro allergic inflammation was accompanied by an increased autophagic flux in rat basophilic leukemia (RBL2H3) cells. 3-MA, an inhibitor of autophagic processes, negatively regulated allergic inflammation both in vitro and in vivo. The role of p62, a selective receptor of autophagy, in allergic inflammation was investigated. P62, increased by antigen stimulation, mediated in vitro allergic inflammation, passive cutaneous anaphylaxis (PCA), and passive systemic anaphylaxis (PSA). P62 mediated cellular interactions during allergic inflammation. It also mediated tumorigenic and metastatic potential of cancer cells enhanced by PSA. TargetScan analysis predicted that miR-135-5p was a negative regulator of p62. Luciferase activity assay showed that miR-135-5p directly regulated p62. MiR-135-5p mimic negatively regulated features of allergic inflammation and inhibited tumorigenic and metastatic potential of cancer cells enhanced by PSA. MiR-135-5p mimic also inhibited cellular interactions during allergic inflammation. Extracellular vesicles mediated allergic inflammation both in vitro and in vivo. Extracellular vesicles were also necessary for cellular interactions during allergic inflammation. Transmission electron microscopy showed p62 within extracellular vesicles of antigen-stimulated rat basophilic leukemia cells (RBL2H3). Extracellular vesicles isolated from antigen-stimulated RBL2H3 cells induced activation of macrophages and enhanced invasion and migration potential of B16F1 mouse melanoma cells in a p62-dependent manner. Extracellular vesicles isolated from PSA-activated BALB/C mouse enhanced invasion and migration potential of B16F1 cells, and induced features of allergic inflammation in RBL2H3 cells. Thus, miR-135-5p-p62 axis might serve as a target for developing anti-allergy drugs.
Collapse
Affiliation(s)
- Misun Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Yeongseo Park
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Youngmi Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Jaehwan Byun
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Myeong Seon Jeong
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea.,Chuncheon Center, Korean Basic Science Institute, Chuncheon, South Korea
| | - Han-Ul Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Ji Young Mun
- Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
15
|
Gao Y, Wang B, Luo H, Zhang Q, Xu M. RETRACTED: miR-217 represses TGF-β1-induced airway smooth muscle cell proliferation and migration through targeting ZEB1. Biomed Pharmacother 2018; 108:27-35. [PMID: 30212709 DOI: 10.1016/j.biopha.2018.09.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 09/05/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. The journal was alerted to several suspected image similarities within Figures 2C and 3B, between Figure 3B and 5E, and an image in Figure 3A appears to be present in another publication, as detailed here: https://pubpeer.com/publications/F4E8CA0032EF5375E7867504F3FC4A. These findings were confirmed as part of an internal investigation, and in addition, a portion of Figure 2C, ‘Scramble’ group appears to contain image similarities with Figure 5D ‘miR-217 mimic + ZEB1’ group. The journal requested the authors provide explanations and source data relating to these affected figures, but the Authors did not respond to these concerns. The Editor-in-Chief assessed this case and decided to retract the article.
Collapse
Affiliation(s)
- Ying Gao
- Otolaryngology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Shaanxi, China
| | - Botao Wang
- Otolaryngology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Shaanxi, China
| | - Huanan Luo
- Otolaryngology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Shaanxi, China
| | - Qing Zhang
- Otolaryngology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Shaanxi, China
| | - Min Xu
- Otolaryngology Department, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Shaanxi, China.
| |
Collapse
|
16
|
Yue H, Yan W, Ji X, Zhang Y, Li G, Sang N. Maternal exposure to NO 2 enhances airway sensitivity to allergens in BALB/c mice through the JAK-STAT6 pathway. CHEMOSPHERE 2018; 200:455-463. [PMID: 29501036 DOI: 10.1016/j.chemosphere.2018.02.116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/04/2018] [Accepted: 02/19/2018] [Indexed: 06/08/2023]
Abstract
Previous studies have indicated that nitrogen dioxide (NO2) exposure could increase airway sensitivity to allergens for children. Recently, fetal stress was proposed as a crucial factor for allergic airway response occurring in offspring. Considering that there is inadequate evidence linking maternal NO2 exposure to offspring airway sensitivity to allergens, pregnant Balb/c mice were exposed daily to 2.5 ppm NO2 throughout the gestation period; then, the offspring were challenged to an allergen (ovalbumin, OVA) to evaluate airway sensitivity. For air + saline group and air + OVA group, offspring mice were maternally exposed to clean air followed by treatment with saline and OVA, respectively, in adulthood. For NO2 + saline group and NO2 + OVA group, offspring mice were maternally exposed to NO2 followed by treatment with saline and OVA, respectively, in adulthood. The results showed that maternal NO2 exposure increased the level of OVA-immunoglobulin (Ig) E in serum and caused airway hyper-responsiveness and pathological changes in offspring. Furthermore, maternal NO2 exposure altered the expression of pro-inflammatory factors and impaired the T helper (Th) 1/Th2 balance. In addition, janus kinase)-signal transducer and activator of transcription 6 pathway participated in OVA-induced airway sensitivity of offspring. Our study showed that the potential risk of airway sensitivity to allergens in offspring is enhanced by maternal NO2 exposure and proposed a possible mechanism for preventing, alleviating, and evaluating the outcomes in polluted environments.
Collapse
Affiliation(s)
- Huifeng Yue
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Wei Yan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China.
| |
Collapse
|
17
|
Intestinal dysbacteriosis potentiates ovalbumin-induced allergic airway inflammation by inhibiting microRNA-130a to upregulate tumor necrosis factor α. Int Immunopharmacol 2018; 60:34-40. [PMID: 29702281 DOI: 10.1016/j.intimp.2018.04.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 04/08/2018] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
Allergic airway diseases (AAD), including chronic disorders such as allergic rhinitis, are resulted from complicated immunological interactions. Intestinal dysbacteriosis (ID) has been implicated in immune response to respiratory infections. We aimed to investigate the effect of ID on a mouse model of AAD, and the potential molecular factors involved. Ovalbumin (OVA) was employed to sensitize and challenge mice to elicit allergic inflammation in the upper as well as the lower airways. OVA-induced AAD model mice and control mice were raised with or without antibiotics treatment to establish the combinational AAD + ID mouse model. Characteristic symptoms of AAD were evaluated in regard to allergic symptoms, serum OVA specific IgE level, as well as inflammation cells, cytokines and microRNA expression profile in nasal lavage fluid (NALF) and bronchoalveolar lavage fluid (BALF). In AAD mice, ID caused increased nasal rubbing, sneezing, serum OVA specific IgE level and pro-inflammatory cytokine tumor necrosis factor α (TNF-α) in NALF and BALF. ID also inhibited microRNA-130a of AAD mice. Further molecular experiments indicated that microRNA-130a could specifically target and repress TNF-α. ID increases the susceptibility to AAD and allergic inflammatory response, possibly by inhibiting microRNA-130a to upregulate TNF-α.
Collapse
|
18
|
Coffey AR, Smallwood TL, Albright J, Hua K, Kanke M, Pomp D, Bennett BJ, Sethupathy P. Systems genetics identifies a co-regulated module of liver microRNAs associated with plasma LDL cholesterol in murine diet-induced dyslipidemia. Physiol Genomics 2017; 49:618-629. [PMID: 28916633 DOI: 10.1152/physiolgenomics.00050.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/21/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023] Open
Abstract
Chronically altered levels of circulating lipids, termed dyslipidemia, is a significant risk factor for a number of metabolic and cardiovascular morbidities. MicroRNAs (miRNAs) have emerged as important regulators of lipid balance, have been implicated in dyslipidemia, and have been proposed as candidate therapeutic targets in lipid-related disorders including atherosclerosis. A major limitation of most murine studies of miRNAs in lipid metabolic disorders is that they have been performed in just one (or very few) inbred strains, such as C57BL/6. Moreover, although individual miRNAs have been associated with lipid phenotypes, it is well understood that miRNAs likely work together in functional modules. To address these limitations, we implemented a systems genetics strategy using the Diversity Outbred (DO) mouse population. Specifically, we performed gene and miRNA expression profiling in the livers from ~300 genetically distinct DO mice after 18 wk on either a high-fat/high-cholesterol diet or a high-protein diet. Large-scale correlative analysis of these data with a wide range of cardio-metabolic end points revealed a co-regulated module of miRNAs significantly associated with circulating low-density lipoprotein cholesterol (LDL-C) levels. The hubs of this module were identified as miR-199a, miR-181b, miR-27a, miR-21_-_1, and miR-24. In sum, we demonstrate that a high-fat/high-cholesterol diet robustly rewires the miRNA regulatory network, and we identify a small group of co-regulated miRNAs that may exert coordinated effects to control circulating LDL-C.
Collapse
Affiliation(s)
- Alisha R Coffey
- Curriculum in Genetics and Molecular Biology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina.,Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina
| | - Tangi L Smallwood
- Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina
| | - Jody Albright
- US Department of Agriculture, ARS Western Human Nutrition Research Center, University of California, Davis, Davis, California; and
| | - Kunjie Hua
- Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina
| | - Matt Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Daniel Pomp
- Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina
| | - Brian J Bennett
- Curriculum in Genetics and Molecular Biology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina.,Department of Genetics, School of Medicine, University of North Carolina Chapel Hill, Chapel Hill, North Carolina.,US Department of Agriculture, ARS Western Human Nutrition Research Center, University of California, Davis, Davis, California; and
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| |
Collapse
|
19
|
Moffett HF, Cartwright ANR, Kim HJ, Godec J, Pyrdol J, Äijö T, Martinez GJ, Rao A, Lu J, Golub TR, Cantor H, Sharpe AH, Novina CD, Wucherpfennig KW. The microRNA miR-31 inhibits CD8 + T cell function in chronic viral infection. Nat Immunol 2017; 18:791-799. [PMID: 28530712 PMCID: PMC5753758 DOI: 10.1038/ni.3755] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/27/2017] [Indexed: 12/13/2022]
Abstract
During infection, antigen-specific T cells undergo tightly regulated developmental transitions controlled by transcriptional and post-transcriptional regulation of gene expression. We found that the microRNA miR-31 was strongly induced by activation of the T cell antigen receptor (TCR) in a pathway involving calcium and activation of the transcription factor NFAT. During chronic infection with lymphocytic choriomeningitis virus (LCMV) clone 13, miR-31-deficent mice recovered from clinical disease, while wild-type mice continued to show signs of disease. This disease phenotype was explained by the presence of larger numbers of cytokine-secreting LCMV-specific CD8+ T cells in miR-31-deficent mice than in wild-type mice. Mechanistically, miR-31 increased the sensitivity of T cells to type I interferons, which interfered with effector T cell function and increased the expression of several proteins related to T cell dysfunction during chronic infection. These studies identify miR-31 as an important regulator of T cell exhaustion in chronic infection.
Collapse
Affiliation(s)
- Howell F Moffett
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Adam N R Cartwright
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hye-Jung Kim
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jernej Godec
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jason Pyrdol
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tarmo Äijö
- Department of Information and Computer Science, Aalto University School of Science, Aalto, Finland
| | - Gustavo J Martinez
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Anjana Rao
- La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Jun Lu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Todd R Golub
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Harvey Cantor
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl D Novina
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology &Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
STEPICHEVA NADEZDAA, SONG JIAL. Function and regulation of microRNA-31 in development and disease. Mol Reprod Dev 2016; 83:654-74. [PMID: 27405090 PMCID: PMC6040227 DOI: 10.1002/mrd.22678] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that orchestrate numerous cellular processes both under normal physiological conditions as well as in diseases. This review summarizes the functional roles and transcriptional regulation of the highly evolutionarily conserved miRNA, microRNA-31 (miR-31). miR-31 is an important regulator of embryonic implantation, development, bone and muscle homeostasis, and immune system function. Its own regulation is disrupted during the onset and progression of cancer and autoimmune disorders such as psoriasis and systemic lupus erythematosus. Limited studies suggest that miR-31 is transcriptionally regulated by epigenetics, such as methylation and acetylation, as well as by a number of transcription factors. Overall, miR-31 regulates diverse cellular and developmental processes by targeting genes involved in cell proliferation, apoptosis, cell differentiation, and cell motility. Mol. Reprod. Dev. 83: 654-674, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - JIA L. SONG
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
21
|
Transcriptomic Analysis of Chronic Hepatitis B and C and Liver Cancer Reveals MicroRNA-Mediated Control of Cholesterol Synthesis Programs. mBio 2015; 6:e01500-15. [PMID: 26646011 PMCID: PMC4676282 DOI: 10.1128/mbio.01500-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chronic hepatitis B (CHB), chronic hepatitis C (CHC), and associated hepatocellular carcinoma (HCC) are characterized by cholesterol imbalance and dyslipidemia; however, the key regulatory drivers of these phenotypes are incompletely understood. Using gene expression microarrays and high-throughput sequencing of small RNAs, we performed integrative analysis of microRNA (miRNA) and gene expression in nonmalignant and matched cancer tissue samples from human subjects with CHB or CHC and HCC. We also carried out follow-up functional studies of specific miRNAs in a cell-based system. These studies led to four major findings. First, pathways affecting cholesterol homeostasis were among the most significantly overrepresented among genes dysregulated in chronic viral hepatitis and especially in tumor tissue. Second, for each disease state, specific miRNA signatures that included miRNAs not previously associated with chronic viral hepatitis, such as miR-1307 in CHC, were identified. Notably, a few miRNAs, including miR-27 and miR-224, were components of the miRNA signatures of all four disease states: CHB, CHC, CHB-associated HCC, and CHC-associated HCC. Third, using a statistical simulation method (miRHub) applied to the gene expression data, we identified candidate master miRNA regulators of pathways controlling cholesterol homeostasis in chronic viral hepatitis and HCC, including miR-21, miR-27, and miR-33. Last, we validated in human hepatoma cells that both miR-21 and miR-27 significantly repress cholesterol synthesis and that miR-27 does so in part through regulation of the gene that codes for the rate-limiting enzyme 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase (HMGCR). Hepatitis B virus (HBV) and hepatitis C virus (HCV) are phylogenetically unrelated hepatotropic viruses that persistently infect hundreds of millions of people world-wide, often leading to chronic liver disease and hepatocellular carcinoma (HCC). Chronic hepatitis B (CHB), chronic hepatitis C (CHC), and associated HCC often lead to cholesterol imbalance and dyslipidemia. However, the regulatory mechanisms underlying the dysregulation of lipid pathways in these disease states are incompletely understood. MicroRNAs (miRNAs) have emerged as critical modulators of lipid homeostasis. Here we use a blend of genomic, molecular, and biochemical strategies to identify key miRNAs that drive the lipid phenotypes of chronic viral hepatitis and HCC. These findings provide a panoramic view of the miRNA landscape in chronic viral hepatitis, which could contribute to the development of novel and more-effective miRNA-based therapeutic strategies.
Collapse
|