1
|
Banack SA, Dunlop RA, Mehta P, Mitsumoto H, Wood SP, Han M, Cox PA. A microRNA diagnostic biomarker for amyotrophic lateral sclerosis. Brain Commun 2024; 6:fcae268. [PMID: 39280119 PMCID: PMC11398878 DOI: 10.1093/braincomms/fcae268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/13/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Blood-based diagnostic biomarkers for amyotrophic lateral sclerosis will improve patient outcomes and positively impact novel drug development. Critical to the development of such biomarkers is robust method validation, optimization and replication with adequate sample sizes and neurological disease comparative blood samples. We sought to test an amyotrophic lateral sclerosis biomarker derived from diverse samples to determine if it is disease specific. Extracellular vesicles were extracted from blood plasma obtained from individuals diagnosed with amyotrophic lateral sclerosis, primary lateral sclerosis, Parkinson's disease and healthy controls. Immunoaffinity purification was used to create a neural-enriched extracellular vesicle fraction. MicroRNAs were measured across sample cohorts using real-time polymerase chain reaction. A Kruskal-Wallis test was used to assess differences in plasma microRNAs followed by post hoc Mann-Whitney tests to compare disease groups. Diagnostic accuracy was determined using a machine learning algorithm and a logistic regression model. We identified an eight-microRNA diagnostic signature for blood samples from amyotrophic lateral sclerosis patients with high sensitivity and specificity and an area under the curve calculation of 98% with clear statistical separation from neurological controls. The eight identified microRNAs represent disease-related biological processes consistent with amyotrophic lateral sclerosis. The direction and magnitude of gene fold regulation are consistent across four separate patient cohorts with real-time polymerase chain reaction analyses conducted in two laboratories from diverse samples and sample collection procedures. We propose that this diagnostic signature could be an aid to neurologists to supplement current clinical metrics used to diagnose amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
| | | | - Paul Mehta
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA 30033, USA
| | - Hiroshi Mitsumoto
- Eleanor and Lou Gehrig MND/ALS Research Center, Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Moon Han
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA 30033, USA
| | | |
Collapse
|
2
|
Hubert A, Achour D, Grare C, Zarcone G, Muntaner M, Hamroun A, Gauthier V, Amouyel P, Matran R, Zerimech F, Lo-Guidice JM, Dauchet L. The relationship between residential exposure to atmospheric pollution and circulating miRNA in adults living in an urban area in northern France. ENVIRONMENT INTERNATIONAL 2023; 174:107913. [PMID: 37037173 DOI: 10.1016/j.envint.2023.107913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
INTRODUCTION MicroRNAs are epigenetic regulatory factors capable of silencing the expression of target genes and might mediate the effects of air pollution on health. The objective of the present population-based study was to investigate the association between microRNA expression and long-term, residential exposure to atmospheric PM10 and NO2. METHOD We included 998 non-smoking adult participants from the cross-sectional ELISABET survey (2010-2014) in the Lille urban area of France. The mean residential annual pollution levels were estimated with an atmospheric dispersion modelling system. Ten microRNAs were selected on the basis of the literature data, together with two housekeeping microRNAs (miR-93-5p and miR-191-5p) and were quantified with RT-qPCRs. Multivariate linear regression models were used to study the association between microRNAs and air pollution. The threshold for statistical significance (after correction for the FDR) was set to p < 0.1. RESULTS The mean annual exposure between 2011 and the year of inclusion was 26.4 ± 2.0 µg/m3 for PM10 and 24.7 ± 5.1 µg/m3 for NO2. Each 2 µg/m3 increment in PM10 exposure was associated with an 8.6% increment (95%CI [3.1; 14.3]; pFDR = 0.019) in miR-451a expression. A 5 µg/m3 increment in NO2 exposure was associated with a 5.3% increment ([0.7; 10]; pFDR = 0.056) in miR451a expression, a 3.6% decrement (95%CI [-6.1; -1.1]; pFDR = 0.052) in miR-223-3p expression, a 3.8% decrement (95%CI[-6.8; -0.7]; pFDR = 0.079) in miR-28-3p expression, a 4.3% decrement (95%CI [-7.7; -0.8]; pFDR = 0.055) in miR-146a-5p expression, and a 4.0% decrement (95% CI[-7.4; -0.4]; pFDR = 0.059) in miR-23a-5p expression. The difference between the two housekeeping microRNAs miR-93-5p and miR-191-5p was also associated with PM10 and NO2 exposure. CONCLUSION Our results suggest that circulating miRNAs are potentially valuable biomarkers of the effects of air pollution.
Collapse
Affiliation(s)
- Audrey Hubert
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| | - Djamal Achour
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Céline Grare
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Gianni Zarcone
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Manon Muntaner
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| | - Aghiles Hamroun
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| | - Victoria Gauthier
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| | - Philippe Amouyel
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| | - Régis Matran
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Farid Zerimech
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Jean-Marc Lo-Guidice
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483 - IMPECS - IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France.
| | - Luc Dauchet
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France.
| |
Collapse
|
3
|
Jin J, Yang Z, Liu H, Guo M, Chen B, Zhu H, Wang Y, Lin J, Wang S, Chen S. Effects of acupuncture on the miR-146a-mediated IRAK1/TRAF6/NF-κB signaling pathway in rats with sarcopenia induced by D-galactose. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:47. [PMID: 36819511 PMCID: PMC9929824 DOI: 10.21037/atm-22-6082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Background Sarcopenia during aging is closely linked to sterile, low-grade, chronic inflammation. However, considering the increasingly aging global population, the effectiveness of existing treatments for sarcopenia is not exact, and acupuncture, as an effective anti-inflammatory therapy, has the potential to treat it. Methods Fifty Sprague-Dawley rats were randomly allocated into five groups, including Control group, D-galactose (D-gal) group, D-gal + acupuncture (DA) group, D-gal + non-acupoint (DN) group and D-gal amino acid mixture (DAA) group. An aging rat was model constructed using D-gal for 12 weeks. Rats in the control group received 0.9% physiological saline daily. Treatment groups were acupunctured or given amino acid mixture interventions daily, and lasted for last 4 consecutive weeks. The effects of acupuncture were evaluated by the hematoxylin and eosin staining (H&E), transmission electron microscopic (TEM) examination and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. The anti-inflammatory mechanism of acupuncture was studied by using the expressions of microRNA-146a (miR-146a) mediated nuclear factor-kappa B (NF-κB) signaling pathway-related proteins were detected by immunofluorescence, western blotting, quantitative real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). Results Rats injected by D-galactose (D-gal) revealed apparent skeletal muscle atrophy with significantly reduced cross-sectional area and fiber diameter. In contrast, acupuncture treatment alleviated these hallmarks of skeletal muscle atrophy and mitigated the mitochondrial aberrations and skeletal muscle apoptosis in D-gal rats. In addition, acupuncture also downgraded the overexpression of inflammatory factors in skeletal muscle, influenced miR-146a and the target genes level, and inhibited NF-κB nuclear translation in D-gal rats. Conclusions Acupuncture may ameliorate skeletal muscle atrophy, and its effects may be associated with the control of mitochondrial function regulation and the suppression of inflammation.
Collapse
Affiliation(s)
- Jing Jin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhengyu Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haichao Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingling Guo
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Borui Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Haoming Zhu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianping Lin
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shizhong Wang
- The School of Health, Fujian Medical University, Fuzhou, China
| | - Shaoqing Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
4
|
Ziętara KJ, Lejman J, Wojciechowska K, Lejman M. The Importance of Selected Dysregulated microRNAs in Diagnosis and Prognosis of Childhood B-Cell Precursor Acute Lymphoblastic Leukemia. Cancers (Basel) 2023; 15:428. [PMID: 36672378 PMCID: PMC9856444 DOI: 10.3390/cancers15020428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a frequent type of childhood hematological malignancy. The disease is classified into several subtypes according to genetic abnormalities. MicroRNAs (miRNAs) are involved in pathological processes (e.g., proliferation, apoptosis, differentiation). A miRNA is a group of short non-coding RNAs with relevant regulatory effects on gene expression achieved by suppression of the translation or degradation of messenger RNA (mRNA). These molecules act as tumor suppressors and/or oncogenes in the pathogenesis of pediatric leukemias. The characteristic features of miRNAs are their stable form and the possibility of secretion to the circulatory system. The role of miRNA in BCP-ALL pathogenesis is still emerging, but several studies have suggested using miRNA expression profiles as biomarkers for diagnosis, prognosis, and response to therapy in leukemia. The dysregulation of some miRNAs involved in childhood acute lymphoid leukemia, such as miR-155, miR-200c, miR-100, miR-181a, miR125b, and miR146a is discussed, showing their possible employment as therapeutic targets. In the current review, the capabilities of miRNAs in non-invasive diagnostics and their prognostic potential as biomarkers are presented.
Collapse
Affiliation(s)
- Karolina Joanna Ziętara
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Jan Lejman
- Independent Public Health Care Facility of The Ministry of Internal Affairs and Administration in Lublin, 20-331 Lublin, Poland
| | - Katarzyna Wojciechowska
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
5
|
Mirza S, Penny C, Jain NK, Rawal RM. Curcumin mediated dendritic cell maturation by modulating cancer associated fibroblasts-derived exosomal miRNA-146a. J Cancer Res Ther 2023; 19:S649-S657. [PMID: 38384034 DOI: 10.4103/jcrt.jcrt_1286_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/20/2022] [Indexed: 02/23/2024]
Abstract
BACKGROUND Though cancer associated fibroblasts (CAFs), being a main component of tumor microenvironment (TME), are known to modulate immune response through secretion of various growth hormones, exosomes carrying miRNAs and cytokines; their effect on dendritic cells (DCs) are yet to be elucidated. Thus, aim of this study was to assess the effect of miRNAs and cytokines released by lung-CAFs and to evaluate immunomodulatory potential of curcumin on DC maturation through modulating their TME. MATERIAL AND METHODS To check the effect of CAFs derived exosomes on DC maturation, we cultured imDCs in the presence of CAFs derived conditioned media (CAFs-CM) and characterized by the presence of maturation markers CD80, CD83, CD86 and CTLA4 using qRT-PCR. Additionally, expression of miR-221, miR-222, miR-155, miR-142-3p and miR-146a was assessed to evaluate the role of epigenetic regulators on DC maturation. Likewise, cytokine profiling of CAFs-CM as well as CAFs-CM treated with curcumin was also conducted using ELISA. RESULTS Results revealed the generation of regulatory DCs which were characterized by decreased expression of maturation markers in the presence of CAFs-CM. In addition, such DCs showed higher expression of epigenetic regulator miR-146a which was positively correlated with increased expression of anti-inflammatory cytokines like IL-6, IL-10, TGF-β and decreased expression of TNF-α (pro-inflammatory). Moreover, curcumin had the potential to convert regulatory DCs generated by CAFs into mDCs, which were characterized by high expression of co-stimulatory molecules, low expression of CTLA4, lower levels of immune suppressive cytokines production and lower levels of miR-146a. CONCLUSION Collectively, these findings provide insight into understanding the immunomodulatory role of curcumin in targeting CAFs and modulating TME, thus enhancing antitumor immune response in DC based therapy.
Collapse
Affiliation(s)
- Sheefa Mirza
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
- Division of Medicinal Chemistry and Pharmacogenomics, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nayan K Jain
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Rakesh M Rawal
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
- Division of Medicinal Chemistry and Pharmacogenomics, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| |
Collapse
|
6
|
Wicks EE, Ran KR, Kim JE, Xu R, Lee RP, Jackson CM. The Translational Potential of Microglia and Monocyte-Derived Macrophages in Ischemic Stroke. Front Immunol 2022; 13:897022. [PMID: 35795678 PMCID: PMC9251541 DOI: 10.3389/fimmu.2022.897022] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response to ischemic stroke is an area of study that is at the forefront of stroke research and presents promising new avenues for treatment development. Upon cerebral vessel occlusion, the innate immune system is activated by danger-associated molecular signals from stressed and dying neurons. Microglia, an immune cell population within the central nervous system which phagocytose cell debris and modulate the immune response via cytokine signaling, are the first cell population to become activated. Soon after, monocytes arrive from the peripheral immune system, differentiate into macrophages, and further aid in the immune response. Upon activation, both microglia and monocyte-derived macrophages are capable of polarizing into phenotypes which can either promote or attenuate the inflammatory response. Phenotypes which promote the inflammatory response are hypothesized to increase neuronal damage and impair recovery of neuronal function during the later phases of ischemic stroke. Therefore, modulating neuroimmune cells to adopt an anti-inflammatory response post ischemic stroke is an area of current research interest and potential treatment development. In this review, we outline the biology of microglia and monocyte-derived macrophages, further explain their roles in the acute, subacute, and chronic stages of ischemic stroke, and highlight current treatment development efforts which target these cells in the context of ischemic stroke.
Collapse
|
7
|
Witten A, Martens L, Schäfer AC, Troidl C, Pankuweit S, Vlacil AK, Oberoi R, Schieffer B, Grote K, Stoll M, Markus B. Monocyte subpopulation profiling indicates CDK6-derived cell differentiation and identifies subpopulation-specific miRNA expression sets in acute and stable coronary artery disease. Sci Rep 2022; 12:5589. [PMID: 35379829 PMCID: PMC8979987 DOI: 10.1038/s41598-022-08600-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Coronary artery disease (CAD) is a long-lasting inflammatory disease characterized by monocyte migration into the vessel wall leading to clinical events like myocardial infarction (MI). However, the role of monocyte subsets, especially their miRNA-driven differentiation in this scenario is still in its infancy. Here, we characterized monocyte subsets in controls and disease phenotypes of CAD and MI patients using flow cytometry and miRNA and mRNA expression profiling using RNA sequencing. We observed major differences in the miRNA profiles between the classical (CD14++CD16−) and nonclassical (CD14+CD16++) monocyte subsets irrespective of the disease phenotype suggesting the Cyclin-dependent Kinase 6 (CDK6) to be an important player in monocyte maturation. Between control and MI patients, we found a set of miRNAs to be differentially expressed in the nonclassical monocytes and targeting CCND2 (Cyclin D2) that is able to enhance myocardial repair. Interestingly, miRNAs as miR-125b playing a role in vascular calcification were differentially expressed in the classical subset in patients suffering from CAD and not MI in comparison to control samples. In conclusion, our study describes specific peculiarities of monocyte subset miRNA expression in control and diseased samples and provides basis to further functional analysis and to identify new cardiovascular disease treatment targets.
Collapse
|
8
|
Zhang Y, Yang Y, Guo J, Cui L, Yang L, Li Y, Mou Y, Jia C, Zhang L, Song X. miR-146a enhances regulatory T-cell differentiation and function in allergic rhinitis by targeting STAT5b. Allergy 2022; 77:550-558. [PMID: 34716993 DOI: 10.1111/all.15163] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 10/24/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND MicroRNA (miR)-146a, as an important immune regulatory factor with an anti-inflammatory effect, plays a crucial role in regulatory T-cell (Tregs) differentiation and function in allergic rhinitis (AR). The present study aimed to investigate the regulatory mechanism employed by miR-146a to control Treg differentiation and function in AR. METHODS Expression of miR-146a and STAT5b in peripheral blood mononuclear cells (PBMCs) and nasal mucosa from patients with AR was detected by qPCR and Western blotting. Tregs were quantified by flow cytometry in miR-146a knockdown or STAT5b knockdown PBMCs. FOXP3, IL-10, and TGF-β levels were detected by Western blotting or ELISA in miR-146a knockdown or STAT5b overexpressing PBMCs, as well as in STAT5b knockdown PBMCs overexpressing miR-146a. The effect of miR-146a on STAT5b was observed by luciferase assay and knockdown experiments. RESULTS Levels of miR146a and STAT5b in the nasal mucosa or PBMCs were significantly lower in the AR group than in the control group. There were significantly fewer Tregs in miR-146a knockdown or STAT5b knockdown PBMCs compared to control PBMCs. Expression of FOXP3, IL-10, and TGF-β was decreased in the miR-146a knockdown group but increased in the STAT5b overexpression group. In contrast, miR-146a overexpression increased the levels of these factors, but knockdown of STAT5b significantly inhibited this effect. Luciferase assay and knockdown experiments showed that miR-146a bound directly to STAT5b. CONCLUSIONS miR-146a enhances Treg differentiation and function in AR by positively targeting STAT5b.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Jing Guo
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Limei Cui
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Liping Yang
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Yumei Li
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Yakui Mou
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Chuanliang Jia
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| | - Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery Beijing Tongren HospitalCapital Medical University Beijing China
- Beijing Key Laboratory of Nasal Diseases Beijing Institute of Otolaryngology Beijing China
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery Yantai Yuhuangding Hospital Qingdao University Yantai China
| |
Collapse
|
9
|
Staats KA, Borchelt DR, Tansey MG, Wymer J. Blood-based biomarkers of inflammation in amyotrophic lateral sclerosis. Mol Neurodegener 2022; 17:11. [PMID: 35073950 PMCID: PMC8785449 DOI: 10.1186/s13024-022-00515-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease in which many processes are detected including (neuro)inflammation. Many drugs have been tested for ALS in clinical trials but most have failed to reach their primary endpoints. The development and inclusion of different types of biomarkers in diagnosis and clinical trials can assist in determining target engagement of a drug, in distinguishing between ALS and other diseases, and in predicting disease progression rate, drug responsiveness, or an adverse event. Ideally, among other characteristics, a biomarker in ALS correlates highly with a disease process in the central nervous system or with disease progression and is conveniently obtained in a peripheral tissue. Here, we describe the state of biomarkers of inflammation in ALS by focusing on peripherally detectable and cellular responses from blood cells, and provide new (combinatorial) directions for exploration that are now feasible due to technological advancements.
Collapse
Affiliation(s)
- Kim A. Staats
- Staats Life Sciences Consulting, LLC, Los Angeles, CA USA
| | - David R. Borchelt
- Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida USA
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease at The University of Florida College of Medicine, Gainesville, Florida USA
| | - James Wymer
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida USA
| |
Collapse
|
10
|
Dai S, Wen Y, Luo P, Ma L, Liu Y, Ai J, Shi C. Therapeutic implications of exosomes in the treatment of radiation injury. BURNS & TRAUMA 2022; 10:tkab043. [PMID: 35071650 PMCID: PMC8778593 DOI: 10.1093/burnst/tkab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/11/2021] [Indexed: 12/28/2022]
Abstract
Radiotherapy is one of the main cancer treatments, but it may damage normal tissue and cause various side effects. At present, radioprotective agents used in clinics have side effects such as nausea, vomiting, diarrhea and hypotension, which limit their clinical application. It has been found that exosomes play an indispensable role in radiation injury. Exosomes are lipid bilayer vesicles that carry various bioactive substances, such as proteins, lipids and microRNA (miRNA), that play a key role in cell-to-cell communication and affect tissue injury and repair. In addition, studies have shown that radiation can increase the uptake of exosomes in cells and affect the composition and secretion of exosomes. Here, we review the existing studies and discuss the effects of radiation on exosomes and the role of exosomes in radiation injury, aiming to provide new insights for the treatment of radiation injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Junhua Ai
- Correspondence. Junhua Ai, ; Chunmeng Shi,
| | | |
Collapse
|
11
|
Alwani A, Andreasik A, Szatanek R, Siedlar M, Baj-Krzyworzeka M. The Role of miRNA in Regulating the Fate of Monocytes in Health and Cancer. Biomolecules 2022; 12:100. [PMID: 35053248 PMCID: PMC8773712 DOI: 10.3390/biom12010100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 12/23/2022] Open
Abstract
Monocytes represent a heterogeneous population of blood cells that provide a link between innate and adaptive immunity. The unique potential of monocytes as both precursors (e.g., of macrophages) and effector cells (as phagocytes or cytotoxic cells) makes them an interesting research and therapeutic target. At the site of a tumor, monocytes/macrophages constitute a major population of infiltrating leukocytes and, depending on the type of tumor, may play a dual role as either a bad or good indicator for cancer recovery. The functional activity of monocytes and macrophages derived from them is tightly regulated at the transcriptional and post-transcriptional level. This review summarizes the current understanding of the role of small regulatory miRNA in monocyte formation, maturation and function in health and cancer development. Additionally, signatures of miRNA-based monocyte subsets and the influence of exogenous miRNA generated in the tumor environment on the function of monocytes are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, 30-663 Kraków, Poland; (A.A.); (A.A.); (R.S.); (M.S.)
| |
Collapse
|
12
|
Abstract
Periodontitis is a multi-etiologic infection characterized clinically by pathologic loss of the periodontal ligament and alveolar bone. Herpesviruses and specific bacterial species are major periodontal pathogens that cooperate synergistically in producing severe periodontitis. Cellular immunity against herpesviruses and humoral immunity against bacteria are key periodontal host defenses. Genetic, epigenetic, and environmental factors are modifiers of periodontal disease severity. MicroRNAs are a class of noncoding, gene expression-based, posttranscriptional regulatory RNAs of great importance for maintaining tissue homeostasis. Aberrant expression of microRNAs has been associated with several medical diseases. Periodontal tissue cells and herpesviruses elaborate several microRNAs that are of current research interest. This review attempts to conceptualize the role of periodontal microRNAs in the pathogenesis of periodontitis. The diagnostic potential of salivary microRNAs is also addressed. Employment of microRNA technology in periodontics represents an interesting new preventive and therapeutic possibility.
Collapse
Affiliation(s)
- Afsar R Naqvi
- Mucosal Immunology Laboratory, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jørgen Slots
- Department of Periodontology, University of Southern California School of Dentistry, Los Angeles, California, USA
| |
Collapse
|
13
|
Olivieri F, Prattichizzo F, Giuliani A, Matacchione G, Rippo MR, Sabbatinelli J, Bonafè M. miR-21 and miR-146a: The microRNAs of inflammaging and age-related diseases. Ageing Res Rev 2021; 70:101374. [PMID: 34082077 DOI: 10.1016/j.arr.2021.101374] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
The first paper on "inflammaging" published in 2001 paved the way for a unifying theory on how and why aging turns out to be the main risk factor for the development of the most common age-related diseases (ARDs). The most exciting challenge on this topic was explaining how systemic inflammation steeps up with age and why it shows different rates among individuals of the same chronological age. The "epigenetic revolution" in the past twenty years conveyed that the assessment of the individual genetic make-up is not enough to depict the trajectories of age-related inflammation. Accordingly, others and we have been focusing on the role of non-coding RNA, i.e. microRNAs (miRNAs), in inflammaging. The results obtained in the latest 10 years underpinned the key role of a miRNA subset that we have called inflammamiRs, owing to their ability to master (NF-κB)-driven inflammatory pathways. In this review, we will focus on two inflammamiRs, i.e. miR-21-5p and miR-146a-5p, which target a variety of molecules belonging to the NF-κB/NLRP3 pathways. The interplay between miR-146a-5p and IL-6 in the context of aging and ARDs will also be highlighted. We will also provide the most relevant evidence suggesting that circulating inflammamiRs, along with IL-6, can measure the degree of inflammaging.
Collapse
|
14
|
Munir MT, Kay MK, Kang MH, Rahman MM, Al-Harrasi A, Choudhury M, Moustaid-Moussa N, Hussain F, Rahman SM. Tumor-Associated Macrophages as Multifaceted Regulators of Breast Tumor Growth. Int J Mol Sci 2021; 22:6526. [PMID: 34207035 PMCID: PMC8233875 DOI: 10.3390/ijms22126526] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most commonly occurring cancer in women of Western countries and is the leading cause of cancer-related mortality. The breast tumor microenvironment contains immune cells, fibroblasts, adipocytes, mesenchymal stem cells, and extracellular matrix. Among these cells, macrophages or tumor-associated macrophages (TAMs) are the major components of the breast cancer microenvironment. TAMs facilitate metastasis of the breast tumor and are responsible for poor clinical outcomes. High TAM density was also found liable for the poor prognosis of breast cancer. These observations make altering TAM function a potential therapeutic target to treat breast cancer. The present review summarizes the origin of TAMs, mechanisms of macrophage recruitment and polarization in the tumor, and the contributions of TAMs in tumor progression. We have also discussed our current knowledge about TAM-targeted therapies and the roles of miRNAs and exosomes in re-educating TAM function.
Collapse
Affiliation(s)
- Maliha Tabassum Munir
- Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.T.M.); (N.M.-M.)
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Matthew K. Kay
- Texas A&M University Health Sciences Center, College Station, TX 77843, USA; (M.K.K.); (M.C.)
| | - Min H. Kang
- Cancer Center, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Md Mizanur Rahman
- Department of Biological and Environmental Sciences, Qatar University, Doha 2713, Qatar;
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz 616, Oman;
| | - Mahua Choudhury
- Texas A&M University Health Sciences Center, College Station, TX 77843, USA; (M.K.K.); (M.C.)
| | - Naima Moustaid-Moussa
- Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.T.M.); (N.M.-M.)
- Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Fazle Hussain
- Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA;
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz 616, Oman;
| |
Collapse
|
15
|
Garo LP, Ajay AK, Fujiwara M, Gabriely G, Raheja R, Kuhn C, Kenyon B, Skillin N, Kadowaki-Saga R, Saxena S, Murugaiyan G. MicroRNA-146a limits tumorigenic inflammation in colorectal cancer. Nat Commun 2021; 12:2419. [PMID: 33893298 PMCID: PMC8065171 DOI: 10.1038/s41467-021-22641-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation can drive tumor development. Here, we have identified microRNA-146a (miR-146a) as a major negative regulator of colonic inflammation and associated tumorigenesis by modulating IL-17 responses. MiR-146a-deficient mice are susceptible to both colitis-associated and sporadic colorectal cancer (CRC), presenting with enhanced tumorigenic IL-17 signaling. Within myeloid cells, miR-146a targets RIPK2, a NOD2 signaling intermediate, to limit myeloid cell-derived IL-17-inducing cytokines and restrict colonic IL-17. Accordingly, myeloid-specific miR-146a deletion promotes CRC. Moreover, within intestinal epithelial cells (IECs), miR-146a targets TRAF6, an IL-17R signaling intermediate, to restrict IEC responsiveness to IL-17. MiR-146a within IECs further suppresses CRC by targeting PTGES2, a PGE2 synthesis enzyme. IEC-specific miR-146a deletion therefore promotes CRC. Importantly, preclinical administration of miR-146a mimic, or small molecule inhibition of the miR-146a targets, TRAF6 and RIPK2, ameliorates colonic inflammation and CRC. MiR-146a overexpression or miR-146a target inhibition represent therapeutic approaches that limit pathways converging on tumorigenic IL-17 signaling in CRC.
Collapse
Affiliation(s)
- Lucien P Garo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Amrendra K Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mai Fujiwara
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brendan Kenyon
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nathaniel Skillin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryoko Kadowaki-Saga
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shrishti Saxena
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Ridzuan N, Zakaria N, Widera D, Sheard J, Morimoto M, Kiyokawa H, Mohd Isa SA, Chatar Singh GK, Then KY, Ooi GC, Yahaya BH. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD). Stem Cell Res Ther 2021; 12:54. [PMID: 33436065 PMCID: PMC7805108 DOI: 10.1186/s13287-020-02088-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is an incurable and debilitating chronic disease characterized by progressive airflow limitation associated with abnormal levels of tissue inflammation. Therefore, stem cell-based approaches to tackle the condition are currently a focus of regenerative therapies for COPD. Extracellular vesicles (EVs) released by all cell types are crucially involved in paracrine, extracellular communication. Recent advances in the field suggest that stem cell-derived EVs possess a therapeutic potential which is comparable to the cells of their origin. METHODS In this study, we assessed the potential anti-inflammatory effects of human umbilical cord mesenchymal stem cell (hUC-MSC)-derived EVs in a rat model of COPD. EVs were isolated from hUC-MSCs and characterized by the transmission electron microscope, western blotting, and nanoparticle tracking analysis. As a model of COPD, male Sprague-Dawley rats were exposed to cigarette smoke for up to 12 weeks, followed by transplantation of hUC-MSCs or application of hUC-MSC-derived EVs. Lung tissue was subjected to histological analysis using haematoxylin and eosin staining, Alcian blue-periodic acid-Schiff (AB-PAS) staining, and immunofluorescence staining. Gene expression in the lung tissue was assessed using microarray analysis. Statistical analyses were performed using GraphPad Prism 7 version 7.0 (GraphPad Software, USA). Student's t test was used to compare between 2 groups. Comparison among more than 2 groups was done using one-way analysis of variance (ANOVA). Data presented as median ± standard deviation (SD). RESULTS Both transplantation of hUC-MSCs and application of EVs resulted in a reduction of peribronchial and perivascular inflammation, alveolar septal thickening associated with mononuclear inflammation, and a decreased number of goblet cells. Moreover, hUC-MSCs and EVs ameliorated the loss of alveolar septa in the emphysematous lung of COPD rats and reduced the levels of NF-κB subunit p65 in the tissue. Subsequent microarray analysis revealed that both hUC-MSCs and EVs significantly regulate multiple pathways known to be associated with COPD. CONCLUSIONS In conclusion, we show that hUC-MSC-derived EVs effectively ameliorate by COPD-induced inflammation. Thus, EVs could serve as a new cell-free-based therapy for the treatment of COPD.
Collapse
Affiliation(s)
- Noridzzaida Ridzuan
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Jonathan Sheard
- Stem Cell Biology and Regenerative Medicine, School of Pharmacy, University of Reading, Reading, RG6 6AP, UK
| | - Mitsuru Morimoto
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Hirofumi Kiyokawa
- RIKEN Centre for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuou-ku, Kobe, 650-0047, Japan
| | - Seoparjoo Azmel Mohd Isa
- Department of Pathology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kubang Kerian, Malaysia
| | - Gurjeet Kaur Chatar Singh
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia
| | - Kong-Yong Then
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Ghee-Chien Ooi
- CryoCord Sdn Bhd, Bio-X Centre, 63000, Cyberjaya, Selangor, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Bertam, Penang, Malaysia.
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, 11800, Gelugor, Penang, Malaysia.
| |
Collapse
|
17
|
Chen C, Liu JM, Luo YP. MicroRNAs in tumor immunity: functional regulation in tumor-associated macrophages. J Zhejiang Univ Sci B 2020; 21:12-28. [PMID: 31898439 DOI: 10.1631/jzus.b1900452] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and are critical for cancer initiation and progression. MicroRNAs (miRNAs) could notably influence the phenotype of TAMs through various targets and signal pathways during cancer progression due to their post-transcriptional regulation. In this review, we discuss mainly the regulatory function of miRNAs on macrophage differentiation, functional polarization, and cellular crosstalk. Firstly, during the generation process, miRNAs take part in the differentiation from myeloid cells to mature macrophages, and this maturation process directly influences their recruitment into the TME, attracted by tumor cells. Secondly, macrophages in the TME can be either tumor-promoting or tumor-suppressing, depending on their functional polarization. Large numbers of miRNAs can influence the polarization of macrophages, which is crucial for tumor progression, including tumor cell invasion, intravasation, extravasation, and premetastatic site formation. Thirdly, crosstalk between tumor cells and macrophages is essential for TME formation and tumor progression, and miRNAs can be the mediator of communication in different forms, especially when encapsulated in microvesicles or exosomes. We also assess the potential value of certain macrophage-related miRNAs (MRMs) as diagnostic and prognostic markers, and discuss the possible development of MRM-based therapies.
Collapse
Affiliation(s)
- Chong Chen
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.,Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Jia-Ming Liu
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.,Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Yun-Ping Luo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.,Collaborative Innovation Center for Biotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences; School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
18
|
Park J, Chang JY, Kim JY, Lee JE. Monocyte Transmodulation: The Next Novel Therapeutic Approach in Overcoming Ischemic Stroke? Front Neurol 2020; 11:578003. [PMID: 33193029 PMCID: PMC7642685 DOI: 10.3389/fneur.2020.578003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The immune response following neuroinflammation is a vital element of ischemic stroke pathophysiology. After the onset of ischemic stroke, a specialized vasculature system that effectively protects central nervous system tissues from the invasion of blood cells and other macromolecules is broken down within minutes, thereby triggering the inflammation cascade, including the infiltration of peripheral blood leukocytes. In this series of processes, blood-derived monocytes have a significant effect on the outcome of ischemic stroke through neuroinflammatory responses. As neuroinflammation is a necessary and pivotal component of the reparative process after ischemic stroke, understanding the role of infiltrating monocytes in the modulation of inflammatory responses may offer a great opportunity to explore new therapies for ischemic stroke. In this review, we discuss and highlight the function and involvement of monocytes in the brain after ischemic injury, as well as their impact on tissue damage and repair.
Collapse
Affiliation(s)
- Joohyun Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Young Chang
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
19
|
Hsu KH, Wei CW, Su YR, Chou T, Lin YL, Yang FC, Tsou AP, Hsu CL, Tseng PH, Chen NJ, Jeng KS, Leu CM. Upregulation of RelB in the miR-122 knockout mice contributes to increased levels of proinflammatory chemokines/cytokines in the liver and macrophages. Immunol Lett 2020; 226:22-30. [DOI: 10.1016/j.imlet.2020.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/04/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
|
20
|
Al-Rawi NH, Al-Marzooq F, Al-Nuaimi AS, Hachim MY, Hamoudi R. Salivary microRNA 155, 146a/b and 203: A pilot study for potentially non-invasive diagnostic biomarkers of periodontitis and diabetes mellitus. PLoS One 2020; 15:e0237004. [PMID: 32756589 PMCID: PMC7406085 DOI: 10.1371/journal.pone.0237004] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Dysregulated expression of MicroRNAs (miRNAs) plays substantial role in the initiation and progression of both diabetes and periodontitis. The aim of the present study was to validate four miRNAs in saliva as potential predictive biomarkers of periodontal disease among patients with and without diabetes mellitus (DM). MiRNAs were extracted from the saliva of 24 adult subjects with DM and 29 healthy controls. Each group was subdivided into periodontally healthy or having periodontitis. In silico analysis identified 4 miRNAs (miRNA 155, 146 a/b and 203) as immune modulators. The expression of miRNAs-146a/b, 155, and 203 was tested using quantitative PCR. The expression levels in the study groups were compared to explore the effect of diabetes on periodontal status and vice versa. In our cohort, the four miRNAs expression were higher in patients with periodontitis and/or diabetes. miRNA-155 was the most reliable predictors of periodontitis among non-diabetics with an optimum cut-off value of < 8.97 with accuracy = 82.6%. MiRNA 146a, on the other hand, was the only reliable predictor of periodontitis among subjects with diabetes with optimum cut-off value of ≥11.04 with accuracy = 86.1%. The results of the present study concluded that MiRNA-146a and miRNA155 in saliva provide reliable, non-invasive, diagnostic and prognostic biomarkers that can be used to monitor periodontal health status among diabetic and non-diabetic patients.
Collapse
Affiliation(s)
- Natheer H. Al-Rawi
- Department of Oral & Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| | - Farah Al-Marzooq
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, UAE University, Al Ain, UAE
| | | | - Mahmood Y. Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| |
Collapse
|
21
|
Banack SA, Dunlop RA, Cox PA. An miRNA fingerprint using neural-enriched extracellular vesicles from blood plasma: towards a biomarker for amyotrophic lateral sclerosis/motor neuron disease. Open Biol 2020; 10:200116. [PMID: 32574550 PMCID: PMC7333885 DOI: 10.1098/rsob.200116] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Biomarkers for amyotrophic lateral sclerosis/motor neuron disease (ALS/MND) are currently not clinically available for disease diagnosis or analysis of disease progression. If identified, biomarkers could improve patient outcomes by enabling early intervention and assist in the determination of treatment efficacy. We hypothesized that neural-enriched extracellular vesicles could provide microRNA (miRNA) fingerprints with unequivocal signatures of neurodegeneration. Using blood plasma from ALS/MND patients and controls, we extracted neural-enriched extracellular vesicle fractions and conducted next-generation sequencing and qPCR of miRNA components of the transcriptome. We here report eight miRNA sequences which significantly distinguish ALS/MND patients from controls in a replicated experiment using a second cohort of patients and controls. miRNA sequences from patient blood samples using neural-enriched extracellular vesicles may yield unique insights into mechanisms of neurodegeneration and assist in early diagnosis of ALS/MND.
Collapse
Affiliation(s)
- Sandra Anne Banack
- Brain Chemistry Labs, Institute for Ethnomedicine, PO Box 3464, Jackson, WY 83001, USA
| | - Rachael Anne Dunlop
- Brain Chemistry Labs, Institute for Ethnomedicine, PO Box 3464, Jackson, WY 83001, USA
| | - Paul Alan Cox
- Brain Chemistry Labs, Institute for Ethnomedicine, PO Box 3464, Jackson, WY 83001, USA
| |
Collapse
|
22
|
Zhang L, Hofer TP, Zawada AM, Rotter B, Krezdorn N, Noessner E, Devaux Y, Heine G, Ziegler-Heitbrock L. Epigenetics in non-classical monocytes support their pro-inflammatory gene expression. Immunobiology 2020; 225:151958. [PMID: 32517883 DOI: 10.1016/j.imbio.2020.151958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 01/12/2023]
Abstract
Non-classical human monocytes are characterized by high-level expression of cytokines like TNF, but the mechanisms involved are elusive. We have identified miRNAs and CpG-methylation sites that are unique to non-classical monocytes, defined via CD14 and CD16 expression levels. For down-regulated miRNAs that are linked to up-regulated mRNAs the dominant gene ontology term was intracellular signal transduction. This included down-regulated miRNA-20a-5p and miRNA-106b-5p, which both are linked to increased mRNA for the TRIM8 signaling molecule. Methylation analysis revealed 16 hypo-methylated CpG sites upstream of 14 differentially increased mRNAs including 2 sites upstream of TRIM8. Consistent with a positive role in signal transduction, high TRIM8 levels went along with high basal TNF mRNA levels in non-classical monocytes. Since cytokine expression levels in monocytes strongly increase after stimulation with toll-like-receptor ligands, we have analyzed non-classical monocytes (defined via slan expression) after stimulation with lipopolysaccharide (LPS). LPS-stimulated cells continued to have low miRNA-20a and miRNA-106b and high TRIM8 mRNA levels and they showed a 10-fold increase in TNF mRNA. These data suggest that decreased miRNAs and CpG hypo-methylation is linked to enhanced expression of TRIM8 and that this can contribute to the increased TNF levels in non-classical human monocytes.
Collapse
Affiliation(s)
- Lu Zhang
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg
| | - Thomas P Hofer
- Immunoanalytics Research Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| | - Adam M Zawada
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany
| | | | | | - Elfriede Noessner
- Immunoanalytics Research Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg
| | - Gunnar Heine
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany
| | | |
Collapse
|
23
|
Silencing of long non-coding RNA MALAT1 suppresses inflammation in septic mice: role of microRNA-23a in the down-regulation of MCEMP1 expression. Inflamm Res 2020; 69:179-190. [PMID: 31893303 DOI: 10.1007/s00011-019-01306-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Sepsis is a life-threatening disease without ideal biomarkers. Some long non-coding RNAs (lncRNAs) are found to be implicated in sepsis. Thus, we investigated the effects of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on inflammation in septic mice and the potential mechanisms of the MALAT1/microRNA-23a (miR-23a)/MCEMP1 axis. METHODS The sepsis mice model was generated by cecal ligation and puncture (CLP). Then the expressions of lncRNA MALAT1, mast cell-expressed membrane protein 1 (MCEMP1), and miR-23a in septic mice were determined. The interaction between lncRNA MALAT1, miR-23a and MCEMP1 was confirmed. Loss- and gain-of-function approaches were used to verify the roles of the lncRNA MALAT1, miR-23a, and MCEMP1 in inflammation, cell proliferation and apoptosis in septic mice. RESULTS AND CONCLUSION The myeloperoxidase (MPO) activity and the expression of interleukin 6 (IL-6), IL-1β, IL-10, and tumor necrosis factor-α (TNF-α) were detected. High expression of the lncRNA MALAT1 and MCEMP1, as well as low expression of miR-23a, was observed in septic mice. LncRNA MALAT1 competitively bound to miR-23a, and miR-23a targeted MCEMP1. Moreover, the down-regulation of lncRNA MALAT1 repressed the expression of MPO, IL-6, IL-10, TNF-α, and IL-1β. Silencing of lncRNA MALAT1 or overexpression of miR-23a reduced inflammation, inhibited cell proliferation, and promoted cell apoptosis in septic mice. Taken together, MALAT1 promotes the inflammation in septic mice by binding to miR-23a to up-regulate MCEMP1. Therefore, silencing of lncRNA MALAT1 might provide a novel therapeutic target for sepsis.
Collapse
|
24
|
van der Vorst EPC, Weber C. Novel Features of Monocytes and Macrophages in Cardiovascular Biology and Disease. Arterioscler Thromb Vasc Biol 2019; 39:e30-e37. [PMID: 30673349 DOI: 10.1161/atvbaha.118.312002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Emiel P C van der Vorst
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany (E.P.C.v.d.V., C.W.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany (E.P.C.v.d.V., C.W.).,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (C.W.).,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.W.)
| |
Collapse
|
25
|
Pareek S, Traboulsi H, Allard B, Rico de Souza A, Eidelman DH, Baglole CJ. Pulmonary neutrophilia caused by absence of the NF-κB member RelB is dampened by exposure to cigarette smoke. Mol Immunol 2019; 114:395-409. [PMID: 31476634 DOI: 10.1016/j.molimm.2019.08.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/23/2019] [Accepted: 08/14/2019] [Indexed: 01/01/2023]
Abstract
Inflammation is a response to injury and infection. Although protective under physiological conditions, excessive and persistent inflammation is linked to numerous diseases. As the lungs are continuously exposed to the external environment, the respiratory system is particularly liable to damage from inflammation. RelB is a member of the non-canonical NF-κB pathway that may control lung inflammation caused by cigarette smoke (CS), a leading cause of morbidity and mortality worldwide. Our lab has previously shown that RelB protects against CS-induced inflammation in vitro, leading us to hypothesize that RelB would protect against acute CS-induced pulmonary inflammation in vivo. We exposed wild-type (Relb+/+) and RelB-deficient mice (Relb-/-) mice to room air or to CS and found that CS exposure caused a sustained decrease in pulmonary granulocytes in Relb-/- mice that was predominated by a decrease in neutrophils. Pulmonary inflammation caused by other irritants, including chlorine, ovalbumin (OVA; to mimic features of asthma) and lipopolysaccharide (LPS) was not controlled by RelB. Differential cytokine analysis suggests that alterations in chemotactic cytokines do not fully account for the CS-specific decrease in neutrophils in Relb-/- mice. Flow cytometric analysis of the bronchoalveolar lavage and bone marrow cells also reveal that it is unlikely that the sustained decrease is caused by excessive cell death or decreased hematopoiesis from the bone marrow. Overall, our results indicate that RelB regulates acute CS-induced pulmonary inflammation. Understanding how RelB regulates CS-induced inflammation may potentiate the discovery of new therapeutic strategies for many of the inflammatory diseases caused by CS.
Collapse
Affiliation(s)
- Swati Pareek
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Departments of Pathology, McGill University, Montreal, Quebec, Canada
| | - Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Benoit Allard
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Medicine, McGill University, Montreal, Quebec, Canada
| | - Angela Rico de Souza
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Carolyn J Baglole
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Departments of Pathology, McGill University, Montreal, Quebec, Canada; Medicine, McGill University, Montreal, Quebec, Canada; Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
26
|
Xu SJ, Hu HT, Li HL, Chang S. The Role of miRNAs in Immune Cell Development, Immune Cell Activation, and Tumor Immunity: With a Focus on Macrophages and Natural Killer Cells. Cells 2019; 8:cells8101140. [PMID: 31554344 PMCID: PMC6829453 DOI: 10.3390/cells8101140] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) is the primary arena where tumor cells and the host immune system interact. Bidirectional communication between tumor cells and the associated stromal cell types within the TME influences disease initiation and progression, as well as tumor immunity. Macrophages and natural killer (NK) cells are crucial components of the stromal compartment and display either pro- or anti-tumor properties, depending on the expression of key regulators. MicroRNAs (miRNAs) are emerging as such regulators. They affect several immune cell functions closely related to tumor evasion of the immune system. This review discusses the role of miRNAs in the differentiation, maturation, and activation of immune cells as well as tumor immunity, focusing particularly on macrophages and NK cells.
Collapse
Affiliation(s)
- Shi Jun Xu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Hong Tao Hu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Hai Liang Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China.
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Suhwan Chang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea.
| |
Collapse
|
27
|
Ablation of miR-146b in mice causes hematopoietic malignancy. Blood Adv 2019; 2:3483-3491. [PMID: 30530754 DOI: 10.1182/bloodadvances.2018017954] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 10/21/2018] [Indexed: 12/23/2022] Open
Abstract
Excessive and constitutive activation of nuclear factor-κB (NF-κB) leads to abnormal cell proliferation and differentiation, leading to the development of malignant tumors, including lymphoma. MicroRNA 146a (miR-146a) and miR-146b, both of which carry an identical seed sequence, have been shown to contribute to inflammatory diseases and tumors by suppressing the expression of key molecules required for NF-κB activation. However, the functional and physiological differences between miR-146a and miR-146b in disease onset have not been fully elucidated. In this study, we generated miR-146b-knockout (KO) and miR-146a-KO mice by genome editing and found that both strains developed hematopoietic malignancies such as B-cell lymphoma and acute myeloid leukemia during aging. However, the B-cell lymphomas observed in miR-146a- and miR-146b-KO mice were histologically different in their morphology, and the malignancy rate is lower in miR-146b mice than miR-146a mice. Upon mitogenic stimulation, the expression of miR-146a and miR-146b was increased, but miR-146b expression was lower than that of miR-146a. Using a previously developed screening system for microRNA targets, we observed that miR-146a and miR-146b could target the same mRNAs, including TRAF6, and inhibit subsequent NF-κB activity. Consistent with these findings, both miR-146a- and miR-146b-KO B cells showed a high proliferative capacity. Taken together, sustained NF-κB activation in miR-146b KO mice could lead to the development of hematopoietic malignancy with aging.
Collapse
|
28
|
Duroux-Richard I, Robin M, Peillex C, Apparailly F. MicroRNAs: Fine Tuners of Monocyte Heterogeneity. Front Immunol 2019; 10:2145. [PMID: 31608049 PMCID: PMC6768098 DOI: 10.3389/fimmu.2019.02145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/27/2019] [Indexed: 01/13/2023] Open
Abstract
Small non-coding microRNAs (miRNAs) have been found to play critical roles in many biological processes by controlling gene expression at the post-transcriptional level. They appear to fine-tune the immune response by targeting key regulatory molecules, and their abnormal expression is associated with immune-mediated inflammatory disorders. Monocytes actively contribute to tissue homeostasis by triggering acute inflammatory reactions as well as the resolution of inflammation and tissue regeneration, in case of injury or pathogen invasion. Their contribution to tissue homeostasis can have many aspects because they are able to differentiate into different cell types including macrophages, dendritic cells, and osteoclasts, which fulfill functions as different as bone remodeling and immune response. Monocytes consist of different subsets with subset-specific expression of miRNAs linked to distinct biological processes dedicated to specific roles. Therefore, understanding the role of miRNAs in the context of monocyte heterogeneity may provide clues as to which subset gives rise to which cell type in tissues. In addition, because monocytes are involved in the pathogenesis of chronic inflammation, associated with loss of tissue homeostasis and function, identifying subset-specific miRNAs might help in developing therapeutic strategies that target one subset while sparing the others. Here, we give an overview of the state-of-the-art research regarding miRNAs that are differentially expressed between monocyte subsets and how they influence monocyte functional heterogeneity in health and disease, with descriptions of specific miRNAs. We also revisit the existing miRNome data to propose a canonical signature for each subset.
Collapse
Affiliation(s)
| | - Maxime Robin
- IRMB, INSERM, University of Montpellier, Montpellier, France
| | - Cindy Peillex
- IRMB, INSERM, University of Montpellier, Montpellier, France
| | - Florence Apparailly
- IRMB, INSERM, University of Montpellier, Montpellier, France
- Clinical Department for Osteoarticular Diseases, University Hospital of Montpellier, Montpellier, France
| |
Collapse
|
29
|
Wolf AA, Yáñez A, Barman PK, Goodridge HS. The Ontogeny of Monocyte Subsets. Front Immunol 2019; 10:1642. [PMID: 31379841 PMCID: PMC6650567 DOI: 10.3389/fimmu.2019.01642] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Classical and non-classical monocytes, and the macrophages and monocyte-derived dendritic cells they produce, play key roles in host defense against pathogens, immune regulation, tissue repair and many other processes throughout the body. Recent studies have revealed previously unappreciated heterogeneity among monocytes that may explain this functional diversity, but our understanding of mechanisms controlling the functional programming of distinct monocyte subsets remains incomplete. Resolving monocyte heterogeneity and understanding how their functional identity is determined holds great promise for therapeutic immune modulation. In this review, we examine how monocyte origins and developmental influences shape the phenotypic and functional characteristics of monocyte subsets during homeostasis and in the context of infection, inflammation, and cancer. We consider how extrinsic signals and transcriptional regulators impact monocyte production and functional programming, as well as the influence of epigenetic and metabolic mechanisms. We also examine the evidence that functionally distinct monocyte subsets are produced via different developmental pathways during homeostasis and that inflammatory stimuli differentially target progenitors during an emergency response. We highlight the need for a more comprehensive understanding of the relationship between monocyte ontogeny and heterogeneity, including multiparametric single-cell profiling and functional analyses. Studies defining mechanisms of monocyte subset production and maintenance of unique monocyte identities have the potential to facilitate the design of therapeutic interventions to target specific monocyte subsets in a variety of disease contexts, including infectious and inflammatory diseases, cancer, and aging.
Collapse
Affiliation(s)
- Anja A Wolf
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Alberto Yáñez
- Departament de Microbiologia i Ecologia, Universitat de València, Burjassot, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Pijus K Barman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Helen S Goodridge
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
30
|
Lu D, Yamawaki T, Zhou H, Chou WY, Chhoa M, Lamas E, Escobar SS, Arnett HA, Ge H, Juan T, Wang S, Li CM. Limited differential expression of miRNAs and other small RNAs in LPS-stimulated human monocytes. PLoS One 2019; 14:e0214296. [PMID: 30908559 PMCID: PMC6433273 DOI: 10.1371/journal.pone.0214296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/11/2019] [Indexed: 01/01/2023] Open
Abstract
Monocytes are a distinct subset of myeloid cells with diverse functions in early inflammatory immune modulation. While previous studies have surveyed the role of miRNA regulation on different myeloid cell lines and primary cultures, the time-dependent kinetics of inflammatory stimulation on miRNA expression and the relationship between miRNA-to-target RNA expression have not been comprehensively profiled in monocytes. In this study, we use next-generation sequencing and RT-PCR assays to analyze the non-coding small RNA transcriptome of unstimulated and lipopolysaccharide (LPS)-stimulated monocytes at 6 and 24 hours. We identified a miRNA signature consisting of five mature miRNAs (hsa-mir-146a, hsa-mir-155, hsa-mir-9, hsa-mir-147b, and hsa-mir-193a) upregulated by LPS-stimulated monocytes after 6 hours and found that most miRNAs were also upregulated after 24 hours of stimulation. Only one miRNA gene was down-regulated and no other small RNAs were found dysregulated in monocytes after LPS treatment. In addition, novel tRNA-derived fragments were also discovered in monocytes although none showed significant changes upon LPS stimulation. Interrogation of validated miRNA targets by transcriptomic analysis revealed that absolute expression of most miRNA targets implicating in innate immune response decreased over time in LPS-stimulated monocytes although their expression patterns along the treatment were heterogeneous. Our findings reveal a potential role by which selective miRNA upregulation and stable expression of other small RNAs enable monocytes to develop finely tuned cellular responses during acute inflammation.
Collapse
Affiliation(s)
- Daniel Lu
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Tracy Yamawaki
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Hong Zhou
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Wen-Yu Chou
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Mark Chhoa
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Edwin Lamas
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Sabine S. Escobar
- Inflammation/Oncology TA, Amgen Research, South San Francisco, California, United States of America
| | - Heather A. Arnett
- Inflammation/Oncology TA, Amgen Research, South San Francisco, California, United States of America
| | - Huanying Ge
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Todd Juan
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Songli Wang
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Research, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
31
|
Yin S, Ji C, Wu P, Jin C, Qian H. Human umbilical cord mesenchymal stem cells and exosomes: bioactive ways of tissue injury repair. Am J Transl Res 2019; 11:1230-1240. [PMID: 30972158 PMCID: PMC6456565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/10/2019] [Indexed: 06/09/2023]
Abstract
Mesenchymal stem cells (MSCs) can be recruited to damaged tissues directly for regeneration. Exosomes, acting as an important ingredient of MSCs-involved intercellular communication through paracrine actions, also play significant roles in tissue damage repair and have a prospect of potential clinical application. It is generally recognized that MSC-derived exosomes (MSC-exosomes) enhance tissue regeneration and repair through reducing inflammatory responses, promoting proliferation, inhibiting apoptosis and facilitating angiogenesis. This review summarizes the positive effects of human umbilical cord mesenchymal stem cells (hucMSCs) and hucMSC-derived exosomes (hucMSC-exosomes) on tissue damage and the specific mechanisms of repair action.
Collapse
Affiliation(s)
- Siqi Yin
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Cheng Ji
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Peipei Wu
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Can Jin
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Hui Qian
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
32
|
Pischiutta F, Sammali E, Parolini O, Carswell HVO, Zanier ER. Placenta-Derived Cells for Acute Brain Injury. Cell Transplant 2019; 27:151-167. [PMID: 29562781 PMCID: PMC6434489 DOI: 10.1177/0963689717732992] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute brain injury resulting from ischemic/hemorrhagic or traumatic damage is one of the leading causes of mortality and disability worldwide and is a significant burden to society. Neuroprotective options to counteract brain damage are very limited in stroke and traumatic brain injury (TBI). Given the multifaceted nature of acute brain injury and damage progression, several therapeutic targets may need to be addressed simultaneously to interfere with the evolution of the injury and improve the patient’s outcome. Stem cells are ideal candidates since they act on various mechanisms of protection and repair, improving structural and functional outcomes after experimental stroke or TBI. Stem cells isolated from placenta offer advantages due to their early embryonic origin, ease of procurement, and ethical acceptance. We analyzed the evidence for the beneficial effects of placenta-derived stem cells in acute brain injury, with the focus on experimental studies of TBI and stroke, the engineering strategies pursued to foster cell potential, and characterization of the bioactive molecules secreted by placental cells, known as their secretome, as an alternative cell-free strategy. Results from the clinical application of placenta-derived stem cells for acute brain injury and ongoing clinical trials are summarily discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Eliana Sammali
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.,2 Department of Cerebrovascular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ornella Parolini
- 3 Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy.,4 Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Hilary V O Carswell
- 5 Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, United Kingdom
| | - Elisa R Zanier
- 1 Department of Neuroscience, Laboratory of Acute Brain Injury and Therapeutic Strategies, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
33
|
Chipont A, Esposito B, Challier I, Montabord M, Tedgui A, Mallat Z, Loyer X, Potteaux S. MicroRNA-21 Deficiency Alters the Survival of Ly-6C
lo
Monocytes in
ApoE
−/−
Mice and Reduces Early-Stage Atherosclerosis—Brief Report. Arterioscler Thromb Vasc Biol 2019; 39:170-177. [DOI: 10.1161/atvbaha.118.311942] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective—
To determine the role of microRNA-21 (miR-21) on the homeostasis of monocyte subsets and on atherosclerosis development in
ApoE
−/−
(apolipoprotein E) mice.
Approach and Results—
In
ApoE
−/−
mice, miR-21 expression was increased in circulating Ly-6C
lo
nonclassical monocytes in comparison to Ly-6C
hi
monocytes. The absence of miR-21 significantly altered the survival and number of circulating Ly-6C
lo
nonclassical monocytes in
ApoE
−/−
mice. In the early stages of atherosclerosis, the absence of miR-21 limited lesion development both in the aortic sinus (by almost 30%) and in the aorta (by almost 50%). This was associated with less monocyte availability in circulation and increased apoptosis of local macrophages in plaques. At later stages of atherosclerosis, lesion size in the aortic root was similar in
ApoE
−/−
and
ApoE
−/−
miR-21
−/−
mice, but plaques showed a less stable phenotype (larger necrotic cores) in the latter. The loss of protection in advanced stages was most likely because of excessive inflammatory apoptosis related to an impairment of local efficient efferocytosis.
Conclusions—
Gene deletion of miR-21 in
ApoE
−/−
mice alters Ly-6C
lo
nonclassical monocytes homeostasis and contribute to limit early-stage atherosclerosis.
Collapse
Affiliation(s)
- Anna Chipont
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Bruno Esposito
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Inès Challier
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Mélanie Montabord
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Alain Tedgui
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Ziad Mallat
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Xavier Loyer
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| | - Stephane Potteaux
- From the Inserm U970, Paris Cardiovascular Research Center (PARCC), Université René Descartes Paris 5, France
| |
Collapse
|
34
|
Tonacci A, Bagnato G, Pandolfo G, Billeci L, Sansone F, Conte R, Gangemi S. MicroRNA Cross-Involvement in Autism Spectrum Disorders and Atopic Dermatitis: A Literature Review. J Clin Med 2019; 8:jcm8010088. [PMID: 30646527 PMCID: PMC6352260 DOI: 10.3390/jcm8010088] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 12/27/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a category of neurodevelopmental disturbances seriously affecting social skills, to which the scientific community has paid great attention in last decades. To date, their pathogenesis is still unknown, but several studies highlighted the relevance of gene-environment interactions in the onset of ASD. In addition, an immune involvement was seen in a wide number of ASD subjects, leading several researchers to hypothesize a possible common pathogenesis between ASD and immune disturbances, including Atopic Dermatitis (AD). In general, among potential contributing factors, microRNAs (miRNAs), small molecules capable of controlling gene expression and targeting mRNA transcripts, might represent one of the major circulating link, possibly unraveling the connections between neurodevelopmental and immune conditions. Under such premises, we conducted a systematic literature review, under the PRISMA guidelines, trying to define the panel of common miRNAs involved in both ASD and AD. The review retrieved articles published between January 1, 2005, and December 13, 2018, in PubMed, ScienceDirect, PsycARTICLES, and Google Scholar. We found a handful of works dealing with miRNAs in ASD and AD, with the most overlapping dysregulated miRNAs being miR-146 and miR-155. Two possible compounds are abnormally regulated in both ASD and AD subjects, possibly cross-contributing to the interactions between the two disorders, setting the basis to investigate more precisely the possible link between ASD and AD from another, not just clinical, perspective.
Collapse
Affiliation(s)
- Alessandro Tonacci
- Clinical Physiology Institute-National Research Council of Italy (IFC-CNR), Via Moruzzi 1, 56124 Pisa, Italy.
| | - Gianluca Bagnato
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino", Via Consolare Valeria SNC, 98125 Messina, Italy.
| | - Gianluca Pandolfo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| | - Lucia Billeci
- Clinical Physiology Institute-National Research Council of Italy (IFC-CNR), Via Moruzzi 1, 56124 Pisa, Italy.
| | - Francesco Sansone
- Clinical Physiology Institute-National Research Council of Italy (IFC-CNR), Via Moruzzi 1, 56124 Pisa, Italy.
| | - Raffaele Conte
- Clinical Physiology Institute-National Research Council of Italy (IFC-CNR), Via Moruzzi 1, 56124 Pisa, Italy.
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino", Via Consolare Valeria SNC, 98125 Messina, Italy.
| |
Collapse
|
35
|
A miR-150/TET3 pathway regulates the generation of mouse and human non-classical monocyte subset. Nat Commun 2018; 9:5455. [PMID: 30575719 PMCID: PMC6303340 DOI: 10.1038/s41467-018-07801-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 11/19/2018] [Indexed: 12/30/2022] Open
Abstract
Non-classical monocyte subsets may derive from classical monocyte differentiation and the proportion of each subset is tightly controlled. Deregulation of this repartition is observed in diverse human diseases, including chronic myelomonocytic leukemia (CMML) in which non-classical monocyte numbers are significantly decreased relative to healthy controls. Here, we identify a down-regulation of hsa-miR-150 through methylation of a lineage-specific promoter in CMML monocytes. Mir150 knock-out mice demonstrate a cell-autonomous defect in non-classical monocytes. Our pulldown experiments point to Ten-Eleven-Translocation-3 (TET3) mRNA as a hsa-miR-150 target in classical human monocytes. We show that Tet3 knockout mice generate an increased number of non-classical monocytes. Our results identify the miR-150/TET3 axis as being involved in the generation of non-classical monocytes.
Collapse
|
36
|
Ammari M, Presumey J, Ponsolles C, Roussignol G, Roubert C, Escriou V, Toupet K, Mausset-Bonnefont AL, Cren M, Robin M, Georgel P, Nehmar R, Taams L, Grün J, Grützkau A, Häupl T, Pers YM, Jorgensen C, Duroux-Richard I, Courties G, Apparailly F. Delivery of miR-146a to Ly6C high Monocytes Inhibits Pathogenic Bone Erosion in Inflammatory Arthritis. Am J Cancer Res 2018; 8:5972-5985. [PMID: 30613275 PMCID: PMC6299444 DOI: 10.7150/thno.29313] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
Rationale: Monocytes play critical roles in the pathogenesis of arthritis by contributing to the inflammatory response and bone erosion. Among genes involved in regulating monocyte functions, miR-146a negatively regulates the inflammatory response and osteoclast differentiation of monocytes. It is also the only miRNA reported to differentially regulate the cytokine response of the two classical Ly6Chigh and non-classical Ly6Clow monocyte subsets upon bacterial challenge. Although miR-146a is overexpressed in many tissues of arthritic patients, its specific role in monocyte subsets under arthritic conditions remains to be explored. Methods: We analyzed the monocyte subsets during collagen-induced arthritis (CIA) development by flow cytometry. We quantified the expression of miR-146a in classical and non-classical monocytes sorted from healthy and CIA mice, as well as patients with rheumatoid arthritis (RA). We monitored arthritis features in miR-146a-/- mice and assessed in vivo the therapeutic potential of miR-146a mimics delivery to Ly6Chigh monocytes. We performed transcriptomic and pathway enrichment analyses on both monocyte subsets sorted from wild type and miR-146a-/- mice. Results: We showed that the expression of miR-146a is reduced in the Ly6Chigh subset of CIA mice and in the analogous monocyte subset (CD14+CD16-) in humans with RA as compared with healthy controls. The ablation of miR-146a in mice worsened arthritis severity, increased osteoclast differentiation in vitro and bone erosion in vivo. In vivo delivery of miR-146a to Ly6Chigh monocytes, and not to Ly6Clow monocytes, rescues bone erosion in miR-146a-/- arthritic mice and reduces osteoclast differentiation and pathogenic bone erosion in CIA joints of miR-146a+/+ mice, with no effect on inflammation. Silencing of the non-canonical NF-κB family member RelB in miR-146a-/- Ly6Chigh monocytes uncovers a role for miR-146a as a key regulator of the differentiation of Ly6Chigh, and not Ly6Clow, monocytes into osteoclasts under arthritic conditions. Conclusion: Our results show that classical monocytes play a critical role in arthritis bone erosion. They demonstrate the theranostics potential of manipulating miR-146a expression in Ly6Chigh monocytes to prevent joint destruction while sparing inflammation in arthritis.
Collapse
|
37
|
Dayan N, Schlosser K, Stewart DJ, Delles C, Kaur A, Pilote L. Circulating MicroRNAs Implicate Multiple Atherogenic Abnormalities in the Long-Term Cardiovascular Sequelae of Preeclampsia. Am J Hypertens 2018; 31:1093-1097. [PMID: 29800045 PMCID: PMC6132124 DOI: 10.1093/ajh/hpy069] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/10/2018] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Women who have had preeclampsia (PE) are at increased risk for premature cardiovascular disease (CVD). The underlying pathophysiology of this risk remains unclear, but potentially involves subclinical vascular damage or dysfunction. Alterations in the levels of circulating microRNAs may be implicated, as they are known to play pervasive roles in vascular biology. We investigated whether levels of circulating microRNAs are altered between women with premature acute coronary syndrome (ACS), with and without a history of PE. METHODS Women with premature ACS (age ≤ 55 years) were categorized based on a prior history of PE or normotensive pregnancy. Relative plasma levels of 372 microRNAs were initially assessed by polymerase chain reaction array in a subset of subjects (n = 12–13/group) matched for age, chronic hypertension, dyslipidemia, and smoking status. Candidate microRNAs were then validated in a larger cohort of ACS patients (n = 176). RESULTS MicroRNAs previously linked to angiogenesis (miR-126-3p), inflammation (miR-146a-5p), and cholesterol metabolism (miR-122-5p) were significantly decreased in women with prior PE compared to women with prior normotensive pregnancy (P = 0.002, 0.017, and 0.009, respectively), even after adjustment for chronic hypertension. CONCLUSIONS Circulating levels of miR-126-3p, -146a-5p, and -122-5p were significantly decreased in women with premature ACS who reported prior PE compared to those with prior normotensive pregnancy. These data provide novel insight into potential pathways that may contribute to the increased risk of CVD following PE.
Collapse
Affiliation(s)
- Natalie Dayan
- Department of Medicine, Division of General Internal Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Kenny Schlosser
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Duncan J Stewart
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, UK
| | - Amanpreet Kaur
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - Louise Pilote
- Department of Medicine, Division of General Internal Medicine, McGill University Health Centre, Montreal, Quebec, Canada
- Research Institute, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
38
|
MicroRNAs and immunity in periodontal health and disease. Int J Oral Sci 2018; 10:24. [PMID: 30078842 PMCID: PMC6080405 DOI: 10.1038/s41368-018-0025-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are critical regulators of the host immune and inflammatory response against bacterial pathogens. In the present review, we discuss target genes, target gene functions, the potential regulatory role of miRNAs in periodontal tissues, and the potential role of miRNAs as biomarkers and therapeutics. In periodontal disease, miRNAs exert control over all aspects of innate and adaptive immunity, including the functions of neutrophils, macrophages, dendritic cells and T and B cells. Previous human studies have highlighted some key miRNAs that are dysregulated in periodontitis patients. In the present study, we mapped the major miRNAs that were altered in our reproducible periodontitis mouse model relative to control animals. The miRNAs that were upregulated as a result of periodontal disease in both human and mouse studies included miR-15a, miR-29b, miR-125a, miR-146a, miR-148/148a and miR-223, whereas miR-92 was downregulated. The association of individual miRNAs with unique aspects of periodontal disease and their stability in gingival crevicular fluid underscores their potential as markers for periodontal disease progression or healthy restitution. Moreover, miRNA therapeutics hold great promise for the future of periodontal therapy because of their ability to modulate the immune response to infection when applied in conjunction with synthetic antagomirs and/or relatively straightforward delivery strategies.
Collapse
|
39
|
Cho S, Lee HM, Yu IS, Choi YS, Huang HY, Hashemifar SS, Lin LL, Chen MC, Afanasiev ND, Khan AA, Lin SW, Rudensky AY, Crotty S, Lu LF. Differential cell-intrinsic regulations of germinal center B and T cells by miR-146a and miR-146b. Nat Commun 2018; 9:2757. [PMID: 30013024 PMCID: PMC6048122 DOI: 10.1038/s41467-018-05196-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 06/22/2018] [Indexed: 11/24/2022] Open
Abstract
Reciprocal interactions between B and follicular T helper (Tfh) cells orchestrate the germinal center (GC) reaction, a hallmark of humoral immunity. Abnormal GC responses could lead to the production of pathogenic autoantibodies and the development of autoimmunity. Here we show that miR-146a controls GC responses by targeting multiple CD40 signaling pathway components in B cells; by contrast, loss of miR-146a in T cells does not alter humoral responses. However, specific deletion of both miR-146a and its paralog, miR-146b, in T cells increases Tfh cell numbers and enhanced GC reactions. Thus, our data reveal differential cell-intrinsic regulations of GC B and Tfh cells by miR-146a and miR-146b. Together, members of the miR-146 family serve as crucial molecular brakes to coordinately control GC reactions to generate protective humoral responses without eliciting unwanted autoimmunity. In the germinal center (GC), B and T cells interact to induce the production of protective antibodies against threats. Here the authors show that microRNA miR-146a modulates CD40 signaling in GC B cells, while both miR-146a and miR-146b synergize to control GC T cell responses, thereby implicating intricate controls of GC response by miR-146.
Collapse
Affiliation(s)
- Sunglim Cho
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Hyang-Mi Lee
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - I-Shing Yu
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Youn Soo Choi
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037, USA.,Department of Medicine, College of Medicine, Seoul National University, Seoul, 03080, Korea
| | - Hsi-Yuan Huang
- Department of Laboratory Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | | | - Ling-Li Lin
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Mei-Chi Chen
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Nikita D Afanasiev
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | | | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Ludwig Center at Memorial Sloan-Kettering Cancer Center, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037, USA.,Division of Infectious Diseases, Department of Medicine, University of California, La Jolla, San Diego, CA, 92037, USA
| | - Li-Fan Lu
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA. .,Moores Cancer Center, University of California, La Jolla, San Diego, CA, 92093, USA. .,Center for Microbiome Innovation, University of California, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
40
|
Ma AJ, Zhu XY, Yang SN, Pan XD, Wang T, Wang Y, Xiao X, Liu SH. Associations of CXCL16, miR‑146a and miR‑146b in atherosclerotic apolipoprotein E‑knockout mice. Mol Med Rep 2018; 18:2995-3002. [PMID: 30015963 DOI: 10.3892/mmr.2018.9270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/14/2018] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. Recent studies have revealed that C‑X‑C motif chemokine ligand 16 (CXCL16), microRNA (miR)‑146a and miR‑146b may have important roles in atherosclerotic diseases. However, the associations of CXCL16, miR‑146a and miR‑146b in atherosclerotic diseases in vivo remain unclear. Previous studies have demonstrated that miR‑146a and miR‑146b may negatively regulate the toll like receptor (TLR4)/nuclear factor (NF)‑κB signaling pathway to repress the inflammatory response. The present study investigated the associations of CXCL16, miR‑146a and miR‑146b in atherosclerotic apolipoprotein E (ApoE)‑/‑ mice in vivo. The expression levels of CXCL16, TLR4/NF‑κB signaling pathway, miR‑146a and miR‑146b in the control and atherosclerotic ApoE‑/‑ mice were investigated via reverse transcription‑quantitative polymerase chain reaction and western blot analysis. The present study demonstrated that the expression of CXCL16 was significantly upregulated in atherosclerotic ApoE‑/‑ mice compared with control ApoE‑/‑ mice. The expression levels of TRL4, interleukin‑1 receptor‑associated kinase 1, tumor necrosis factor receptor associated factor 6, NF‑κB, tumor necrosis factor‑α and interleukin‑1β were also significantly upregulated in atherosclerotic ApoE‑/‑ mice compared with control mice. However, the present study revealed that the expression levels of miR‑146a and miR‑146b were significantly downregulated in atherosclerotic ApoE‑/‑ mice compared with control ApoE‑/‑ mice. Overall, the results of the present study suggested that CXCL16 may regulate the TRL4/NF‑κB/CXCL16 signaling pathway, and that miR‑146a and miR‑146b may negatively regulate CXCL16 via this pathway in atherosclerosis in vivo.
Collapse
Affiliation(s)
- Ai-Jun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Xiao-Yan Zhu
- Department of Critical Care Medicine, The Affiliated Hiser Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Shao-Nan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Xu-Dong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Ting Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Yuan Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Xing Xiao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| | - Shi-Hai Liu
- Medical Animal Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266033, P.R. China
| |
Collapse
|
41
|
Apolipoprotein E and Atherosclerosis: From Lipoprotein Metabolism to MicroRNA Control of Inflammation. J Cardiovasc Dev Dis 2018; 5:jcdd5020030. [PMID: 29789495 PMCID: PMC6023389 DOI: 10.3390/jcdd5020030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022] Open
Abstract
Apolipoprotein (apo) E stands out among plasma apolipoproteins through its unprecedented ability to protect against atherosclerosis. Although best recognized for its ability to mediate plasma lipoprotein clearance in the liver and protect against macrophage foam cell formation, our recent understanding of the influence that apoE can exert to control atherosclerosis has significantly widened. Among apoE’s newfound athero-protective properties include an ability to control exaggerated hematopoiesis, blood monocyte activation and aortic stiffening in mice with hyperlipidemia. Mechanisms responsible for these exciting new properties extend beyond apoE’s ability to prevent cellular lipid excess. Rather, new findings have revealed a role for apoE in regulating microRNA-controlled cellular signaling in cells of the immune system and vascular wall. Remarkably, infusions of apoE-responsive microRNA mimics were shown to substitute for apoE in protecting against systemic and vascular inflammation to suppress atherosclerosis in mice with hyperlipidemia. Finally, more recent evidence suggests that apoE may control the release of microvesicles that could modulate cellular signaling, inflammation and atherosclerosis at a distance. These exciting new findings position apoE within the emerging field of intercellular communication that could introduce new approaches to control atherosclerosis cardiovascular disease.
Collapse
|
42
|
Natural Compounds as Epigenetic Regulators of Human Dendritic Cell-mediated Immune Function. J Immunother 2018; 41:169-180. [DOI: 10.1097/cji.0000000000000201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
43
|
Du L, Chen X, Duan Z, Liu C, Zeng R, Chen Q, Li M. MiR-146a negatively regulates dectin-1-induced inflammatory responses. Oncotarget 2018; 8:37355-37366. [PMID: 28454101 PMCID: PMC5514914 DOI: 10.18632/oncotarget.16958] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/29/2017] [Indexed: 01/16/2023] Open
Abstract
Dectin-1 is the critical sensor for β-glucan from Candida which is the most common human fungal pathogen and cause superficial and system infection. MicroRNAs (miRNAs) play crucial roles in regulating innate immunity. However, the functional role of miRNAs in inflammatory response dependent on the activation of dectin-1 pathway has not been defined. In the present study, we found insoluble β-glucan from the cell wall of Candida albicans (CaIG) was able to increase the production of of IL-6 and TNFα through Dectin-1-Syk-NF-κB and p38MAPK pathway. MiRNAs profiles combined with real-time PCR validation revealed that miR-146a, miR-30-5p, miR-210-3p expression level were increased in THP-1 cells treated with CaIG. The interaction between Dectin-1 and CaIG resulted in an long lasting increase of miR-146a expression dependent on Dectin-1-Syk-NF-κB, p38MAPK, contrasting with a rapid and transient increase of IL-6 and TNFα. Overexpression of miR-146a significantly suppressed the production of IL-6 and TNFα. MiR-146a mimics inhibited CaIG-induced activity of p-IκBα and translocation of NF-κB p65. Luciferase reporter assays showed miR-146a inhibited NF-κB promoter-binding activity. Together, our data suggest miR-146a may play the potent negative feedback regulator in inflammatory response following Dectin-1 stimulation.
Collapse
Affiliation(s)
- Leilei Du
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Xu Chen
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Zhimin Duan
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Caixia Liu
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Rong Zeng
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| | - Qing Chen
- Jiangsu Province Blood Center, Nanjing, Jiangsu 210042, China
| | - Min Li
- From Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science and Peking Union Medical College, Nanjing 210042, China
| |
Collapse
|
44
|
Heuslein JL, McDonnell SP, Song J, Annex BH, Price RJ. MicroRNA-146a Regulates Perfusion Recovery in Response to Arterial Occlusion via Arteriogenesis. Front Bioeng Biotechnol 2018; 6:1. [PMID: 29404323 PMCID: PMC5786509 DOI: 10.3389/fbioe.2018.00001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/03/2018] [Indexed: 01/09/2023] Open
Abstract
The growth of endogenous collateral arteries that bypass arterial occlusion(s), or arteriogenesis, is a fundamental shear stress-induced adaptation with implications for treating peripheral arterial disease. MicroRNAs (miRs) are key regulators of gene expression in response to injury and have strong therapeutic potential. In a previous study, we identified miR-146a as a candidate regulator of vascular remodeling. Here, we tested whether miR-146a regulates in vitro angiogenic endothelial cell (EC) behaviors, as well as perfusion recovery, arteriogenesis, and angiogenesis in response to femoral arterial ligation (FAL) in vivo. We found miR-146a inhibition impaired EC tube formation and migration in vitro. Following FAL, Balb/c mice were treated with a single, intramuscular injection of anti-miR-146a or scramble locked nucleic acid (LNA) oligonucleotides directly into the non-ischemic gracilis muscles. Serial laser Doppler imaging demonstrated that anti-miR-146a treated mice exhibited significantly greater perfusion recovery (a 16% increase) compared mice treated with scramble LNA. Moreover, anti-miR-146a treated mice exhibited a 22% increase in collateral artery diameter compared to controls, while there was no significant effect on in vivo angiogenesis or muscle regeneration. Despite exerting no beneficial effects on angiogenesis, the inhibition of mechanosensitive miR-146a enhances perfusion recovery after FAL via enhanced arteriogenesis.
Collapse
Affiliation(s)
- Joshua L Heuslein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Stephanie P McDonnell
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Ji Song
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Brian H Annex
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
45
|
Menezes S, Melandri D, Anselmi G, Perchet T, Loschko J, Dubrot J, Patel R, Gautier EL, Hugues S, Longhi MP, Henry JY, Quezada SA, Lauvau G, Lennon-Duménil AM, Gutiérrez-Martínez E, Bessis A, Gomez-Perdiguero E, Jacome-Galarza CE, Garner H, Geissmann F, Golub R, Nussenzweig MC, Guermonprez P. The Heterogeneity of Ly6C hi Monocytes Controls Their Differentiation into iNOS + Macrophages or Monocyte-Derived Dendritic Cells. Immunity 2017; 45:1205-1218. [PMID: 28002729 PMCID: PMC5196026 DOI: 10.1016/j.immuni.2016.12.001] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 06/30/2016] [Accepted: 10/24/2016] [Indexed: 12/17/2022]
Abstract
Inflammation triggers the differentiation of Ly6Chi monocytes into microbicidal macrophages or monocyte-derived dendritic cells (moDCs). Yet, it is unclear whether environmental inflammatory cues control the polarization of monocytes toward each of these fates or whether specialized monocyte progenitor subsets exist before inflammation. Here, we have shown that naive monocytes are phenotypically heterogeneous and contain an NR4A1- and Flt3L-independent, CCR2-dependent, Flt3+CD11c−MHCII+PU.1hi subset. This subset acted as a precursor for FcγRIII+PD-L2+CD209a+, GM-CSF-dependent moDCs but was distal from the DC lineage, as shown by fate-mapping experiments using Zbtb46. By contrast, Flt3−CD11c−MHCII−PU.1lo monocytes differentiated into FcγRIII+PD-L2−CD209a−iNOS+ macrophages upon microbial stimulation. Importantly, Sfpi1 haploinsufficiency genetically distinguished the precursor activities of monocytes toward moDCs or microbicidal macrophages. Indeed, Sfpi1+/− mice had reduced Flt3+CD11c−MHCII+ monocytes and GM-CSF-dependent FcγRIII+PD-L2+CD209a+ moDCs but generated iNOS+ macrophages more efficiently. Therefore, intercellular disparities of PU.1 expression within naive monocytes segregate progenitor activity for inflammatory iNOS+ macrophages or moDCs. Murine Ly6ChiCD115+ monocytes are heterogeneous DC-related genes (Cd209a and MHCII) are expressed in a subset of FcγRIII+ monocytes GM-CSF-dependent CD209a+ moDCs are generated by FcγRIII+CD209a+MHCII+ monocytes iNOS+ macrophages are generated by FcγRIII+CD209a−MHCII− monocytes
Collapse
Affiliation(s)
- Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK
| | - Daisy Melandri
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK
| | | | | | | | - Rajen Patel
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK
| | | | | | - M Paula Longhi
- Barts and the London School of Medicine, EC1M 6BQ London, UK
| | | | | | - Grégoire Lauvau
- Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Enrique Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK
| | | | | | | | - Hannah Garner
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, King's College London, SE1 1UL London, UK; Centre for Inflammation Biology and Cancer Immunology, King's College London, SE1 1UL London, UK; Peter Gorer Department of Immunobiology, King's College London, SE1 1UL London, UK.
| |
Collapse
|
46
|
Abstract
Monocytes are short-lived mononuclear phagocytes that circulate in the bloodstream and comprise two main subpopulations that in the mouse are best defined by the Ly6C marker. Intravascular functions of "classical" Ly6C+ monocytes and their interactions with other lymphoid and myeloid leukocytes in the circulation remain poorly understood. Rather, these cells are known to efficiently extravasate into tissues. Indeed, Ly6C+ monocytes and their descendants have emerged as a third, highly plastic and dynamic cellular system that complements the two classical, tissue-resident mononuclear phagocyte compartments, i.e., macrophages and dendritic cells, on demand. Following recruitment to injured tissue, Ly6C+ monocytes respond to local cues and can critically contribute to the initiation and resolution of inflammatory reactions. The second main murine monocyte subset, Ly6C- cells, derive in steady state from Ly6C+ monocytes and remain in the vasculature, where the cells act as scavengers. Moreover, a major fraction of Ly6C- monocytes adheres to the capillary endothelium and patrols the vessel wall for surveillance. Given the central role of monocytes in homeostasis and pathology, in-depth study of this cellular compartment can be highly informative on the health state of the organism and provides an attractive target for therapeutic intervention.
Collapse
|
47
|
Begalli F, Bennett J, Capece D, Verzella D, D'Andrea D, Tornatore L, Franzoso G. Unlocking the NF-κB Conundrum: Embracing Complexity to Achieve Specificity. Biomedicines 2017; 5:E50. [PMID: 28829404 PMCID: PMC5618308 DOI: 10.3390/biomedicines5030050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/04/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Transcription factors of the nuclear factor κB (NF-κB) family are central coordinating regulators of the host defence responses to stress, injury and infection. Aberrant NF-κB activation also contributes to the pathogenesis of some of the most common current threats to global human health, including chronic inflammatory diseases, autoimmune disorders, diabetes, vascular diseases and the majority of cancers. Accordingly, the NF-κB pathway is widely considered an attractive therapeutic target in a broad range of malignant and non-malignant diseases. Yet, despite the aggressive efforts by the pharmaceutical industry to develop a specific NF-κB inhibitor, none has been clinically approved, due to the dose-limiting toxicities associated with the global suppression of NF-κB. In this review, we summarise the main strategies historically adopted to therapeutically target the NF-κB pathway with an emphasis on oncology, and some of the emerging strategies and newer agents being developed to pharmacologically inhibit this pathway.
Collapse
Affiliation(s)
- Federica Begalli
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Jason Bennett
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daria Capece
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daniela Verzella
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daniel D'Andrea
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Laura Tornatore
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Guido Franzoso
- Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
48
|
Abstract
As the largest receptor gene family in the human genome, with >800 members, the signal-transducing G protein-coupled receptors (GPCRs) play critical roles in nearly all conceivable physiological processes, ranging from the sensing of photons and odorants to metabolic homeostasis and migration of leukocytes. Unfortunately, an exhaustive review of the several hundred GPCRs expressed by myeloid cells/macrophages (P.J. Groot-Kormelink, L .Fawcett, P.D. Wright, M. Gosling, and T.C. Kent, BMC Immunol 12:57, 2012, doi:10.1186/1471-2172-13-57) is beyond the scope of this chapter; however, we will endeavor to cover the GPCRs that contribute to the major facets of macrophage biology, i.e., those whose expression is restricted to macrophages and the GPCRs involved in macrophage differentiation/polarization, microbial elimination, inflammation and resolution, and macrophage-mediated pathology. The chemokine receptors, a major group of myeloid GPCRs, will not be extensively covered as they are comprehensively reviewed elsewhere.
Collapse
|
49
|
Cheng HS, Besla R, Li A, Chen Z, Shikatani EA, Nazari-Jahantigh M, Hammoutène A, Nguyen MA, Geoffrion M, Cai L, Khyzha N, Li T, MacParland SA, Husain M, Cybulsky MI, Boulanger CM, Temel RE, Schober A, Rayner KJ, Robbins CS, Fish JE. Paradoxical Suppression of Atherosclerosis in the Absence of microRNA-146a. Circ Res 2017. [PMID: 28637783 PMCID: PMC5542783 DOI: 10.1161/circresaha.116.310529] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Inflammation is a key contributor to atherosclerosis. MicroRNA-146a (miR-146a) has been identified as a critical brake on proinflammatory nuclear factor κ light chain enhancer of activated B cells signaling in several cell types, including endothelial cells and bone marrow (BM)-derived cells. Importantly, miR-146a expression is elevated in human atherosclerotic plaques, and polymorphisms in the miR-146a precursor have been associated with risk of coronary artery disease. OBJECTIVE To define the role of endogenous miR-146a during atherogenesis. METHODS AND RESULTS Paradoxically, Ldlr-/- (low-density lipoprotein receptor null) mice deficient in miR-146a develop less atherosclerosis, despite having highly elevated levels of circulating proinflammatory cytokines. In contrast, cytokine levels are normalized in Ldlr-/-;miR-146a-/- mice receiving wild-type BM transplantation, and these mice have enhanced endothelial cell activation and elevated atherosclerotic plaque burden compared with Ldlr-/- mice receiving wild-type BM, demonstrating the atheroprotective role of miR-146a in the endothelium. We find that deficiency of miR-146a in BM-derived cells precipitates defects in hematopoietic stem cell function, contributing to extramedullary hematopoiesis, splenomegaly, BM failure, and decreased levels of circulating proatherogenic cells in mice fed an atherogenic diet. These hematopoietic phenotypes seem to be driven by unrestrained inflammatory signaling that leads to the expansion and eventual exhaustion of hematopoietic cells, and this occurs in the face of lower levels of circulating low-density lipoprotein cholesterol in mice lacking miR-146a in BM-derived cells. Furthermore, we identify sortilin-1(Sort1), a known regulator of circulating low-density lipoprotein levels in humans, as a novel target of miR-146a. CONCLUSIONS Our study reveals that miR-146a regulates cholesterol metabolism and tempers chronic inflammatory responses to atherogenic diet by restraining proinflammatory signaling in endothelial cells and BM-derived cells.
Collapse
Affiliation(s)
- Henry S Cheng
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Rickvinder Besla
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Angela Li
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Zhiqi Chen
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Eric A Shikatani
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Maliheh Nazari-Jahantigh
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Adel Hammoutène
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - My-Anh Nguyen
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Michele Geoffrion
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Lei Cai
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Nadiya Khyzha
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Tong Li
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Sonya A MacParland
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Mansoor Husain
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Myron I Cybulsky
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Chantal M Boulanger
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Ryan E Temel
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Andreas Schober
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Katey J Rayner
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Clinton S Robbins
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.)
| | - Jason E Fish
- From the Toronto General Hospital Research Institute, University Health Network, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., S.A.M., M.H., M.I.C., C.S.R., J.E.F.); Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada (H.S.C, R.B., A.L., Z.C., E.A.S., N.K., M.H., M.I.C., C.S.R., J.E.F.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Germany (M.N.-J., A.S.); INSERM, Unit 970, Paris Cardiovascular Research Center-PARCC, France (A.H., C.M.B.); University of Ottawa Heart Institute, Ontario, Canada (M.-A.N., M.G., K.J.R.); and Pharmacology and Nutritional Sciences, University of Kentucky, Lexington (L.C., T.L., R.E.T.).
| |
Collapse
|
50
|
Zawada AM, Zhang L, Emrich IE, Rogacev KS, Krezdorn N, Rotter B, Fliser D, Devaux Y, Ziegler-Heitbrock L, Heine GH. Reprint of: MicroRNA profiling of human intermediate monocytes. Immunobiology 2017; 222:831-840. [PMID: 28578934 DOI: 10.1016/j.imbio.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 01/23/2023]
Abstract
Among the three human monocyte subsets, intermediate CD14++CD16+ monocytes have been characterized as particularly proinflammatory cells in experimental studies and as potential biomarkers of cardiovascular risk in clinical cohorts. To further substantiate the distinct role of intermediate monocytes within human monocyte heterogeneity, we assessed subset-specific expression of miRNAs as central epigenetic regulators of gene expression. We hypothesized that intermediate monocytes have a distinct miRNA profile compared to classical and non-classical monocytes. By using small RNA-seq we analyzed 662 miRNAs in the three monocyte subsets. We identified 38 miRNAs that are differentially expressed in intermediate monocytes compared to both classical and non-classical monocytes with a p value of <10-10, of which two miRNAs - miR-6087 (upregulated) and miR-150-5p (downregulated) - differed in their expression more than ten-fold. Pathway analysis of the 38 differentially expressed miRNAs linked intermediate monocytes to distinct biological processes such as gene regulation, cell differentiation, toll-like receptor signaling as well as antigen processing and presentation. Moreover, differentially expressed miRNAs were connected to those genes that we previously identified as markers of intermediate monocytes. In aggregation, we provide first genome-wide miRNA data in the context of monocyte heterogeneity, which substantiate the concept of monocyte trichotomy in human immunity. The identification of miRNAs that are specific for intermediate monocytes may allow to develop strategies, which particularly target this cell population while sparing the other two subsets.
Collapse
Affiliation(s)
- Adam M Zawada
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany
| | - Lu Zhang
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg
| | - Insa E Emrich
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany
| | - Kyrill S Rogacev
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany; University Heart Center Luebeck, Medical Clinic II (Cardiology/Angiology/Intensive Care Medicine), University Hospital Schleswig-Holstein, Luebeck, Germany
| | | | | | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, Luxembourg
| | - Loems Ziegler-Heitbrock
- EvA Study Center, Comprehensive Pneumology Center Helmholtz Zentrum Muenchen - German Research Center for Environmental Health, Gauting, Germany
| | - Gunnar H Heine
- Department of Internal Medicine IV, Saarland University Medical Center, Homburg, Germany.
| |
Collapse
|