51
|
MicroRNAs as Potential Biomarkers in Atherosclerosis. Int J Mol Sci 2019; 20:ijms20225547. [PMID: 31703274 PMCID: PMC6887712 DOI: 10.3390/ijms20225547] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a complex multifactorial disease that, despite advances in lifestyle management and drug therapy, remains to be the major cause of high morbidity and mortality rates from cardiovascular diseases (CVDs) in industrialized countries. Therefore, there is a great need in reliable diagnostic/prognostic biomarkers and effective treatment alternatives to reduce its burden. It was established that microRNAs (miRNAs/miRs), a class of non-coding single-stranded RNA molecules, can regulate the expression of genes at the post-transcriptional level and, accordingly, coordinate the cellular protein expression. Thus, they are involved not only in cell-specific physiological functions but also in the cellular and molecular mechanisms of human pathologies, including atherosclerosis. MiRNAs may be significant in the dysregulation that affects endothelial integrity, the function of vascular smooth muscle and inflammatory cells, and cellular cholesterol homeostasis that drives the initiation and growth of an atherosclerotic plaque. Besides, distinct expression patterns of several miRNAs are attributed to atherosclerotic and cardiovascular patients. In this article, the evidence indicating the multiple critical roles of miRNAs and their relevant molecular mechanisms related to atherosclerosis development and progression was reviewed. Moreover, the effects of miRNAs on atherosclerosis enabled to exploit them as novel diagnostic biomarkers and therapeutic targets that may lead to better management of atherosclerosis and CVDs.
Collapse
|
52
|
Seeree P, Janvilisri T, Kangsamaksin T, Tohtong R, Kumkate S. Downregulation of ABCA1 and ABCG1 transporters by simvastatin in cholangiocarcinoma cells. Oncol Lett 2019; 18:5173-5184. [PMID: 31612028 PMCID: PMC6781495 DOI: 10.3892/ol.2019.10874] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Disturbances in cholesterol homeostasis of the bile duct epithelium, including transport interruption and the hyperaccumulation of intracellular cholesterol can lead to the initiation and progression of cholangiocarcinoma (CCA). Statins, which are lipid-lowering drugs, have been previously documented to exhibit anti-cancer properties. The role of statins in CCA cell cholesterol transport through the expression and function of ATP-binding cassette (ABC) A1 and ABCG1 was investigated in the current study. In four CCA cell lines, ABCA1 and ABCG1 expression was identified. However, neither ABCG5 nor ABCG8 expression was observed. Immunocytochemistry revealed that the expression of ABCA1 was localized in the proximity of the nucleus, while ABCG1 was more dispersed throughout the cytoplasm of KKU-100 cells. A cholesterol efflux assay was performed using bodipy cholesterol, and the translocation of cholesterol via ABCA1 and ABCG1 to Apo-A1 and high density lipoprotein was confirmed, respectively. Simvastatin and atorvastatin demonstrated the inhibitory effects on CCA cell viability. A reduction in intracellular lipid level and a lower expression of ABCA1 and ABCG1 were observed in KKU-100 cells under simvastatin treatment. The pre-exposure of KKU-100 cells to cholesterol diminished the statin effect. Furthermore, when KKU-100 cells were pre-loaded with cholesterol, ABCA1 and ABCG1-mediated exports were unaffected even though they were treated with simvastatin. The results of the current study indicated the limitations of the use of statin in CCA therapy, particularly under hypercholesterolemia conditions.
Collapse
Affiliation(s)
- Pattaya Seeree
- Department of Biology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Thaned Kangsamaksin
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Supeecha Kumkate
- Department of Biology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
53
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
54
|
Pillay P, Vatish M, Duarte R, Moodley J, Mackraj I. Exosomal microRNA profiling in early and late onset preeclamptic pregnant women reflects pathophysiology. Int J Nanomedicine 2019; 14:5637-5657. [PMID: 31413567 PMCID: PMC6661992 DOI: 10.2147/ijn.s208865] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Preeclampsia is the leading cause of maternal and fetal mortality due to the inability to diagnose and treat the disorder early in pregnancy. This is attributed to the complex pathophysiology and unknown etiology of the disorder, which is modulated by several known and unknown factors. Exosomes have recently been implicated as possible mediators of the pathogenesis of preeclampsia, with, however, no evidence linking these nanovesicles to the pathophysiology of preeclampsia and its subtypes. Methods: To better understand the pathophysiological role of exosomes in preeclampsia, we have analyzed the exosomal microRNA in early and late onset preeclamptic women in comparison to their gestationally matched normotensive controls using Digital Direct Detection (NanoString Technologies). Results: For the first time, distinct exosomal microRNA signatures in early and late onset preeclampsia have been identified. Moreover, these signatures indicate that exosomes are involved in key pathological features associated with preeclampsia and differentiate between the subtypes. Conclusion: This study forms the basis for the diagnostic and functional validation of the identified signatures as biomarkers of preeclampsia and its subtypes.
Collapse
Affiliation(s)
- Preenan Pillay
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of Kwazulu-Natal, Durban, South Africa
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Raquel Duarte
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jack Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, University of Kwazulu-Natal, Durban, South Africa
| | - Irene Mackraj
- School of Laboratory Medicine and Medical Sciences, University of Kwazulu-Natal, Durban, South Africa
| |
Collapse
|
55
|
Zusso M, Barbierato M, Facci L, Skaper SD, Giusti P. Neuroepigenetics and Alzheimer's Disease: An Update. J Alzheimers Dis 2019; 64:671-688. [PMID: 29991138 DOI: 10.3233/jad-180259] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epigenetics is the study of changes in gene expression which may be triggered by both genetic and environmental factors, and independent from changes to the underlying DNA sequence-a change in phenotype without a change in genotype-which in turn affects how cells read genes. Epigenetic changes represent a regular and natural occurrence but can be influenced also by factors such as age, environment, and disease state. Epigenetic modifications can manifest themselves not only as the manner in which cells terminally differentiate, but can have also deleterious effects, resulting in diseases such as cancer. At least three systems including DNA methylation, histone modification, and non-coding RNA (ncRNA)-associated gene silencing are thought to initiate and sustain epigenetic change. For example, in Alzheimer's disease (AD), both genetic and non-genetic factors contribute to disease etiopathology. While over 250 gene mutations have been related to familial AD, less than 5% of AD cases are explained by known disease genes. More than likely, non-genetic factors, probably triggered by environmental factors, are causative factors of late-onset AD. AD is associated with dysregulation of DNA methylation, histone modifications, and ncRNAs. Among the classes of ncRNA, microRNAs (miRNAs) have a well-established regulatory relevance. MicroRNAs are highly expressed in CNS neurons, where they play a major role in neuron differentiation, synaptogenesis, and plasticity. MicroRNAs impact higher cognitive functions, as their functional impairment is involved in the etiology of neurological diseases, including AD. Alterations in the miRNA network contribute to AD disease processes, e.g., in the regulation of amyloid peptides, tau, lipid metabolism, and neuroinflammation. MicroRNAs, both as biomarkers for AD and therapeutic targets, are in the early stages of exploration. In addition, emerging data suggest that altered transcription of long ncRNAs, endogenous, ncRNAs longer than 200 nucleotides, may be involved in an elevated risk for AD.
Collapse
Affiliation(s)
- Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti, Padua, Italy
| |
Collapse
|
56
|
Sharma B, Agnihotri N. Role of cholesterol homeostasis and its efflux pathways in cancer progression. J Steroid Biochem Mol Biol 2019; 191:105377. [PMID: 31063804 DOI: 10.1016/j.jsbmb.2019.105377] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/09/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022]
Abstract
Tumor cells show high avidity for cholesterol in order to support their inherent nature to divide and proliferate. This results in the rewiring of cholesterol homeostatic pathways by influencing not only de novo synthesis but also uptake or efflux pathways of cholesterol. Recent findings have pointed towards the importance of cholesterol efflux in tumor pathogenesis. Cholesterol efflux is the first and foremost step in reverse cholesterol transport and any perturbation in this pathway may lead to the accumulation of intracellular cholesterol, thereby altering the cellular equilibrium. This review addresses the different mechanisms of cholesterol efflux from the cell and highlights their role and regulation in context to tumor development. There are four different routes by which cholesterol can be effluxed from the cell namely, 1) passive diffusion of cholesterol to mature HDL particles, 2) SR-B1 mediated facilitated diffusion, 3) Active efflux to apo A1 via ABCA1 and 4) ABCG1 mediated efflux to mature HDL. These molecular players facilitating cholesterol efflux are engaged in a complex interplay with different signaling pathways. Thus, an understanding of the efflux pathways, their regulation and cross-talk with signaling molecules may provide novel prognostic markers and therapeutic targets to combat the onset of carcinogenesis.
Collapse
Affiliation(s)
- Bhoomika Sharma
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| | - Navneet Agnihotri
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| |
Collapse
|
57
|
Overexpression of miR-758 inhibited proliferation, migration, invasion, and promoted apoptosis of non-small cell lung cancer cells by negatively regulating HMGB. Biosci Rep 2019; 39:BSR20180855. [PMID: 30446524 PMCID: PMC6340954 DOI: 10.1042/bsr20180855] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/04/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most fatal types of cancer with significant mortality and morbidity worldwide. MicroRNAs (miRs) have been confirmed to have positive functions in NSCLC. In the present study, we try to explore the role of miR-758 in proliferation, migration, invasion, and apoptosis of NSCLC cells by regulating high-mobility group box (HMGB) 3 (HMGB3.) NSCLC and adjacent tissues were collected. Reverse transcription quantitative PCR (RT-qPCR) was employed to detect expression of miR-758 and HMGB3 in NSCLC and adjacent tissues, in BEAS-2B cells and NSCLC cell lines. The targetted relationship between miR-758 and HMGB3 was identified by dual luciferase reporter gene assay. The effects of miR-758 on proliferation, migration, invasion, cell cycle, and apoptosis of A549 cells. MiR-758 expression was lower in NSCLC tissues, which was opposite to HMGB3 expression. The results also demonstrated that miR-758 can target HMGB3. The cells transfected with miR-758 mimic had decreased HMGB3 expression, proliferation, migration, and invasion, with more arrested cells in G1 phase and increased apoptosis. Our results supported that the overexpression of miR-758 inhibits proliferation, migration, and invasion, and promotes apoptosis of NSCLC cells by negative regulating HMGB2. The present study may provide a novel target for NSCLC treatment.
Collapse
|
58
|
Yu XH, Zhang DW, Zheng XL, Tang CK. Cholesterol transport system: An integrated cholesterol transport model involved in atherosclerosis. Prog Lipid Res 2018; 73:65-91. [PMID: 30528667 DOI: 10.1016/j.plipres.2018.12.002] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/30/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease (CVD), is closely associated with cholesterol accumulation in the arterial intima. Excessive cholesterol is removed by the reverse cholesterol transport (RCT) pathway, representing a major antiatherogenic mechanism. In addition to the RCT, other pathways are required for maintaining the whole-body cholesterol homeostasis. Thus, we propose a working model of integrated cholesterol transport, termed the cholesterol transport system (CTS), to describe body cholesterol metabolism. The novel model not only involves the classical view of RCT but also contains other steps, such as cholesterol absorption in the small intestine, low-density lipoprotein uptake by the liver, and transintestinal cholesterol excretion. Extensive studies have shown that dysfunctional CTS is one of the major causes for hypercholesterolemia and atherosclerosis. Currently, several drugs are available to improve the CTS efficiently. There are also several therapeutic approaches that have entered into clinical trials and shown considerable promise for decreasing the risk of CVD. In recent years, a variety of novel findings reveal the molecular mechanisms for the CTS and its role in the development of atherosclerosis, thereby providing novel insights into the understanding of whole-body cholesterol transport and metabolism. In this review, we summarize the latest advances in this area with an emphasis on the therapeutic potential of targeting the CTS in CVD patients.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta T2N 4N1, Canada
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical Research Experiment Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
59
|
Abnormally expressed miR-23b in Chinese Mongolian at high cardiovascular risk may contribute to monocyte/macrophage inflammatory reaction in atherosclerosis. Biosci Rep 2018; 38:BSR20180673. [PMID: 30314997 PMCID: PMC6240720 DOI: 10.1042/bsr20180673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/14/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background: The prevalence of coronary heart disease (CHD) appears to be high among Chinese Mongolians. MiR-23b has been proven to play a key role in atherosclerosis. The expression and role of miR-23b in the Mongolians at high cardiovascular risk were explored in the present study. Methods: Forty cases of blood samples from the Mongolians at high cardiovascular risk were enrolled in the present study. The expression of miR-23b was quantified by quantitative real-time PCR. To induce monocytes differentiation into macrophages, HP-1 cells were cultured with phorbol 12-myristate 13-acetate. The level of inflammatory markers was determined by the enzyme-linked immunosorbent assay. The interaction between miR-23b and A20 was explored by the dual luciferase reporter assay. Results: The expression of miR-23b in the Mongolian at high cardiovascular risk was higher than that in healthy Mongolian volunteers. Decrease in ATP-binding cassette transporter A1 caused by miR-23b is responsible for TC accumulation in the Mongolian at high cardiovascular risk. MiR-23b enhanced the oxidized low-density lipoprotein (oxLDL)-induced inflammatory response of THP-1 derived macrophage. MiR-23b regulated nuclear factor-κB (NF-κB) pathway through targeting A20. MiR-23b mediated oxLDL-induced inflammatory response of peripheral blood mononuclear cell in the Mongolian at high cardiovascular risk. Conclusion MiR-23b enhanced oxLDL-induced inflammatory response of macrophages in the Mongolian at high cardiovascular risk through the A20/NF-κB signaling pathway, and thus contributing to atherosclerosis.
Collapse
|
60
|
Donaldson CJ, Lao KH, Zeng L. The salient role of microRNAs in atherogenesis. J Mol Cell Cardiol 2018; 122:98-113. [DOI: 10.1016/j.yjmcc.2018.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
|
61
|
Maguire EM, Pearce SWA, Xiao Q. Foam cell formation: A new target for fighting atherosclerosis and cardiovascular disease. Vascul Pharmacol 2018; 112:54-71. [PMID: 30115528 DOI: 10.1016/j.vph.2018.08.002] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/17/2018] [Accepted: 08/03/2018] [Indexed: 12/23/2022]
Abstract
During atherosclerosis, the gradual accumulation of lipids into the subendothelial space of damaged arteries results in several lipid modification processes followed by macrophage uptake in the arterial wall. The way in which these modified lipoproteins are dealt with determines the likelihood of cholesterol accumulation within the monocyte-derived macrophage and thus its transformation into the foam cell that makes up the characteristic fatty streak observed in the early stages of atherosclerosis. The unique expression of chemokine receptors and cellular adhesion molecules expressed on the cell surface of monocytes points to a particular extravasation route that they can take to gain entry into atherosclerotic site, in order to undergo differentiation into the phagocytic macrophage. Indeed several GWAS and animal studies have identified key genes and proteins required for monocyte recruitment as well cholesterol handling involving lipid uptake, cholesterol esterification and cholesterol efflux. A re-examination of the previously accepted paradigm of macrophage foam cell origin has been called into question by recent studies demonstrating shared expression of scavenger receptors, cholesterol transporters and pro-inflammatory cytokine release by alternative cell types present in the neointima, namely; endothelial cells, vascular smooth muscle cells and stem/progenitor cells. Thus, therapeutic targets aimed at a more heterogeneous foam cell population with shared functions, such as enhanced protease activity, and signalling pathways, mediated by non-coding RNA molecules, may provide greater therapeutic outcome in patients. Finally, studies targeting each aspect of foam cell formation and death using both genetic knock down and pharmacological inhibition have provided researchers with a clearer understanding of the cellular processes at play, as well as helped researchers to identify key molecular targets, which may hold significant therapeutic potential in the future.
Collapse
Affiliation(s)
- Eithne M Maguire
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Stuart W A Pearce
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
62
|
Shao D, Lian Z, Di Y, Zhang L, Rajoka MSR, Zhang Y, Kong J, Jiang C, Shi J. Dietary compounds have potential in controlling atherosclerosis by modulating macrophage cholesterol metabolism and inflammation via miRNA. NPJ Sci Food 2018; 2:13. [PMID: 31304263 PMCID: PMC6550192 DOI: 10.1038/s41538-018-0022-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 06/12/2018] [Accepted: 06/28/2018] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis (AS) is a typical example of a widespread fatal cardiovascular disease. Accumulation of cholesterol-laden macrophages in the artery wall forms the starting point of AS. Increased influx of oxidized low-density lipoprotein to macrophages and decreased efflux of free cholesterol out of macrophages constitute major factors promoting the development of AS. Inflammation further aggravates the development of AS along or via interaction with the cholesterol metabolism. Many microRNAs (miRNAs) are related to the regulation of macrophage in AS in aspects of cholesterol metabolism and inflammation signaling. Dietary compounds perform AS inhibitory effects via miRNAs in the cholesterol metabolism (miR-19b, miR-378, miR-10b, miR-33a, and miR-33b) and two miRNAs in the inflammation signaling (miR-155 and miR-146a). The targeted miRNAs in the cholesterol metabolism vary greatly among different food compounds; however, in inflammation signaling, most food compounds target miR-155. Many receptors are involved in macrophages via miRNAs, including ABCA1 and ABCG1 as major receptors in the cholesterol metabolism, while nuclear factor-κB (NF-κB) and Nrf2 signaling and PI3K/AKT signaling pathways are targeted during inflammation. This article reviews current literature to investigate possible AS therapy with dietary compounds via targeting miRNAs. Currently existing problems were also discussed to guide further studies.
Collapse
Affiliation(s)
- Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Ziyang Lian
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Yichao Di
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Lei Zhang
- Department of Microbiology and Pathogeny Biology, Xi’an Medical University, 1 Xinwang Road, Xi’an, 710072 Shaanxi China
| | - Muhammad shahid riaz Rajoka
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Yudan Zhang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Jie Kong
- MOE Key Laboratory of Space Applied Physics and Chemistry, Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Science, Northwestern Polytechnical University, Xi’an, 710072 Shaanxi China
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi’an, 710072 Shaanxi China
| |
Collapse
|
63
|
Chalcone Derivatives Enhance ATP-Binding Cassette Transporters A1 in Human THP-1 Macrophages. Molecules 2018; 23:molecules23071620. [PMID: 29970865 PMCID: PMC6100038 DOI: 10.3390/molecules23071620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/20/2018] [Accepted: 06/29/2018] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis is a process of imbalanced lipid metabolism in the vascular walls. The underlying pathology mainly involves the deposition of oxidized lipids in the endothelium and the accumulation of cholesterol in macrophages. Macrophages export excessive cholesterol (cholesterol efflux) through ATP-binding cassette transporter A1 (ABCA1) to counter the progression of atherosclerosis. We synthesized novel chalcone derivatives and assessed their effects and the underlying mechanisms on ABCA1 expression in macrophages. Human THP-1 macrophages were treated with synthetic chalcone derivatives for 24 h. In Western blot and flow cytometry analyses, a chalcone derivative, (E)-1-(3,4-diisopropoxyphenyl)-3-(4-isopropoxy-3-methoxyphenyl)prop- 2-en-1-one (1m), was observed to significantly enhance ABCA1 protein expression in THP-1 cells (10 µM, 24 h). Levels of mRNA of ABCA1 and liver X receptor alpha (LXRα) were quantified using a real-time quantitative polymerase chain reaction technique and were found to be significantly increased after treatment with the novel chalcone derivative 1m. Several microRNAs, including miR155, miR758, miR10b, miR145, miR33, and miR106b, which functionally inhibit ABCA1 expression were suppressed after treatment with 1m. Collectively, 1m increases ABCA1 expression in human THP-1 macrophages. The mechanisms involve the activation of the LXRα-ABCA1 pathway and suppression of certain microRNAs that regulate ABCA1 expression.
Collapse
|
64
|
Das A, Samidurai A, Salloum FN. Deciphering Non-coding RNAs in Cardiovascular Health and Disease. Front Cardiovasc Med 2018; 5:73. [PMID: 30013975 PMCID: PMC6036139 DOI: 10.3389/fcvm.2018.00073] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/29/2018] [Indexed: 12/16/2022] Open
Abstract
After being long considered as “junk” in the human genome, non-coding RNAs (ncRNAs) currently represent one of the newest frontiers in cardiovascular disease (CVD) since they have emerged in recent years as potential therapeutic targets. Different types of ncRNAs exist, including small ncRNAs that have fewer than 200 nucleotides, which are mostly known as microRNAs (miRNAs), and long ncRNAs that have more than 200 nucleotides. Recent discoveries on the role of ncRNAs in epigenetic and transcriptional regulation, atherosclerosis, myocardial ischemia/reperfusion (I/R) injury and infarction (MI), adverse cardiac remodeling and hypertrophy, insulin resistance, and diabetic cardiomyopathy prompted vast interest in exploring candidate ncRNAs for utilization as potential therapeutic targets and/or diagnostic/prognostic biomarkers in CVDs. This review will discuss our current knowledge concerning the roles of different types of ncRNAs in cardiovascular health and disease and provide some insight on the cardioprotective signaling pathways elicited by the non-coding genome. We will highlight important basic and clinical breakthroughs that support employing ncRNAs for treatment or early diagnosis of a variety of CVDs, and also depict the most relevant limitations that challenge this novel therapeutic approach.
Collapse
Affiliation(s)
- Anindita Das
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Arun Samidurai
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Fadi N Salloum
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
65
|
Zhang X, Price NL, Fernández-Hernando C. Non-coding RNAs in lipid metabolism. Vascul Pharmacol 2018; 114:93-102. [PMID: 29929012 DOI: 10.1016/j.vph.2018.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/01/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD), the leading cause of death and morbidity in the Western world, begins with lipid accumulation in the arterial wall, which is the initial step in atherogenesis. Alterations in lipid metabolism result in increased risk of cardiometabolic disorders, and treatment of lipid disorders remains the most common strategy aimed at reducing the incidence of CVD. Work done over the past decade has identified numerous classes of non-coding RNA molecules including microRNAs (miRNAs) and long-non-coding RNAs (lncRNAs) as critical regulators of gene expression involved in lipid metabolism and CVD, mostly acting at post-transcriptional level. A number of miRNAs, including miR-33, miR-122 and miR-148a, have been demonstrated to play important role in controlling the risk of CVD through regulation of cholesterol homeostasis and lipoprotein metabolism. lncRNAs are recently emerging as important regulators of lipid and lipoprotein metabolism. However, much additional work will be required to fully understand the impact of lncRNAs on CVD and lipid metabolism, due to the high abundance of lncRNAs and the poor-genetic conservation between species. This article reviews the role of miRNAs and lncRNAs in lipid and lipoprotein metabolism and their potential implications for the treatment of CVD.
Collapse
Affiliation(s)
- Xinbo Zhang
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA.
| |
Collapse
|
66
|
Liu X, Garban J, Jones PJ, Vanden Heuvel J, Lamarche B, Jenkins DJ, Connelly PW, Couture P, Pu S, Fleming JA, West SG, Kris-Etherton PM. Diets Low in Saturated Fat with Different Unsaturated Fatty Acid Profiles Similarly Increase Serum-Mediated Cholesterol Efflux from THP-1 Macrophages in a Population with or at Risk for Metabolic Syndrome: The Canola Oil Multicenter Intervention Trial. J Nutr 2018; 148:721-728. [PMID: 30053283 PMCID: PMC6669947 DOI: 10.1093/jn/nxy040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/09/2017] [Accepted: 02/12/2018] [Indexed: 01/02/2023] Open
Abstract
Background Cholesterol efflux plays an important role in preventing atherosclerosis progression. Vegetable oils with varying unsaturated fatty acid profiles favorably affect multiple cardiovascular disease risk factors; however, their effects on cholesterol efflux remain unclear. Objective The objectives of this study were to examine the effects of diets low in saturated fatty acids (SFAs) with varying unsaturated fatty acid profiles on serum-mediated cholesterol efflux and its association with the plasma lipophilic index and central obesity. Methods The present study is a randomized, crossover, controlled-feeding study. Participants [men: n = 50; women: n = 51; mean ± SE age: 49.5 ± 1.2 y; body mass index (in kg/m2): 29.4 ± 0.4] at risk for or with metabolic syndrome (MetS) were randomly assigned to 5 isocaloric diets containing the treatment oils: canola oil, high oleic acid-canola oil, DHA-enriched high oleic acid-canola oil, corn oil and safflower oil blend, and flax oil and safflower oil blend. These treatment oils were incorporated into smoothies that participants consumed 2 times/d. For a 3000-kcal diet, 60 g of treatment oil was required to provide 18% of total energy per day. Each diet period was 4 wk followed by a 2- to 4-wk washout period. We quantified cholesterol efflux capacity with a validated ex vivo high-throughput cholesterol efflux assay. Statistical analyses were performed with the use of the SAS mixed-model procedure. Results The 5 diets increased serum-mediated cholesterol efflux capacity from THP-1 macrophages similarly by 39%, 34%, 55%, 49% and 51%, respectively, compared with baseline (P < 0.05 for all). Waist circumference and abdominal adiposity were negatively correlated with serum-mediated cholesterol efflux capacity (r = -0.25, P = 0.01, r = -0.33, P = 0.02, respectively). Conclusion Diets low in SFAs with different monounsaturated fatty acid and polyunsaturated fatty acid profiles improved serum-mediated cholesterol efflux capacity in individuals with or at risk for MetS. This mechanism may account, in part, for the cardiovascular disease benefits of diets low in SFAs and high in unsaturated fatty acids. Importantly, central obesity is inversely associated with cholesterol efflux capacity. This trial was registered at www.clinicaltrials.gov as NCT01351012.
Collapse
Affiliation(s)
- Xiaoran Liu
- Departments of Nutritional Sciences, Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Josephine Garban
- Departments of Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Peter J Jones
- Richardson Center for Functional Foods and Nutraceuticals, University of Manitoba, Winnipeg, Canada
| | - Jack Vanden Heuvel
- Departments of Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Benoît Lamarche
- Institute of Nutrition and Functional Foods, Laval University, Québec, Canada
| | - David J Jenkins
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Philip W Connelly
- Keenan Research Centre for Biomedical Science of St Michael's Hospital, Toronto, Canada
| | - Patrick Couture
- Institute of Nutrition and Functional Foods, Laval University, Québec, Canada
| | - Shuaihua Pu
- Departments of Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Jennifer A Fleming
- Departments of Nutritional Sciences, Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Sheila G West
- Departments of Nutritional Sciences, Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| | - Penny M Kris-Etherton
- Departments of Nutritional Sciences, Veterinary and Biomedical Sciences, and Biobehavioral Health, The Pennsylvania State University, University Park, PA
| |
Collapse
|
67
|
Zambrano T, Hirata RD, Hirata MH, Cerda Á, Salazar LA. Statins differentially modulate microRNAs expression in peripheral cells of hyperlipidemic subjects: A pilot study. Eur J Pharm Sci 2018; 117:55-61. [DOI: 10.1016/j.ejps.2018.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 01/27/2018] [Accepted: 02/06/2018] [Indexed: 01/13/2023]
|
68
|
Glucagon-like peptide-1 contributes to increases ABCA1 expression by downregulating miR-758 to regulate cholesterol homeostasis. Biochem Biophys Res Commun 2018; 497:652-658. [DOI: 10.1016/j.bbrc.2018.02.126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 02/14/2018] [Indexed: 12/17/2022]
|
69
|
Chu M, Zhao Y, Yu S, Hao Y, Zhang P, Feng Y, Zhang H, Ma D, Liu J, Cheng M, Li L, Shen W, Cao H, Li Q, Min L. MicroRNA-221 may be involved in lipid metabolism in mammary epithelial cells. Int J Biochem Cell Biol 2018; 97:118-127. [PMID: 29474925 DOI: 10.1016/j.biocel.2018.02.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/09/2018] [Accepted: 02/15/2018] [Indexed: 12/27/2022]
Abstract
Milk lipids, important for infant growth and development, are produced and secreted by mammary gland under the regulation of steroid hormones, growth factors, and microRNAs (miRNAs). miR-221 has been identified in milk and adipocytes and it plays important roles in regulating normal mammary epithelial hierarchy and breast cancer stem cells; however, its roles in lipid metabolism in mammary epithelial cells (MECs), the cells of lipid synthesis and secretion, are as yet unknown. Through overexpression or inhibition of miR-221 expression, we found that it regulated lipid metabolism in MECs and was expressed differentially at various stages during murine mammary gland development. Inhibition of miR-221 expression increased lipid content in MECs through elevation of the lipid synthesis enzyme FASN, while overexpression of miR-221 reduced MEC lipid content. Moreover, the steroid hormones estradiol and progesterone decreased miR-221 expression with a subsequent increase in lipid formation in MECs. The expression of miR-221 was lower during lactation, which suggests that it may be involved in milk production. Therefore, miR-221 might be a useful target for influencing milk lipid production.
Collapse
Affiliation(s)
- Meiqiang Chu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yong Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Shuai Yu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yanan Hao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Pengfei Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yanni Feng
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Dongxue Ma
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jing Liu
- Core Laboratories of Qingdao Agricultural University, Qingdao 266109, PR China
| | - Ming Cheng
- Qingdao Veterinary and Livestock Administration, Qingdao, 266000, PR China
| | - Lan Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Wei Shen
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Hongfang Cao
- Laiwu Veterinary and Livestock Administration, Laiwu, 271100, PR China
| | - Qiang Li
- Laiwu Veterinary and Livestock Administration, Laiwu, 271100, PR China
| | - Lingjiang Min
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
70
|
O'Neill S, Larsen MB, Gregersen S, Hermansen K, O'Driscoll L. miR-758-3p: a blood-based biomarker that's influence on the expression of CERP/ABCA1 may contribute to the progression of obesity to metabolic syndrome. Oncotarget 2018; 9:9379-9390. [PMID: 29507696 PMCID: PMC5823618 DOI: 10.18632/oncotarget.24314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/02/2018] [Indexed: 01/22/2023] Open
Abstract
Due to increasing prevalence of obesity, a simple method or methods for the diagnosis of metabolic syndrome are urgently required to reduce the risk of associated cardiovascular disease, diabetes and cancer. This study aimed to identify a miRNA biomarker that may distinguish metabolic syndrome from obesity and to investigate if such a miRNA may have functional relevance for metabolic syndrome. 52 adults with clinical obesity (n=26) or metabolic syndrome (n=26) were recruited. Plasma specimens were procured from all and were randomly designated to discovery and validation cohorts. miRNA discovery profiling was performed, using array technology, on plasma RNA. Validation was performed by quantitative polymerase chain reaction. The functional effect of miR-758-3p on its predicted target, cholesterol efflux regulatory protein/ATP-binding cassette transporter, was investigated using HepG2 liver cells. Custom miRNA profiling of 25 miRNAs in the discovery cohort found miR-758-3p to be detected in the obese cohort but undetected in the metabolic syndrome cohort. miR-758-3p was subsequently validated as a potential biomarker for metabolic syndrome by quantitative polymerase chain reaction. Bioinformatics analysis identified cholesterol efflux regulatory protein/ATP-binding cassette transporter as miR-758-3p’s predicted target. Specifically, mimicking miR-758-3p in HepG2 cells suppressed cholesterol efflux regulatory protein/ATP-binding cassette transporter protein expression; conversely, inhibiting miR-758-3p increased cholesterol efflux regulatory protein/ATP-binding cassette transporter protein expression. miR-758-3p holds potential as a blood-based biomarker for distinguishing progression from obesity to metabolic syndrome and as a driver in controlling cholesterol efflux regulatory protein/ATP-binding cassette transporter expression, indicating it potential role in cholesterol control in metabolic syndrome.
Collapse
Affiliation(s)
- Sadhbh O'Neill
- School of Pharmacy & Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mette Bohl Larsen
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Søren Gregersen
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Kjeld Hermansen
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Lorraine O'Driscoll
- School of Pharmacy & Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
71
|
Abstract
Significant advancements have been made in unraveling and understanding the non-coding elements of the human genome. New insights into the structure and function of noncoding RNAs have emerged. Their relevance in the context of both physiological cellular homeostasis and human diseases is getting appreciated. As a result, exploration of noncoding RNAs, in particular microRNAs (miRs), as therapeutic agents or targets of therapeutic strategies is under way. This review summarizes and discusses in depth the current literature on the role of miRs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Salil Sharma
- Department of Psychological and Brain Sciences, The Linda and Jack Gill Center for Bimolecular Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, The Linda and Jack Gill Center for Bimolecular Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
72
|
Chu M, Zhao Y, Feng Y, Zhang H, Liu J, Cheng M, Li L, Shen W, Cao H, Li Q, Min L. MicroRNA-126 participates in lipid metabolism in mammary epithelial cells. Mol Cell Endocrinol 2017; 454:77-86. [PMID: 28599789 DOI: 10.1016/j.mce.2017.05.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/31/2017] [Accepted: 05/31/2017] [Indexed: 12/14/2022]
Abstract
Lipids are a major component of milk and are important for infant growth and development. MicroRNA-126 (miR-126) has previously been observed in mammary glands and adipocytes and is known to be involved in lipid metabolism during the process of atherosclerosis. However, it remains unknown whether miR-126 also participates in lipid metabolism in mammary luminal epithelial cells (MECs). In the current investigation, miR-126-3p inhibition stimulated lipid synthesis in MECs in part through increasing levels of the lipid synthesis enzymes FASN, ACSL1, and Insig1. Overexpression of miR-126-3p decreased lipid content in MECs with a reduction in FASN and Insig1. Furthermore, the expression of miR-126-3p was diminished by the steroid hormones estradiol and progesterone with a subsequent elevation of lipid formation in MECs. We also noted that miR-126-3p was expressed differentially at various stages of murine mammary gland development, exhibiting a negative correlation with FASN. Together these findings suggest that miR-126-3 might be involved in lipid metabolism in mammary gland.
Collapse
Affiliation(s)
- Meiqiang Chu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yong Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yanni Feng
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Jing Liu
- Core Laboratories of Qingdao Agricultural University, Qingdao 266109, PR China
| | - Ming Cheng
- Qingdao Veterinary and Livestock Administration, Qingdao 266000, PR China
| | - Lan Li
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Wei Shen
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Hongfang Cao
- Laiwu Veterinary and Livestock Administration, Laiwu 271100, PR China
| | - Qiang Li
- Laiwu Veterinary and Livestock Administration, Laiwu 271100, PR China
| | - Lingjiang Min
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
73
|
Jiang D, Cho WC, Li Z, Xu X, Qu Y, Jiang Z, Guo L, Xu G. MiR-758-3p suppresses proliferation, migration and invasion of hepatocellular carcinoma cells via targeting MDM2 and mTOR. Biomed Pharmacother 2017; 96:535-544. [PMID: 29032337 DOI: 10.1016/j.biopha.2017.10.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/12/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocelluar carcinoma (HCC) is one of the most frequently diagnosed cancers worldwide and among the leading causes of cancer-related death. Although deregulation of microRNAs has been frequently described in HCC, imperfection is known about the precise molecular mechanisms by which microRNAs modulate the process of tumorogenesis and behavior of cancer cells. In this study, we demonstrated that miR-758-3p could suppress cell proliferation, migration and invasion in hepatocellular carcinoma cells. We screened and identified two novel miR-758-3p targets, MDM2 and mTOR. Up-regulation of miR-758-3p could specifically and markedly down-regulate the expression of MDM2 and mTOR. Additionally, miR-758-3p over-expression displayed significant suppression in HCC development. To identify the mechanisms, we further investigated the P53 and mTOR pathway and found that p-p70S6 kinase(Ser371), p-p70 S6 kinase(Thr389) and p-4E-BP1were dramatically down-regulated after miR-758-3p transfection, while an enhanced expression of P53, AKT and PRAS40 were visualized, thus suggesting that the role of miR-758-3p in HCC progression may be associated with MDM2-p53 and mTOR signaling pathways. Collectively, our results indicate that miR-758-3pserves as a tumor suppressor and plays a crucial role in inhibiting the proliferation, migration and invasion of HCC via targeting MDM2 and mTOR and implicate its potential application in cancer therapy.
Collapse
Affiliation(s)
- Dan Jiang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| | - Zhenhao Li
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiangrong Xu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yuliang Qu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhongjia Jiang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Le Guo
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China
| | - Guangxian Xu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China; Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, Yinchuan, 750004, China.
| |
Collapse
|
74
|
miR-758-5p regulates cholesterol uptake via targeting the CD36 3'UTR. Biochem Biophys Res Commun 2017; 494:384-389. [PMID: 28965954 DOI: 10.1016/j.bbrc.2017.09.150] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 09/27/2017] [Indexed: 12/20/2022]
Abstract
miR-758-3p plays an important role via regulting ABCA1-mediated cholesterol efflux in atherosclerosis. However, the mechanism of miR-758-5p in cholesterol metabolism is still unclear. Here, we revealed that miR-758-5p decreased total cholesterol accumulation in THP-1 macrophage derived foam cells through markedly reducing cholesterol uptake, and no effect on the cholesterol efflux. Interestingly, computational analysis suggests that CD36 may be a target gene of miR-758-5p. Our study further demonstrated that miR-758-5p decreased CD36 expression at both protein and mRNA levels via targeting the CD36 3'UTR in THP-1 macrophage derived foam cells. The present present study concluded that miR-758-5p decreases lipid accumulation of foam cell via regulating CD36-mediated the cholesterol uptake. Therefore, targeting miR-758-5p may offer a promising strategy to treat atherosclerotic vascular disease.
Collapse
|
75
|
Meng X, Zhao Y, Wang J, Gao Z, Geng Q, Liu X. Regulatory roles of miRNA-758 and matrix extracellular phosphoglycoprotein in cervical cancer. Exp Ther Med 2017; 14:2789-2794. [PMID: 28928798 PMCID: PMC5590035 DOI: 10.3892/etm.2017.4887] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/03/2017] [Indexed: 01/03/2023] Open
Abstract
The present study aimed to examine the role and underlying mechanism of miRNA-758 (miR-758) expression in cancer tissues, blood and cervical exfoliated cells from patients with cervical cancer. A total of 49 patients with cervical cancer and 26 healthy people for cervical cancer screening were included in the present study. The patients with cervical cancer were treated with resection, and the tumor and adjacent tissues, blood and cervical exfoliated cells were collected. The expression levels of miR-758 and matrix extracellular phosphoglycoprotein (MEPE) mRNA in each sample were detected by reverse transcription-quantitative polymerase chain reaction. In addition, western blot analysis was used to detect the MEPE protein in tumor tissues, while ELISA was applied to detect the MEPE protein expression in the blood and cervical exfoliated cells. Compared with the normal control, MEPE mRNA expression was upregulated in cervical cancer tissues, blood and cervical exfoliated cells. At the protein level, MEPE was also upregulated significantly in patients with cervical cancer. miR-758 expression was decreased significantly in cervical cancer tissues, blood and cervical exfoliated cells (P<0.05), which was opposite to the trend observed for MEPE mRNA expression. Furthermore, MEPE expression was increased in the tumor tissue, blood and cervical exfoliated cells of cervical cancer patients, which was associated to the downregulated miR-758. Therefore, miR-758 may regulate the infiltration and invasion of cervical cancer by targeting MEPE.
Collapse
Affiliation(s)
- Xianhua Meng
- Department of Gynaecology and Obstetrics, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Yinghui Zhao
- Department of Gynaecology and Obstetrics, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Jinyun Wang
- Department of Gynaecology and Obstetrics, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Zheng Gao
- Department of Gynaecology and Obstetrics, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| | - Qingxia Geng
- Department of Gynaecology and Obstetrics, Laiwu Maternal and Child Care Hospital, Laiwu, Shandong 271100, P.R. China
| | - Xiaoxia Liu
- Department of Gynaecology and Obstetrics, Laiwu City People's Hospital, Laiwu, Shandong 271199, P.R. China
| |
Collapse
|
76
|
Abstract
Despite rapid advances in cardiovascular research and therapeutic strategies, ischemic heart disease (IHD) remains the leading cause of mortality worldwide. MicroRNAs (miRNAs) are small, noncoding RNAs which post transcriptionally regulate gene expression. In the past few years, miRNAs have emerged as key tools for the understanding of the pathophysiology of IHD, with potential uses as new biomarkers and therapeutic targets. Several studies report a regulatory role of miRNAs, with regard to fundamental components of IHD pathogenesis and progression, such as lipoprotein metabolism, atherogenesis, vascular calcification, platelet function, and angiogenesis. Due to their high stability in biofluids, circulating miRNAs have attracted attention as promising biomarkers of IHD, especially in cardiovascular risk prediction and the diagnosis of myocardial infarction. Furthermore, experimental studies have demonstrated the potential of miRNA-targeted therapy in improving hyperlipidemia, atherosclerosis, and angiogenesis. In this review, the current knowledge on the role of miRNAs in IHD and translational perspectives of their use is discussed.
Collapse
|
77
|
Liang B, Wang X, Song X, Bai R, Yang H, Yang Z, Xiao C, Bian Y. MicroRNA-20a/b regulates cholesterol efflux through post-transcriptional repression of ATP-binding cassette transporter A1. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:929-938. [PMID: 28602962 DOI: 10.1016/j.bbalip.2017.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/27/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022]
Abstract
ATP-binding cassette transporter A1 (ABCA1) plays a crucial role in reverse cholesterol transport and exhibits anti-atherosclerosis effects. Some microRNAs (miRs) regulate ABCA1 expression, and recent studies have shown that miR-20a/b might play a critical role in atherosclerotic diseases. Here, we attempted to clarify the potential contribution of miR-20a/b in post-transcriptional regulation of ABCA1, cholesterol efflux, and atherosclerosis. We performed bioinformatics analysis and found that miR-20a/b was highly conserved and directly bound to ABCA1 mRNA with low binding free energy. Luciferase-reporter assay also confirmed that miR-20a/b significantly reduced luciferase activity associated with the ABCA1 3' untranslated region reporter construct. Additionally, miR-20a/b decreased ABCA1 expression, which, in turn, decreased cholesterol efflux and increased cholesterol content in THP-1 and RAW 264.7 macrophage-derived foam cells. In contrast, miR-20a/b inhibitors increased ABCA1 expression and cholesterol efflux, decreased cholesterol content, and inhibited foam-cell formation. Consistent with our in vitro results, miR-20a/b-treated ApoE-/- mice showed decreased ABCA1expression in the liver and reductions of reverse cholesterol transport in vivo. Furthermore, miR-20a/b regulated the formation of nascent high-density lipoprotein and promoted atherosclerotic development, whereas miR-20a/b knockdown attenuated atherosclerotic formation. miR-20 is a new miRNA capable of targeting ABCA1 and regulating ABCA1 expression. Therefore, miR-20 inhibition constitutes a new strategy for ABCA1-based treatment of atherosclerosis.
Collapse
Affiliation(s)
- Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China
| | - Xin Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Xiaosu Song
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China
| | - Rui Bai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China
| | - Huiyu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China
| | - Zhiming Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China
| | - Chuanshi Xiao
- Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China; Department of Cardiology, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan, China
| | - Yunfei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China; Key Laboratory of Cardiovascular Medicine and Clinical Pharmacology of Shanxi Province, 382 Wuyi Road, Taiyuan, China.
| |
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW Work over the past decade has identified the important role of microRNAs (miRNAS) in regulating lipoprotein metabolism and associated disorders including metabolic syndrome, obesity, and atherosclerosis. This review summarizes the most recent findings in the field, highlighting the contribution of miRNAs in controlling LDL-cholesterol (LDL-C) and HDL-cholesterol (HDL-C) metabolism. RECENT FINDINGS A number of miRNAs have emerged as important regulators of lipid metabolism, including miR-122 and miR-33. Work over the past 2 years has identified additional functions of miR-33 including the regulation of macrophage activation and mitochondrial metabolism. Moreover, it has recently been shown that miR-33 regulates vascular homeostasis and cardiac adaptation in response to pressure overload. In addition to miR-33 and miR-122, recent GWAS have identified single-nucleotide polymorphisms in the proximity of miRNA genes associated with abnormal levels of circulating lipids in humans. Several of these miRNAs, such as miR-148a and miR-128-1, target important proteins that regulate cellular cholesterol metabolism, including the LDL receptor (LDLR) and the ATP-binding cassette A1 (ABCA1). SUMMARY MicroRNAs have emerged as critical regulators of cholesterol metabolism and promising therapeutic targets for treating cardiometabolic disorders including atherosclerosis. Here, we discuss the recent findings in the field, highlighting the novel mechanisms by which miR-33 controls lipid metabolism and atherogenesis, and the identification of novel miRNAs that regulate LDL metabolism. Finally, we summarize the recent findings that identified miR-33 as an important noncoding RNA that controls cardiovascular homeostasis independent of its role in regulating lipid metabolism.
Collapse
Affiliation(s)
- Binod Aryal
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Abhishek K. Singh
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Nathan Price
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, 10 Amistad St., New Haven, CT 06510. USA
- Corresponding author: Carlos Fernández-Hernando. Phone: +1 (203)-737-4615.
| |
Collapse
|
79
|
Ouimet M, Ediriweera H, Afonso MS, Ramkhelawon B, Singaravelu R, Liao X, Bandler RC, Rahman K, Fisher EA, Rayner KJ, Pezacki JP, Tabas I, Moore KJ. microRNA-33 Regulates Macrophage Autophagy in Atherosclerosis. Arterioscler Thromb Vasc Biol 2017; 37:1058-1067. [PMID: 28428217 DOI: 10.1161/atvbaha.116.308916] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Defective autophagy in macrophages leads to pathological processes that contribute to atherosclerosis, including impaired cholesterol metabolism and defective efferocytosis. Autophagy promotes the degradation of cytoplasmic components in lysosomes and plays a key role in the catabolism of stored lipids to maintain cellular homeostasis. microRNA-33 (miR-33) is a post-transcriptional regulator of genes involved in cholesterol homeostasis, yet the complete mechanisms by which miR-33 controls lipid metabolism are unknown. We investigated whether miR-33 targeting of autophagy contributes to its regulation of cholesterol homeostasis and atherogenesis. APPROACH AND RESULTS Using coherent anti-Stokes Raman scattering microscopy, we show that miR-33 drives lipid droplet accumulation in macrophages, suggesting decreased lipolysis. Inhibition of neutral and lysosomal hydrolysis pathways revealed that miR-33 reduced cholesterol mobilization by a lysosomal-dependent mechanism, implicating repression of autophagy. Indeed, we show that miR-33 targets key autophagy regulators and effectors in macrophages to reduce lipid droplet catabolism, an essential process to generate free cholesterol for efflux. Notably, miR-33 regulation of autophagy lies upstream of its known effects on ABCA1 (ATP-binding cassette transporter A1)-dependent cholesterol efflux, as miR-33 inhibitors fail to increase efflux upon genetic or chemical inhibition of autophagy. Furthermore, we find that miR-33 inhibits apoptotic cell clearance via an autophagy-dependent mechanism. Macrophages treated with anti-miR-33 show increased efferocytosis, lysosomal biogenesis, and degradation of apoptotic material. Finally, we show that treating atherosclerotic Ldlr-/- mice with anti-miR-33 restores defective autophagy in macrophage foam cells and plaques and promotes apoptotic cell clearance to reduce plaque necrosis. CONCLUSIONS Collectively, these data provide insight into the mechanisms by which miR-33 regulates cellular cholesterol homeostasis and atherosclerosis.
Collapse
Affiliation(s)
- Mireille Ouimet
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Hasini Ediriweera
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Milessa Silva Afonso
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Bhama Ramkhelawon
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Ragunath Singaravelu
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Xianghai Liao
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Rachel C Bandler
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Karishma Rahman
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Katey J Rayner
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - John P Pezacki
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Ira Tabas
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.).
| |
Collapse
|
80
|
Desgagné V, Bouchard L, Guérin R. microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application. Clin Chem Lab Med 2017; 55:667-686. [PMID: 27987357 DOI: 10.1515/cclm-2016-0575] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are short (~22 nucleotides), non-coding, single-stranded RNA molecules that regulate the expression of target genes by partial sequence-specific base-pairing to the targeted mRNA 3'UTR, blocking its translation, and promoting its degradation or its sequestration into processing bodies. miRNAs are important regulators of several physiological processes including developmental and metabolic functions, but their concentration in circulation has also been reported to be altered in many pathological conditions such as familial hypercholesterolemia, cardiovascular diseases, obesity, type 2 diabetes, and cancers. In this review, we focus on the role of miRNAs in lipoprotein and lipid metabolism, with special attention to the well-characterized miR-33a/b, and on the huge potential of miRNAs for clinical application as biomarkers and therapeutics in the context of cardiometabolic diseases.
Collapse
Affiliation(s)
| | - Luigi Bouchard
- Département de biochimie, Université de Sherbrooke, Sherbrooke, Québec
| | - Renée Guérin
- Département de biochimie, Université de Sherbrooke, Sherbrooke, Québec
| |
Collapse
|
81
|
Loh WP, Yang Y, Lam KP. miR-92a enhances recombinant protein productivity in CHO cells by increasing intracellular cholesterol levels. Biotechnol J 2017; 12. [PMID: 28146316 DOI: 10.1002/biot.201600488] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/27/2016] [Accepted: 01/31/2017] [Indexed: 12/27/2022]
Abstract
MicroRNAs (miRNAs) have emerged as promising targets for engineering of CHO cell factories to enhance recombinant protein productivity. Manipulation of miRNA levels in CHO cells have been shown to improve product yield by increasing proliferation and specific productivity (qP), resisting apoptosis and enhancing oxidative metabolism. The authors previously demonstrated that over-expressing miR-92a results in increases in qP and titer of CHO-IgG cells. However, the mechanisms by which miR-92a enhances qP in CHO cells are still uninvestigated. Here, the authors report the identification of insig1, a regulator of cholesterol biosynthesis, as a target of miR-92a using computational prediction. Both transient and stable over-expression of miR-92a decreased the expression levels of insig1. Insig1 was further validated as a target of miR-92a using 3' UTR reporter assay. Intracellular cholesterol concentration of two high-producing miR-92a clones were significantly increased by ≈30% compared to the blank-transfected pool. Relative Golgi surface area was also found to be 18-26% higher in these clones. Our findings suggest that miR-92a may affect cholesterol metabolism by repressing insig1, resulting in raised intracellular cholesterol levels and Golgi volume and hence enhanced protein secretion.
Collapse
Affiliation(s)
- Wan Ping Loh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kong Peng Lam
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
82
|
Gabunia K, Herman AB, Ray M, Kelemen SE, England RN, DeLa Cadena R, Foster WJ, Elliott KJ, Eguchi S, Autieri MV. Induction of MiR133a expression by IL-19 targets LDLRAP1 and reduces oxLDL uptake in VSMC. J Mol Cell Cardiol 2017; 105:38-48. [PMID: 28257760 DOI: 10.1016/j.yjmcc.2017.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.
Collapse
Affiliation(s)
- Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Raul DeLa Cadena
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - William J Foster
- Departments of Ophthalmology & Bioengineering, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Katherine J Elliott
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
83
|
Laffont B, Rayner KJ. MicroRNAs in the Pathobiology and Therapy of Atherosclerosis. Can J Cardiol 2017; 33:313-324. [PMID: 28232017 DOI: 10.1016/j.cjca.2017.01.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs are short noncoding RNAs, expressed in humans and involved in sequence-specific post-transcriptional regulation of gene expression. They have emerged as key players in a wide array of biological processes, and changes in their expression and/or function have been associated with plethora of human diseases. Atherosclerosis and its related clinical complications, such as myocardial infarction or stroke, represent the leading cause of death in the Western world. Accumulating experimental evidence has revealed a key role for microRNAs in regulating cellular and molecular processes related to atherosclerosis development, ranging from risk factors, to plaque initiation and progression, up to atherosclerotic plaque rupture. In this review, we focus on how microRNAs can influence atherosclerosis biology, as well as the potential clinical applications of microRNAs, which are being developed as targets as well as therapeutic agents for a growing industry hoping to harness the power of RNA-guided gene regulation to fight disease and infection.
Collapse
Affiliation(s)
- Benoit Laffont
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
84
|
Das S, Vasanthi HR, Parjapath R. MitomiRs Keep the Heart Beating. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:431-450. [PMID: 28551801 DOI: 10.1007/978-3-319-55330-6_23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this chapter, we focus on the microRNAs (miRNAs or miRs) that have been found in the mitochondrial compartment, and target either mitochondrial or nuclear encoded genes present in mitochondria, leading to an alteration of mitochondrial function. We term this subset of miRNAs as "MitomiRs".
Collapse
Affiliation(s)
- Samarjit Das
- Department of Pathology, Cardiovascular Division, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Hannah R Vasanthi
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Ramesh Parjapath
- Department of Biotechnology, Pondicherry University, Puducherry, India
| |
Collapse
|
85
|
|
86
|
The role of microRNAs in hepatocyte metabolism and hepatitis B virus replication. Virol Sin 2016; 31:472-479. [PMID: 28063013 DOI: 10.1007/s12250-016-3924-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/22/2016] [Indexed: 02/07/2023] Open
Abstract
Though efficient vaccines against hepatitis B virus (HBV) and antiviral therapies are available, chronic HBV infection is still a global health problem. The process of HBV infection and HBV life cycle are extensively studied in last decades, however, the mechanisms of HBV-induced alterations of host cell metabolisms and host factors involved in modulating of viral replication are not fully understood. Thus, it is an important issue to examine these specific HBV-host interactions for development of novel strategies for antiviral therapies. Recently, microRNAs (miRNAs), a class of post-transcriptional regulatory small RNA, seem to be the relevant fine tuning factors of various cellular activities and pathways, including cell growth, metabolism, and viral replication. In this review, we summarize the up to date knowledge concerning the virus-host interactions and emphasizing on the role of miRNAs in regulation of HBV replication and host cell metabolism.
Collapse
|
87
|
Lauschke VM, Mkrtchian S, Ingelman-Sundberg M. The role of microRNAs in liver injury at the crossroad between hepatic cell death and regeneration. Biochem Biophys Res Commun 2016; 482:399-407. [PMID: 27789285 DOI: 10.1016/j.bbrc.2016.10.084] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/19/2016] [Accepted: 10/23/2016] [Indexed: 02/07/2023]
Abstract
The liver fulfills critical metabolic functions, such as controlling blood sugar and ammonia levels, and is of central importance for lipid metabolism and detoxification of environmental and chemical agents, including drugs. Liver injuries of different etiology can elicit a spectrum of responses. Some hepatocytes initiate molecular programs resulting in cell death, whereas others undergo cellular divisions to regenerate the damaged organ. Interestingly, recent research indicates that microRNAs serve as very rapid as well as long-term regulators in these processes. In this review, we discuss their importance in liver disease etiology and progression as well as for therapy with particular focus on metabolic and inflammatory conditions. Furthermore, we highlight the central role of microRNAs in controlling hepatocyte differentiation and plasticity, which are required for successful regeneration, but under certain conditions, such as chronic liver insults, can result in the formation of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden.
| | - Souren Mkrtchian
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
88
|
Abstract
Numerous studies have examined the role of microRNAs (miRNAs) in cell homeostasis and cardiovascular disease and have markedly improved our understanding of RNA biology in general and the potential role of miRNAs in atherosclerosis. In atherosclerosis, several miRNAs, such as miR-33a,b, miR-92a, miR-126 and others, have been identified that are relevant mediators of pathological processes, including regulation of cholesterol and lipid biosynthesis, lipoprotein metabolism and cholesterol efflux, but also immune responses, endothelial cell biology and vascular function. Further understanding of the specific roles of miRNAs in the distinct cell types involved in atherosclerosis initiation, progression and resolution may reveal new intervention strategies for the prevention and treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hector Giral
- Department of Cardiology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Germany
| | - Adelheid Kratzer
- Department of Cardiology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany; Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), Germany.
| |
Collapse
|
89
|
Zhang X, Schulze PC. MicroRNAs in heart failure: Non-coding regulators of metabolic function. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2276-2287. [PMID: 27544699 DOI: 10.1016/j.bbadis.2016.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is the inability of the heart to provide sufficient cardiac output for the energy demands of the body. Over the last decades, our understanding of the role of microRNAs (miRNAs), a class of small non-coding RNA regulators of gene expression at the post-transcriptional level, in cardiovascular diseases has expanded at a rapid rate. Importantly, multiple miRNAs have been specifically implicated in the progression of HF. Growing evidence suggests that miRNAs regulate central metabolic pathways and thus are highly implicated in the maintenance of energy homeostasis. In this review, we highlight recent discoveries of the mechanistic role of miRNAs in regulating metabolic functions in HF, with specific focus on the implication of miRNAs in metabolic rearrangements, discuss the potential value of miRNA profiles as novel HF biomarkers, and summarize the recent investigations on therapeutic approaches using miRNAs in heart disease. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY, USA
| | - P Christian Schulze
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY, USA; Department of Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University Jena, University Hospital Jena, Jena, Germany.
| |
Collapse
|
90
|
Arrigoni E, Galimberti S, Petrini M, Danesi R, Di Paolo A. ATP-binding cassette transmembrane transporters and their epigenetic control in cancer: an overview. Expert Opin Drug Metab Toxicol 2016; 12:1419-1432. [PMID: 27459275 DOI: 10.1080/17425255.2016.1215423] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Members of the ATP-binding cassette (ABC) transmembrane transporters control the passage of several substrates across cell membranes, including drugs. This means that ABC transporters may exert a significant influence on the kinetics and dynamics of pharmacological agents, being responsible for the occurrence of multidrug-resistant (MDR) phenotype. Pharmacogenetic analyses have shed light on gene expression and polymorphisms as possible markers predictive of transporter activity. However, a non-negligible part of the variability in drug pharmacokinetics and pharmacodynamics still remains. Further research has demonstrated that different epigenetic mechanisms exert a coordinated control over ABC genes, and on the corresponding MDR phenotype. Areas covered: DNA methylation and histone modifications (namely acetylation, methylation, phosphorylation, etc.) significantly impact gene expression, as well as noncoding RNA molecules that are involved in the post-transcriptional control of the ABC transporters ABCB1, ABCC1 and ABCG2. We describe the epigenetic mechanisms of gene expression control for ABC transporters and their relevant association with the MDR phenotype in human cancer. Expert opinion: The clinical meaning of those observations is discussed in the review, highlighting the importance of the epigenetic control of the ABC transporters for the clinical therapeutic outcomes that despite their effects and applications, requires further investigation.
Collapse
Affiliation(s)
- Elena Arrigoni
- a Section of Pharmacology, Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Sara Galimberti
- b Section of Hematology, Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Mario Petrini
- b Section of Hematology, Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Romano Danesi
- a Section of Pharmacology, Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Antonello Di Paolo
- a Section of Pharmacology, Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
91
|
Lai L, Azzam KM, Lin WC, Rai P, Lowe JM, Gabor KA, Madenspacher JH, Aloor JJ, Parks JS, Näär AM, Fessler MB. MicroRNA-33 Regulates the Innate Immune Response via ATP Binding Cassette Transporter-mediated Remodeling of Membrane Microdomains. J Biol Chem 2016; 291:19651-60. [PMID: 27471270 DOI: 10.1074/jbc.m116.723056] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression by promoting degradation and/or repressing translation of specific target mRNAs. Several miRNAs have been identified that regulate the amplitude of the innate immune response by directly targeting Toll-like receptor (TLR) pathway members and/or cytokines. miR-33a and miR-33b (the latter present in primates but absent in rodents and lower species) are located in introns of the sterol regulatory element-binding protein (SREBP)-encoding genes and control cholesterol/lipid homeostasis in concert with their host gene products. These miRNAs regulate macrophage cholesterol by targeting the lipid efflux transporters ATP binding cassette (ABC)A1 and ABCG1. We and others have previously reported that Abca1(-/-) and Abcg1(-/-) macrophages have increased TLR proinflammatory responses due to augmented lipid raft cholesterol. Given this, we hypothesized that miR-33 would augment TLR signaling in macrophages via a raft cholesterol-dependent mechanism. Herein, we report that multiple TLR ligands down-regulate miR-33 in murine macrophages. In the case of lipopolysaccharide, this is a delayed, Toll/interleukin-1 receptor (TIR) domain-containing adapter-inducing interferon-β-dependent response that also down-regulates Srebf-2, the host gene for miR-33. miR-33 augments macrophage lipid rafts and enhances proinflammatory cytokine induction and NF-κB activation by LPS. This occurs through an ABCA1- and ABCG1-dependent mechanism and is reversible by interventions upon raft cholesterol and by ABC transporter-inducing liver X receptor agonists. Taken together, these findings extend the purview of miR-33, identifying it as an indirect regulator of innate immunity that mediates bidirectional cross-talk between lipid homeostasis and inflammation.
Collapse
Affiliation(s)
- Lihua Lai
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Kathleen M Azzam
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Wan-Chi Lin
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Prashant Rai
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Julie M Lowe
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Kristin A Gabor
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Jennifer H Madenspacher
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Jim J Aloor
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157
| | - Anders M Näär
- Massachusetts General Hospital Cancer Center, Charlestown, Massachusetts 02129, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Michael B Fessler
- From the Immunity, Inflammation and Disease Laboratory, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709,
| |
Collapse
|
92
|
Smirnova AV, Sukhorukov VN, Karagodin VP, Orekhov AN. [Epigenetic factors in atherogenesis: microRNA]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2016; 62:134-40. [PMID: 27143369 DOI: 10.18097/pbmc20166202134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
MicroRNAs (miRNAs) are small (~22 nucleotides in length) noncoding RNA sequences regulating gene expression at posttranscriptional level. MicroRNAs bind complementarily to certain mRNA and cause gene silencing. The involvement of miRNAs in the regulation of lipid metabolism, inflammatory response, cell cycle progression and proliferation, oxidative stress, platelet activation, endothelial and vascular smooth muscle cells (VSMC) function, angiogenesis and plaque formation and rapture indicates important roles in the initiation and progression of atherosclerosis. The key role of microRNAs in pathophysiology of cardiovascular diseases (CVDs), including atherosclerosis, was demonstrated in recent studies. Creating antisense oligonucleotides is a novel technique for selective changes in gene expression both in vitro and in vivo. In this review, we draw attention to the role of miRNAs in atherosclerosis progression, using miRNA as the potential biomarkers and targets in the CVDs, as well as possible application of antisense oligonucleotides.
Collapse
Affiliation(s)
- A V Smirnova
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - V N Sukhorukov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia
| | - V P Karagodin
- Plekhanov Russian University of Economics, Moscow, Russia
| | - A N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia; Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia
| |
Collapse
|
93
|
Casado-Díaz A, Anter J, Müller S, Winter P, Quesada-Gómez JM, Dorado G. Transcriptomic Analyses of Adipocyte Differentiation From Human Mesenchymal Stromal-Cells (MSC). J Cell Physiol 2016; 232:771-784. [PMID: 27349923 DOI: 10.1002/jcp.25472] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
Adipogenesis is a physiological process required for fat-tissue development, mainly involved in regulating the organism energetic-state. Abnormal distribution-changes and dysfunctions in such tissue are associated to different pathologies. Adipocytes are generated from progenitor cells, via a complex differentiating process not yet well understood. Therefore, we investigated differential mRNA and miRNA expression patterns of human mesenchymal stromal-cells (MSC) induced and not induced to differentiate into adipocytes by next (second)-generation sequencing. A total of 2,866 differentially expressed genes (101 encoding miRNA) were identified, with 705 (46 encoding miRNA) being upregulated in adipogenesis. They were related to different pathways, including PPARG, lipid, carbohydrate and energy metabolism, redox, membrane-organelle biosynthesis, and endocrine system. Downregulated genes were related to extracellular matrix and cell migration, proliferation, and differentiation. Analyses of mRNA-miRNA interaction showed that repressed miRNA-encoding genes can act downregulating PPARG-related genes; mostly the PPARG activator (PPARGC1A). Induced miRNA-encoding genes regulate downregulated genes related to TGFB1. These results shed new light to understand adipose-tissue differentiation and physiology, increasing our knowledge about pathologies like obesity, type-2 diabetes and osteoporosis. J. Cell. Physiol. 232: 771-784, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Jaouad Anter
- Dep. Genética, Universidad de Córdoba, Córdoba, Spain
| | | | | | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus de Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
94
|
Orenes-Piñero E, Marín F, Lip GYH. miRNA-197 and miRNA-223 and cardiovascular death in coronary artery disease patients. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:200. [PMID: 27294096 DOI: 10.21037/atm.2016.05.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Esteban Orenes-Piñero
- 1 Proteomic Unit, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 2 Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 3 University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Birmingham, UK
| | - Francisco Marín
- 1 Proteomic Unit, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 2 Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 3 University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Birmingham, UK
| | - Gregory Y H Lip
- 1 Proteomic Unit, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 2 Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca (IMIB-Arrixaca), Universidad de Murcia, Murcia, Spain ; 3 University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Birmingham, UK
| |
Collapse
|
95
|
Boosani CS, Dhar K, Agrawal DK. Down-regulation of hsa-miR-1264 contributes to DNMT1-mediated silencing of SOCS3. Mol Biol Rep 2016; 42:1365-76. [PMID: 26047583 DOI: 10.1007/s11033-015-3882-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Previously we found decreased expression of SOCS3 in neointimal hyperplastic region following balloon angioplasty in atherosclerotic micro swine. In our recent in vitro studies using human coronary artery smooth muscle cells (HCASMC), we observed the inhibition of SOCS3 expression in the presence of both TNF-α and IGF-1, correlating with the in vivo findings in microswine. We also reported that two independent mechanisms, JAK/STAT3/NFκB and promoter methylation of SOCS3 were responsible for TNF-α and IGF-1 induced SOCS3 inhibition. In this study, using miRNA array and gene expression approaches, we explored the molecular mechanisms involved in the above SOCS3 repression and identified several miRNAs that are associated with the regulation of SOCS3 expression. Our miRNA expression profiling revealed profound down-regulation of two specific miRNAs, hsa-miR-758 and hsa-miR-1264, whose expression levels were decreased by 8-10 folds in HCASMCs that were treated with both TNF-α and IGF-1. This was accompanied with a significant up-regulation of three specific miRNAs, hsa-miR-155, hsa-miR-146b-5p and hsa-miR-146a, which showed about 3-7 fold increases in their expression levels. Importantly, we also found that the miRNA hsa-miR-1264 targets DNA methyltransferase-1 (DNMT1) transcripts by binding to its 3'UTR region to affect its expression. Expression of hsa-miR-1264 in HCASMCs not only resulted in decreased DNMT1 mRNA transcripts but it also increased SOCS3 expression. The treatment with TNF-α and IGF-1 resulted in drastic decrease in hsa-miR-1264 levels with no change in the expression of DNMT1. Consequently, the DNMT1 activity caused hypermethylation in the CpG island of the SOCS3 promoter region and inhibited its expression. This could be a causative epigenetic mechanism associated with TNF-α and IGF-1 induced smooth muscle cell proliferation involved in the pathogenesis of coronary artery hyperplasia and restenosis.
Collapse
Affiliation(s)
- Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine Creighton University, 2500 California Plaza, Omaha, NE, 68178, USA
| | | | | |
Collapse
|
96
|
Can U, Buyukinan M, Yerlikaya FH. The investigation of circulating microRNAs associated with lipid metabolism in childhood obesity. Pediatr Obes 2016. [PMID: 26223376 DOI: 10.1111/ijpo.12050] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Childhood obesity is an increasing health challenge related to increased risk of chronic diseases. microRNAs (miRNAs) are noncoding short RNA molecules regulating multiple biological processes linked to obesity. OBJECTIVES We aimed at evaluating the association between circulating miRNA levels and lipid metabolism in obese and non-obese children and adolescents. METHODS By constituting study group, 45 obese children and adolescents were recruited. To perform comparisons with study group, 41 lean controls were matched for age and sex. Using real-time quantitative PCR analysis, circulating miRNAs were evaluated in both groups. RESULTS Circulating miR-335 (P < 0.001), miR-143 (P = 0.001) and miR-758 (P = 0.006) in obese children were significantly lower than those of controls. However, circulating miR-27 (P = 0.032), miR-378 (P < 0.001) and miR-370 (P = 0.045) in obese children were significantly higher, compared with those of controls. In addition, circulating miR-33 in obese children was higher than those of controls, but no significant difference was present (P = 0.687). CONCLUSION Our findings showed that a significant association is present between circulating miR-370, miR-33, miR-378, miR-27, miR-335, miR-143 and miR-758 values, and childhood obesity. Low levels of miR-335, miR-143 and miR-758, and high levels of miR-27, miR-378, miR-33 and miR-370 may have been responsible for elevated triglycerides and low-density lipoprotein (LDL-C) levels, and low level of high-density lipoprotein (HDL-C) in obese subjects. Therefore, miRNAs may be a good novel biomarker for childhood obesity.
Collapse
Affiliation(s)
- U Can
- Department of Biochemistry, Konya Education and Research Hospital, Konya, Turkey
| | - M Buyukinan
- Department of Pediatric Endocrinology, Konya Education and Research Hospital, Konya, Turkey
| | - F H Yerlikaya
- Department of Biochemistry, Meram Faculty of Medicine, University of Necmettin Erbakan, Konya, Turkey
| |
Collapse
|
97
|
Yoon H, Flores LF, Kim J. MicroRNAs in brain cholesterol metabolism and their implications for Alzheimer's disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2139-2147. [PMID: 27155217 DOI: 10.1016/j.bbalip.2016.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 01/01/2023]
Abstract
Cholesterol is important for various neuronal functions in the brain. Brain has elaborate regulatory mechanisms to control cholesterol metabolism that are distinct from the mechanisms in periphery. Interestingly, dysregulation of the cholesterol metabolism is strongly associated with a number of neurodegenerative diseases. MicroRNAs are short non-coding RNAs acting as post-transcriptional gene regulators. Recently, several microRNAs are demonstrated to be involved in regulating cholesterol metabolism in the brain. This article reviews the regulatory mechanisms of cellular cholesterol homeostasis in the brain. In addition, we discuss the role of microRNAs in brain cholesterol metabolism and their potential implications for the treatment of Alzheimer's disease. This article is part of a special issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Hyejin Yoon
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, United States; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Luis F Flores
- Biochemistry and Molecular Biology Graduate Program, Mayo Graduate School, Jacksonville, FL, United States
| | - Jungsu Kim
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, United States; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.
| |
Collapse
|
98
|
MicroRNA: a connecting road between apoptosis and cholesterol metabolism. Tumour Biol 2016; 37:8529-54. [PMID: 27105614 DOI: 10.1007/s13277-016-4988-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/10/2016] [Indexed: 12/15/2022] Open
Abstract
Resistance to apoptosis leads to tumorigenesis and failure of anti-cancer therapy. Recent studies also highlight abrogated lipid/cholesterol metabolism as one of the root causes of cancer that can lead to metastatic transformations. Cancer cells are dependent on tremendous supply of cellular cholesterol for the formation of new membranes and continuation of cell signaling. Cholesterol homeostasis network tightly regulates this metabolic need of cancer cells on cholesterol and other lipids. Genetic landscape is also shared between apoptosis and cholesterol metabolism. MicroRNAs (miRNAs) are the new fine tuners of signaling pathways and cellular processes and are known for their ability to post-transcriptionally repress gene expression in a targeted manner. This review summarizes the current knowledge about the cross talk between apoptosis and cholesterol metabolism via miRNAs. In addition, we also emphasize herein recent therapeutic modulations of specific miRNAs and their promising potential for the treatment of deadly diseases including cancer and cholesterol related pathologies. Understanding of the impact of miRNA-based regulation of apoptosis and metabolic processes is still at its dawn and needs further research for the development of future miRNA-based therapies. As both these physiological processes affect cellular homeostasis, we believe that this comprehensive summary of miRNAs modulating both apoptosis and cholesterol metabolism will open uncharted territory for scientific exploration and will provide the foundation for discovering novel drug targets for cancer and metabolic diseases.
Collapse
|
99
|
Canfrán-Duque A, Lin CS, Goedeke L, Suárez Y, Fernández-Hernando C. Micro-RNAs and High-Density Lipoprotein Metabolism. Arterioscler Thromb Vasc Biol 2016; 36:1076-84. [PMID: 27079881 DOI: 10.1161/atvbaha.116.307028] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
Improved prevention and treatment of cardiovascular diseases is one of the challenges in Western societies, where ischemic heart disease and stroke are the leading cause of death. Early epidemiological studies have shown an inverse correlation between circulating high-density lipoprotein-cholesterol (HDL-C) and cardiovascular diseases. The cardioprotective effect of HDL is because of its ability to remove cholesterol from plaques in the artery wall to the liver for excretion by a process known as reverse cholesterol transport. Numerous studies have reported the role that micro-RNAs (miRNA) play in the regulation of the different steps in reverse cholesterol transport, including HDL biogenesis, cholesterol efflux, and cholesterol uptake in the liver and bile acid synthesis and secretion. Because of their ability to control different aspects of HDL metabolism and function, miRNAs have emerged as potential therapeutic targets to combat cardiovascular diseases. In this review, we summarize the recent advances in the miRNA-mediated control of HDL metabolism. We also discuss how HDL particles serve as carriers of miRNAs and the potential use of HDL-containing miRNAs as cardiovascular diseases biomarkers.
Collapse
Affiliation(s)
- Alberto Canfrán-Duque
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Chin-Sheng Lin
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Leigh Goedeke
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Yajaira Suárez
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.)
| | - Carlos Fernández-Hernando
- From the Vascular Biology and Therapeutics Program (A.C.-D., L.G., Y.S., C.F.-H.) and Integrative Cell Signaling and Neurobiology of Metabolism Program, Section of Comparative Medicine and Department of Pathology (A.C.-D., L.G., Y.S., C.F.-H.), Yale University School of Medicine, New Haven, CT; and Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (C.-S.L.).
| |
Collapse
|
100
|
BI DAPENG, YIN CHENGHUA, ZHANG XIAOYUE, YANG NANA, XU JIAYOU. miR-183 functions as an oncogene by targeting ABCA1 in colon cancer. Oncol Rep 2016; 35:2873-9. [DOI: 10.3892/or.2016.4631] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 10/13/2015] [Indexed: 11/05/2022] Open
|