1
|
Ricciotti E, Haines PG, Chai W, FitzGerald GA. Prostanoids in Cardiac and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:558-583. [PMID: 38269585 PMCID: PMC10922399 DOI: 10.1161/atvbaha.123.320045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024]
Abstract
Prostanoids are biologically active lipids generated from arachidonic acid by the action of the COX (cyclooxygenase) isozymes. NSAIDs, which reduce the biosynthesis of prostanoids by inhibiting COX activity, are effective anti-inflammatory, antipyretic, and analgesic drugs. However, their use is limited by cardiovascular adverse effects, including myocardial infarction, stroke, hypertension, and heart failure. While it is well established that NSAIDs increase the risk of atherothrombotic events and hypertension by suppressing vasoprotective prostanoids, less is known about the link between NSAIDs and heart failure risk. Current evidence indicates that NSAIDs may increase the risk for heart failure by promoting adverse myocardial and vascular remodeling. Indeed, prostanoids play an important role in modulating structural and functional changes occurring in the myocardium and in the vasculature in response to physiological and pathological stimuli. This review will summarize current knowledge of the role of the different prostanoids in myocardial and vascular remodeling and explore how maladaptive remodeling can be counteracted by targeting specific prostanoids.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Philip G Haines
- Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence (P.G.H.)
| | - William Chai
- Health and Human Biology, Division of Biology and Medicine, Brown University, Providence, RI (W.C.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Institute for Translational Medicine and Therapeutics (E.R., G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Medicine (G.A.F.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| |
Collapse
|
2
|
Tubular IKKβ Deletion Alleviates Acute Ischemic Kidney Injury and Facilitates Tissue Regeneration. Int J Mol Sci 2022; 23:ijms231710199. [PMID: 36077596 PMCID: PMC9456401 DOI: 10.3390/ijms231710199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/30/2022] Open
Abstract
Acute kidney injury (AKI) is a common renal injury leading to relevant morbidity and mortality worldwide. Most of the clinical cases of AKI are caused by ischemia reperfusion (I/R) injury with renal ischemia injury followed by reperfusion injury and activation of the innate immune response converging to NF-ĸB pathway induction. Despite the clear role of NF-ĸB in inflammation, it has recently been acknowledged that NF-ĸB may impact other cell functions. To identify NF-ĸB function with respect to metabolism, vascular function and oxidative stress after I/R injury and to decipher in detail the underlying mechanism, we generated a transgenic mouse model with targeted deletion of IKKβ along the tubule and applied I/R injury followed by its analysis after 2 and 14 days after I/R injury. Tubular IKKβ deletion ameliorated renal function and reduced tissue damage. RNAseq data together with immunohistochemical, biochemical and morphometric analysis demonstrated an ameliorated vascular organization and mRNA expression profile for increased angiogenesis in mice with tubular IKKβ deletion at 2 days after I/R injury. RNAseq and protein analysis indicate an ameliorated metabolism, oxidative species handling and timely-adapted cell proliferation and apoptosis as well as reduced fibrosis in mice with tubular IKKβ deletion at 14 days after I/R injury. In conclusion, mice with tubular IKKβ deletion upon I/R injury display improved renal function and reduced tissue damage and fibrosis in association with improved vascularization, metabolism, reactive species disposal and fine-tuned cell proliferation.
Collapse
|
3
|
Vu TA, Lema I, Hani I, Cheval L, Atger-Lallier L, Souvannarath V, Perrot J, Souvanheuane M, Marie Y, Fabrega S, Blanchard A, Bouligand J, Kamenickỷ P, Crambert G, Martinerie L, Lombès M, Viengchareun S. miR-324-5p and miR-30c-2-3p Alter Renal Mineralocorticoid Receptor Signaling under Hypertonicity. Cells 2022; 11:cells11091377. [PMID: 35563683 PMCID: PMC9104010 DOI: 10.3390/cells11091377] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
The Mineralocorticoid Receptor (MR) mediates the sodium-retaining action of aldosterone in the distal nephron, but mechanisms regulating MR expression are still poorly understood. We previously showed that RNA Binding Proteins (RBPs) regulate MR expression at the post-transcriptional level in response to variations of extracellular tonicity. Herein, we highlight a novel regulatory mechanism involving the recruitment of microRNAs (miRNAs) under hypertonicity. RT-qPCR validated miRNAs candidates identified by high throughput screening approaches and transfection of a luciferase reporter construct together with miRNAs Mimics or Inhibitors demonstrated their functional interaction with target transcripts. Overexpression strategies using Mimics or lentivirus revealed the impact on MR expression and signaling in renal KC3AC1 cells. miR-324-5p and miR-30c-2-3p expression are increased under hypertonicity in KC3AC1 cells. These miRNAs directly affect Nr3c2 (MR) transcript stability, act with Tis11b to destabilize MR transcript but also repress Elavl1 (HuR) transcript, which enhances MR expression and signaling. Overexpression of miR-324-5p and miR-30c-2-3p alter MR expression and signaling in KC3AC1 cells with blunted responses in terms of aldosterone-regulated genes expression. We also confirm that their expression is increased by hypertonicity in vivo in the kidneys of mice treated with furosemide. These findings may have major implications for the pathogenesis of renal dysfunctions, sodium retention, and mineralocorticoid resistance.
Collapse
Affiliation(s)
- Thi An Vu
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Ingrid Lema
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Imene Hani
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université Paris Cité, 75006 Paris, France; (L.C.); (G.C.)
| | - Laura Atger-Lallier
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Vilayvane Souvannarath
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Julie Perrot
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Mélanie Souvanheuane
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Yannick Marie
- Plateforme de Genotypage Séquençage (iGenSeq), Institut du Cerveau et de la Moelle Epinière, Hôpital Sapêtrière, 75013 Paris, France;
| | - Sylvie Fabrega
- Plateforme Vecteurs Viraux et Transfert de Gènes, Structure Federative de Recherche Necker, UMS 24, UMS 3633, Faculté de Santé, Université Paris Cité, 75015 Paris, France;
| | - Anne Blanchard
- Inserm, Centre d’Investigations Cliniques 9201, 75015 Paris, France;
| | - Jérôme Bouligand
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, 94275 Le Kremlin-Bicêtre, France
| | - Peter Kamenickỷ
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
- Assistance Publique-Hopitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, 94275 Le Kremlin-Bicêtre, France
| | - Gilles Crambert
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université Paris Cité, 75006 Paris, France; (L.C.); (G.C.)
| | - Laetitia Martinerie
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
- Assistance-Publique Hôpitaux de Paris, Hôpital Robert Debré, Service d’Endocrinologie Pédiatrique, Université Paris Cité, 75019 Paris, France
| | - Marc Lombès
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
| | - Say Viengchareun
- Physiologie et Physiopathologie Endocriniennes, Université Paris-Saclay, Inserm, 94276 Le Kremlin-Bicêtre, France; (T.A.V.); (I.L.); (I.H.); (L.A.-L.); (V.S.); (J.P.); (M.S.); (J.B.); (P.K.); (L.M.); (M.L.)
- Correspondence:
| |
Collapse
|
4
|
Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Single nucleotide polymorphism (SNP) variants are abundant, persistent and widely distributed across the genome and are frequently linked to the development of genetic diseases. Identifying SNPs that underpin complex diseases can aid scientists in the discovery of disease-related genes by allowing for early detection, effective medication and eventually disease prevention.
Main body
Various SNP or polymorphism-based studies were used to categorize different SNPs potentially related to three quantitative traits: body mass index (BMI), intelligence quotient (IQ) and blood pressure, and then uncovered common SNPs for these three traits. We employed SNPedia, RefSNP Report, GWAS Catalog, Gene Cards (Data Bases), PubMed and Google Scholar search engines to find relevant material on SNPs associated with three quantitative traits. As a result, we detected three common SNPs for all three quantitative traits in global populations: SNP rs6265 of the BDNF gene on chromosome 11p14.1, SNP rs131070325 of the SL39A8 gene on chromosome 4p24 and SNP rs4680 of the COMT gene on chromosome 22q11.21.
Conclusion
In our review, we focused on the prevalent SNPs and gene expression activities that influence these three quantitative traits. These SNPs have been used to detect and map complex, common illnesses in communities for homogeneity testing and pharmacogenetic studies. High blood pressure, diabetes and heart disease, as well as BMI, schizophrenia and IQ, can all be predicted using common SNPs. Finally, the results of our work can be used to find common SNPs and genes that regulate these three quantitative features across the genome.
Collapse
|
5
|
Török B, Fazekas CL, Szabó A, Zelena D. Epigenetic Modulation of Vasopressin Expression in Health and Disease. Int J Mol Sci 2021; 22:ijms22179415. [PMID: 34502322 PMCID: PMC8430944 DOI: 10.3390/ijms22179415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Vasopressin is a ubiquitous molecule playing an important role in a wide range of physiological processes thereby implicated in the pathomechanism of many disorders. Its effect is well characterized through V2 receptors, which regulates the water resorption in kidney, while its vasoconstrictory effect through V1a receptor also received a lot of attention in the maintenance of blood pressure during shock. However, the most striking is its central effect both through the V1b receptors in stress-axis regulation as well as through V1a receptors regulating many aspects of our behavior (e.g., social behavior, learning and memory). Vasopressin has been implicated in the development of depression, due to its connection with chronic stress, as well as schizophrenia because of its involvement in social interactions and memory processes. Epigenetic changes may also play a role in the development of these disorders. The possible mechanism includes DNA methylation, histone modification and/or micro RNAs, and these possible regulations will be in the focus of our present review.
Collapse
Affiliation(s)
- Bibiána Török
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Adrienn Szabó
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Dóra Zelena
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
6
|
Wang C, Hu F. Long noncoding RNA SOX2OT silencing alleviates cerebral ischemia-reperfusion injury via miR-135a-5p-mediated NR3C2 inhibition. Brain Res Bull 2021; 173:193-202. [PMID: 34022287 DOI: 10.1016/j.brainresbull.2021.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 01/04/2023]
Abstract
OBJECTIVE This study is aimed to investigate the role of the long noncoding RNA SOX2 overlapping transcript (SOX2OT) in cerebral ischemia-reperfusion injury (CIRI) and the underlying regulatory mechanisms. METHODS The oxygen-glucose deprivation/reoxygenation (OGD/R)-treated PC12 cells and middle cerebral artery occlusion/reperfusion (MCAO/R)-treated rats were established to simulate CIRI condition in vitro and in vivo. Quantitative real-time polymerase chain reaction was performed to detect the expression of SOX2OT, microRNA-135a-5p (miR-135a-5p), and nuclear receptor subfamily 3 group C member 2 (NR3C2). The cell viability and apoptosis were analyzed by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays. The levels of lactate dehydrogenase (LDH), malondialdehyde (MDA), superoxide dismutase (SOD), and reactive oxygen species (ROS) or interleukin (IL)-1β and IL-6 were used to evaluate the oxidative stress or inflammation. Dual-luciferase reporter assay was conducted to validate the interactions among SOX2OT, miR-135a-5p, and NR3C2. Additionally, neurological deficit scores (NDS), infarct volume, and brain edema were used to assess brain impairments in vivo. RESULTS The expression of SOX2OT and NR3C2 was increased, while miR-135a-5p was decreased in OGD/R-treated PC12 cells. SOX2OT silencing repressed the levels of LDH, MDA, ROS, IL-1β, IL-6, reduced the numbers of TUNEL positive cells, and elevated viability and SOD level in OGD/R-treated PC12 cells. Besides, SOX2OT targeted miR-135a-5p, and miR-135a-5p targeted NR3C2. Both miR-135a-5p downregulation and NR3C2 upregulation reversed the suppressive effects of SOX2OT knockdown on oxidative stress, apoptosis, and inflammation of OGD/R-treated PC12 cells. Furthermore, injection of sh-SOX2OT reduced the NDS, cerebral infarct, and cerebral edema in MCAO/R-treated rats. CONCLUSIONS Silencing of SOX2OT attenuated CIRI via regulation of the miR-135a-5p/NR3C2 axis, which may provide a novel therapeutic target for CIRI.
Collapse
Affiliation(s)
- Chao Wang
- Department of Neurology, Shanxi Provincial People's Hospital, No. 29, Shuangta Road, Taiyuan City, Shanxi Province, 030012, China
| | - Fengyun Hu
- Department of Neurology, Shanxi Provincial People's Hospital, No. 29, Shuangta Road, Taiyuan City, Shanxi Province, 030012, China.
| |
Collapse
|
7
|
Butterworth MB. Non-coding RNAs and the mineralocorticoid receptor in the kidney. Mol Cell Endocrinol 2021; 521:111115. [PMID: 33301840 PMCID: PMC7796954 DOI: 10.1016/j.mce.2020.111115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
The final steps in the Renin-Angiotensin-Aldosterone signaling System (RAAS) involve binding of the corticosteroid hormone, aldosterone to its mineralocorticoid receptor (MR). The bound MR interacts with response elements to induce or repress the transcription of aldosterone-regulated genes. Along with the classic genomic targets of aldosterone that alter mRNA and protein expression, aldosterone also regulates the expression of non-coding RNAs (ncRNAs). Short ncRNAs termed microRNAs (miRs) have been shown to play a role in transducing aldosterone's actions via MR signaling. The role of miRs in homeostatic regulation of aldosterone signaling, and the potential for aldosterone-regulated miRs to act as feedback regulators of MR have been recently reported. In this review, the role of miRs in RAAS signaling and feedback regulation of MR in kidney epithelial cells will be discussed.
Collapse
Affiliation(s)
- Michael B Butterworth
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Wang Y, Li Y, Jia D, Zheng J, Wang G. Correlation between single nucleotide polymorphisms in CXCR4 microRNA binding site and the susceptibility to knee osteoarthritis in Han Chinese population. J Clin Lab Anal 2020; 35:e23600. [PMID: 32978834 PMCID: PMC7891528 DOI: 10.1002/jcla.23600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
Background This study aimed to investigate the relationship between single nucleotide polymorphisms (SNPs) at the microRNA target sequence in CXCR4 and the susceptibility to knee osteoarthritis (KOA). Methods A total of 305 patients with KOA and 305 healthy controls were recruited into this study. The genotypes of CXCR4 rs1804029 and rs17848060 loci were analyzed. Results The susceptibility to KOA of CXCR4 rs1804029 G allele carriers was 1.33 times (95% CI: 1.09‐1.54, P = .006) that of T allele carriers. The KOA susceptibility in individuals carrying T allele at CXCR4 rs17848060 locus was 1.38 times that of individuals carrying A allele (95% CI: 1.17‐1.57, P < .001). The G allele at CXCR4 rs1804029 locus was the target of hsa‐miR‐146a‐3p, while the A allele at CXCR4 rs17848060 locus could be targeted by hsa‐miR‐20a‐3p. The plasma level of hsa‐miR‐146a‐3p was lower in rs1804029 G allele carriers than T allele carriers (P < .001), whereas plasma level of hsa‐miR‐20a‐3p was higher in rs17848060 T allele carriers than A allele carriers (P < .001). Conclusion The SNPs at rs1804029 and rs17848060 loci in CXCR4 were significantly associated with the susceptibility to KOA in Han Chinese population.
Collapse
Affiliation(s)
- Yang Wang
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Di Jia
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiali Zheng
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
9
|
Ozbaki-Yagan N, Liu X, Bodnar A, Ho J, Butterworth M. Aldosterone-induced microRNAs act as feedback regulators of mineralocorticoid receptor signaling in kidney epithelia. FASEB J 2020; 34:11714-11728. [PMID: 32652691 PMCID: PMC7725848 DOI: 10.1096/fj.201902254rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
The final steps in the Renin-Angiotensin-Aldosterone signaling System (RAAS) involve binding of the corticosteroid hormone, aldosterone to its mineralocorticoid receptor (MR). The bound MR interacts with response elements to induce or repress the transcription of aldosterone-regulated genes. A well characterized aldosterone-induced gene is the serum and glucocorticoid-induced kinase (SGK1), which acts downstream to increase sodium transport in distal kidney nephron epithelial cells. The role of microRNAs (miRs) induced by extended aldosterone stimulation in regulating MR and SGK1 has not been reported. In these studies, miRs predicted to bind to the 3'-UTR of mouse MR were profiled by qRT-PCR after aldosterone stimulation. The miR-466a/b/c/e family was upregulated in mouse kidney cortical collecting duct epithelial cells. A luciferase reporter assay confirmed miR-466 binding to both MR and SGK1 3'-UTRs. Inhibition of miR-466 increased MR and SGK1 mRNA and protein levels. Inhibiting miR-466b and preventing its upregulation after aldosterone stimulation increased amiloride-sensitive sodium transport and sensitivity to aldosterone stimulation. In vivo upregulation of miR-466 was confirmed in distal nephrons of mice on low Na+ diets. Repression of MR and SGK1 by aldosterone-induced miRs may represent a negative feedback loop that contributes to a form of aldosterone escape in vivo.
Collapse
Affiliation(s)
- N. Ozbaki-Yagan
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - X. Liu
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - A.J. Bodnar
- Division of Nephrology in the Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - J. Ho
- Division of Nephrology in the Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - M.B. Butterworth
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
10
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
11
|
Tömböl Z, Turai PI, Decmann Á, Igaz P. MicroRNAs and Adrenocortical Tumors: Where do we Stand on Primary Aldosteronism? Horm Metab Res 2020; 52:394-403. [PMID: 32168526 DOI: 10.1055/a-1116-2366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
MicroRNAs, the endogenous mediators of RNA interference, interact with the renin-angiotensin-aldosterone system, regulate aldosterone secretion and aldosterone effects. Some novel data show that the expression of some microRNAs is altered in primary aldosteronism, and some of these appear to have pathogenic relevance, as well. Differences in the circulating microRNA expression profiles between the two major forms of primary aldosteronism, unilateral aldosterone-producing adenoma and bilateral adrenal hyperplasia have also been shown. Here, we present a brief synopsis of these findings focusing on the potential relevance of microRNA in primary aldosteronism.
Collapse
Affiliation(s)
- Zsófia Tömböl
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter István Turai
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ábel Decmann
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Igaz
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| |
Collapse
|
12
|
Abstract
BACKGROUND The influence of genetic polymorphisms on the development of gestational hypertension (GH) is unclear. The aim of this study was to examine whether single-nucleotide polymorphisms (SNPs) of the nuclear receptor subfamily 3, group C, member 2 (NR3C2) genes, rs5522, rs2070951, rs5534, s2248038, and s9992256 are associated with GH in Han Chinese women. METHOD Sanger sequencing was used to analyze the genotypes of rs5522, rs2070951, rs5534, rs2248038, and rs9992256 loci of the NR3C2 gene in 450 patients with GH and 450 healthy controls. RESULTS The rs5522 dominant model (odds ratio [OR] = 1.30, 95% confidence interval [CI]: 1.13-1.47, P < .001) and the recessive model (OR = 1.64, 95% CI: 1.33-1.86, P < .001) had higher GH risk. The rs2070951 dominant model (OR = 1.18, 95% CI: 1.03-1.35, P = .02) had higher risk of GH, and the recessive model (OR = 1.09, 95% CI: 0.84-1.34, P = .55) was not significant for GH risk. The rs5534 dominant model (OR = 1.25, 95% CI: 1.09-1.43, P = .001) had a higher GH risk. The rs2248038 and rs9992256 sites were not significantly related to GH risk. Gene-gene interactions at the rs5522, rs2070951, and rs5534 loci affected GH risk (OR = 1.34, 95% CI: 1.12-1.64, P < .001). CONCLUSION The SNPs of the NR3C2 gene rs5522, rs2070951, and rs5534 are associated with GH in Han Chinese women.
Collapse
Affiliation(s)
- Zhenghui Cui
- Obstetrical Department, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianyun Xu
- Obstetrical Department, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenying Jiang
- Obstetrical Department, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Nielsen BS, Holmstrøm K. Combined MicroRNA In Situ Hybridization and Immunohistochemical Detection of Protein Markers. Methods Mol Biol 2019; 1953:271-286. [PMID: 30912028 DOI: 10.1007/978-1-4939-9145-7_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs are short (18-23 nucleotides) noncoding RNAs involved in posttranscriptional regulation of gene expression through their specific binding to the 3'UTR of mRNAs. MicroRNAs can be detected in tissues using specific locked nucleic acid (LNA)-enhanced probes. The characterization of microRNA expression in tissues by in situ detection is often crucial following a microRNA biomarker discovery phase in order to validate the candidate microRNA biomarker and allow better interpretation of its molecular functions and derived cellular interactions. The in situ hybridization data provides information about contextual distribution and cellular origin of the microRNA. By combining microRNA in situ hybridization with immunohistochemical staining of protein markers, it is possible to precisely characterize the microRNA-expressing cells and to identify the potential microRNA targets. This combined technology can also help to monitor changes in the level of potential microRNA targets in a therapeutic setting. In this chapter, we present a fluorescence-based detection method that allows the combination of microRNA in situ hybridization with immunohistochemical staining of one and, in this updated version of the paper, two protein markers detected with primary antibodies raised in the same host species.
Collapse
|
14
|
Correlation between miRNA target site polymorphisms in the 3' UTR of AVPR1A and the risk of hypertension in the Chinese Han population. Biosci Rep 2019; 39:BSR20182232. [PMID: 31053625 PMCID: PMC6522731 DOI: 10.1042/bsr20182232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/14/2019] [Accepted: 04/29/2019] [Indexed: 12/26/2022] Open
Abstract
We aimed to study the relationship between rs11174811 and rs3803107 single nucleotide polymorphisms (SNPs) in miRNA target sites of the 3' UTR in the arginine vasopressin receptor 1a gene (AVPR1A) and the risk of hypertension in the Chinese Han population. The genotypes at rs11174811 and rs3803107 were analyzed by direct sequencing in 425 Chinese Han patients with hypertension and 425 healthy subjects. AVPR1A expression was investigated by transfecting miR-526b, miR-375, and miR-186 mimics into human umbilical vein endothelial cells (HUVECs) containing AVPR1A rs11174811 CC, CA/AA and AVPR1A rs3803107 GG, GA/AA genotypes. The A alleles of rs11174811 (adjusted OR = 1.424, 95% CI: 1.231-1.599, P<0.001) and rs3803107 (adjusted OR = 1.222, 95% CI: 1.092-1.355; P=0.001) were high risk factors for hypertension. Plasma levels of miR-526b, miR-375, and miR-186 were higher in the study group than in the control group (P<0.001). The expression levels of AVPR1A mRNA in AVPR1A rs11174811 and rs3803107 mutant HUVECs were higher than those in wild-type cells (t = 8.811, 4.068 and P=0.001, 0.015, respectively). The single nucleotide polymorphisms rs11174811 and rs3803107 in the AVPR1A gene are associated with an increased risk of hypertension in the Chinese Han population. This may be related to the effect of these variants on the regulation of AVPR1A expression by miRNAs.
Collapse
|
15
|
Abstract
The mineralocorticoid hormone aldosterone is released by the adrenal glands in a homeostatic mechanism to regulate blood volume. Several cues elicit aldosterone release, and the long-term action of the hormone is to restore blood pressure and/or increase the retrieval of sodium from filtered plasma in the kidney. While the signaling cascade that results in aldosterone release is well studied, the impact of this hormone on tissues and cells in various organ systems is pleotropic. Emerging evidence indicates aldosterone may alter non-coding RNAs (ncRNAs) to integrate the hormonal response, and these ncRNAs may contribute to the heterogeneity of signaling outcomes in aldosterone target tissues. The best studied of the ncRNAs in aldosterone action are the small ncRNAs, microRNAs. MicroRNA expression is regulated by aldosterone stimulation, and microRNAs are able to modulate protein expression at all steps in the renin-angiotensin-aldosterone-signaling system. The discovery and synthesis of microRNAs will be briefly covered followed by a discussion of the reciprocal role of aldosterone/microRNA regulation, including misregulation of microRNA signaling in aldosterone-linked disease states.
Collapse
|
16
|
TBXA2R rs4523 G allele is associated with decreased susceptibility to Kawasaki disease. Cytokine 2018; 111:216-221. [PMID: 30179800 DOI: 10.1016/j.cyto.2018.08.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/03/2018] [Accepted: 08/25/2018] [Indexed: 02/06/2023]
Abstract
Kawasaki disease is a multi-system vasculitis and a primary cause of acquired heart disease among children. Genetic factors may increase susceptibility to Kawasaki disease. TBXA2R is a G-protein-coupled receptor that participates in tissue inflammation and is associated with susceptibility to several diseases, but its relevance in Kawasaki disease is unclear. We genotyped TBXA2R (rs1131882 and rs4523) in 694 Kawasaki disease cases and 657 healthy controls. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to evaluate the intensity of the associations. We found a significantly decreased risk of Kawasaki disease associated with TBXA2R rs4523 G variant genotypes (AG vs AA: adjusted OR = 0.788, 95%CI = 0.626-0.993; GG vs AA: adjusted OR = 0.459, 95%CI = 0.258-0.815; AG/GG vs AA: adjusted OR = 0.744, 95%CI = 0.595-0.929; GG vs AG/AA: adjusted OR = 0.497, 95% CI = 0.281-0.879). In the combined analysis of the two single-nucleotide polymorphisms (SNPs), we found that individuals with two unfavorable genotypes exhibited decreased risk for Kawasaki disease (adjusted OR = 0.754, 95%CI = 0.577-0.985) compared with those who did not have or one unfavorable genotypes. This cumulative effect on protection is effect-genotype dose-dependent (ptrend = 0.022). Moreover, the combined analysis indicated that the two unfavorable genotypes were associated with a decreased risk of Kawasaki disease in children 12-60 months of age, females and the subgroup with non-coronary artery lesion (NCAL) formation compared with those who did not have or one unfavorable genotypes. In conclusion, the TBXA2R rs4523 G allele may contribute to protection against Kawasaki disease and decreased risk of coronary artery aneurysm complications in a southern Chinese population.
Collapse
|
17
|
Leimena C, Qiu H. Non-Coding RNA in the Pathogenesis, Progression and Treatment of Hypertension. Int J Mol Sci 2018; 19:E927. [PMID: 29561765 PMCID: PMC5979335 DOI: 10.3390/ijms19040927] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/11/2022] Open
Abstract
Hypertension is a complex, multifactorial disease that involves the coexistence of multiple risk factors, environmental factors and physiological systems. The complexities extend to the treatment and management of hypertension, which are still the pursuit of many researchers. In the last two decades, various genes have emerged as possible biomarkers and have become the target for investigations of specialized drug design based on its risk factors and the primary cause. Owing to the growing technology of microarrays and next-generation sequencing, the non-protein-coding RNAs (ncRNAs) have increasingly gained attention, and their status of redundancy has flipped to importance in normal cellular processes, as well as in disease progression. The ncRNA molecules make up a significant portion of the human genome, and their role in diseases continues to be uncovered. Specifically, the cellular role of these ncRNAs has played a part in the pathogenesis of hypertension and its progression to heart failure. This review explores the function of the ncRNAs, their types and biology, the current update of their association with hypertension pathology and the potential new therapeutic regime for hypertension.
Collapse
Affiliation(s)
- Christiana Leimena
- Department of Basic Sciences, Physiological Division, School of Medicine, Loma Linda University, Loma Linda, CA 92324, USA.
| | - Hongyu Qiu
- Department of Basic Sciences, Physiological Division, School of Medicine, Loma Linda University, Loma Linda, CA 92324, USA.
| |
Collapse
|
18
|
De Santi C, Gadi S, Swiatecka-Urban A, Greene CM. Identification of a novel functional miR-143-5p recognition element in the Cystic Fibrosis Transmembrane Conductance Regulator 3'UTR. AIMS GENETICS 2018; 5:53-62. [PMID: 31435512 PMCID: PMC6690249 DOI: 10.3934/genet.2018.1.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/08/2018] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in regulation of gene expression. They bind in a sequence-specific manner to miRNA recognition elements (MREs) located in the 3' untranslated region (UTR) of target mRNAs and prevent mRNA translation. MiRNA expression is dysregulated in cystic fibrosis (CF), affecting several biological processes including ion conductance in the epithelial cells of the lung. We previously reported that miR-143 is up-regulated in CF bronchial brushings compared to non-CF. Here we identified two predicted binding sites for miR-143-5p (starting at residues 558 and 644) on the CFTR mRNA, and aimed to assess whether CFTR is a true molecular target of miR-143-5p. Expression of miR-143-5p was found to be up-regulated in a panel of CF vs non-CF cell lines (1.7-fold, P = 0.0165), and its levels were increased in vitro after 20 hours treatment with bronchoalveolar lavage fluid from CF patients compared to vehicle-treated cells (3.3-fold, P = 0.0319). Luciferase assays were performed to elucidate direct miRNA::target interactions and showed that miR-143-5p significantly decreased the reporter activity when carrying the wild-type full length sequence of CFTR 3'UTR (minus 15%, P = 0.005). This repression was rescued by the disruption of the first, but not the second, predicted MRE, suggesting that the residue starting at 558 was the actual active binding site. In conclusion, we here showed that miR-143-5p modestly but significantly inhibits CFTR, improving the knowledge on functional MREs within the CFTR 3'UTR. This could lead to the development of novel therapeutic strategies where miRNA-mediated CFTR repression is blocked thereby possibly increasing the efficacy of the currently available CFTR modulators.
Collapse
Affiliation(s)
- Chiara De Santi
- Lung Biology Group, Department of Clinical Microbiology, RCSI Education & Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Sucharitha Gadi
- Lung Biology Group, Department of Clinical Microbiology, RCSI Education & Research Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Agnieszka Swiatecka-Urban
- Children's Hospital of Pittsburgh of UPMC, Department of Cell Biology, University of Pittsburgh School of Medicine Pittsburgh, PA, USA
| | - Catherine M. Greene
- Lung Biology Group, Department of Clinical Microbiology, RCSI Education & Research Centre, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
19
|
Bhaumik P, Ghosh P, Biswas A, Ghosh S, Pal S, Sarkar B, Kumar Dey S. Rare Intronic Variations inTP73Gene Found in Patients with Alzheimer’sDisease. INT J HUM GENET 2018. [DOI: 10.1080/09723757.2017.1421438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Pranami Bhaumik
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| | - Priyanka Ghosh
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| | - Atanu Biswas
- Department of Neurology, Bangur Institute of Neurosciences, 52/1A, S.N. Pandit Street, Kolkata 700 025, West Bengal, India
| | - Sujay Ghosh
- Department of Zoology, University of Calcutta, (Ballygunge Science College Campus), 35 Ballygunge Circular Road., Kolkata 700 019, West Bengal, India
| | - Sandip Pal
- Department of Neurology, Burdwan Medical College, Burdwan 713 104, West Bengal, India
| | - Biswanath Sarkar
- DNA Laboratory, Anthropological Survey of India, 27 Jawaharlal Nehru Road Kolkata 700 016, West Bengal, India
| | - Subrata Kumar Dey
- Department of Biotechnology, School of Biotechnology and Biological Sciences, Maulana Abul Kalam Azad University of Technology, West Bengal (Formerly known as West Bengal University of Technology) BF – 142, Salt Lake City, Sector I. Kolkata 700 064, West Bengal, India
| |
Collapse
|
20
|
Cai MY, Cheng J, Zhou MY, Liang LL, Lian SM, Xie XS, Xu S, Liu X, Xiong XD. The association between pre-miR-27a rs895819 polymorphism and myocardial infarction risk in a Chinese Han population. Lipids Health Dis 2018; 17:7. [PMID: 29304813 PMCID: PMC5756394 DOI: 10.1186/s12944-017-0652-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/27/2017] [Indexed: 12/21/2022] Open
Abstract
Background Accumulating evidences have shown that miRNAs are directly or indirectly involved in a variety of biological processes, and closely associated with diverse human diseases, including cardiovascular diseases. SNPs locating within pri/pre-miRNA can affect miRNA processing and binding ability of target genes. MiR-27a, miR-26a-1 miR-100, miR-126 and miR-218 were reported to be associated with pathogenesis of myocardial infarction (MI). Here we aimed to evaluate the potential association of five polymorphisms in these pri/pre-miRNAs with individual susceptibility to MI in a Chinese Han population. Methods Genotyping was performed in 287 MI cases and 646 control subjects using polymerase chain reaction-ligase detection reaction (PCR-LDR) method. The association of these SNPs with MI risk was performed with SPSS software. Results In a logistic regression analysis, we found that AG heterozygote (OR = 0.40, 95% CI = 0.21-0.76, Pa = 0.005) or AA homozygote (OR = 0.40, 95% CI = 0.22-0.75, Pa = 0.004) of pre-miR-27a rs895819 had a reduced susceptibility to MI in comparison with GG homozygote. Similarly, a reduced risk of MI was detected when the AG and AA genotypes were combined (OR = 0.40, 95% CI = 0.22-0.74, Pa = 0.003). However, no significant association between pri-miR-26a-1 pri-miR-100, pri-miR-126 and pri-miR-218 polymorphisms and MI risk was observed under the allelic and established genetic models. Further stratified analysis of pre-miR-27a rs895819 revealed a more significant association of AG + AA genotypes with MI risk among younger, male and smoking subjects. Interestingly, AG and AA genotypes of the rs895819 polymorphism conferred about 0.17 mmol/L and 0.18 mmol/L increase in HDL-C levels compared to GG genotype. Conclusions Our findings suggest that the pre-miR-27a rs895819 polymorphism is associated with MI susceptibility in the Chinese Han population, which probably due to influence the HDL-C levels. Electronic supplementary material The online version of this article (10.1186/s12944-017-0652-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meng-Yun Cai
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Jie Cheng
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China.,Department of Clinical Laboratory, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Meng-Yuan Zhou
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Li-Li Liang
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Si-Min Lian
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Xiao-Shan Xie
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Shun Xu
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China
| | - Xinguang Liu
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China.,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China.,Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xing-Dong Xiong
- Institute of Aging Research, Guangdong Medical University, Xin Cheng Avenue 1#, Songshan Lake, Dongguan, 523808, People's Republic of China. .,Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, People's Republic of China. .,Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Zhanjiang, People's Republic of China.
| |
Collapse
|
21
|
Koshy L, Vijayalekshmi SV, Harikrishnan S, Raman KV, Jissa VT, Jayakumaran Nair A, Gangaprasad A, Nair GM, Sudhakaran PR. Lack of association of mirSNP rs11174811 in AVPR1A gene with arterial blood pressure and hypertension in South Indian population. Clin Exp Hypertens 2017; 40:534-538. [PMID: 29182374 DOI: 10.1080/10641963.2017.1403624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Epigenetic regulation of arterial blood pressure mediated through mirSNPs in renin-angiotensin aldosterone system (RAAS) genes is a less explored hypothesis. Recently, the mirSNP rs11174811 in the 3'UTR of the AVPR1A gene was associated with higher arterial blood pressure in a large study population from the Study of Myocardial Infarctions Leiden (SMILE). The aim of the present study was to replicate the association of mirSNP rs11174811 with blood pressure outcomes and hypertension in a south Indian population. Four hundred and fifteen hypertensive cases and 416 normotensive controls were genotyped using a 5' nuclease allelic discrimination assay. Logistic regression was used to test the association of mirSNP rs11174811 with the hypertension phenotype. Censored normal regression was used to test the association of the polymorphism with continuous blood pressure outcomes such as systolic and diastolic blood pressure. The mirSNP rs11174811 did not show any significant association with hypertension. The adjusted odds ratio was 1.02, with 95% CI of 0.72 to 1.45 (p = 0.909). The mean systolic and diastolic blood pressure values were not significantly different across the three genotypic groups, between hypertensives and normotensives, or when stratified by gender. Despite having a similar minor allele frequency (MAF) of 14.5% compared with the SMILE cohort, our results did not support an association of the mirSNP rs11174811 with the hypertension phenotype or with continuous blood pressure outcomes in the south Indian population.
Collapse
Affiliation(s)
- Linda Koshy
- a Inter-University Centre for Genomics and Gene Technology, Department of Biotechnology , University of Kerala , Trivandrum , India
| | - S V Vijayalekshmi
- a Inter-University Centre for Genomics and Gene Technology, Department of Biotechnology , University of Kerala , Trivandrum , India
| | - S Harikrishnan
- b Department of Cardiology , Sree Chitra Tirunal Institute for Medical Sciences and Technology , Trivandrum , India
| | - Kutty V Raman
- c Achutha Menon Centre for Health Science Studies , Sree Chitra Tirunal Institute for Medical Sciences and Technology , Trivandrum , India
| | - V T Jissa
- c Achutha Menon Centre for Health Science Studies , Sree Chitra Tirunal Institute for Medical Sciences and Technology , Trivandrum , India
| | - A Jayakumaran Nair
- a Inter-University Centre for Genomics and Gene Technology, Department of Biotechnology , University of Kerala , Trivandrum , India
| | - A Gangaprasad
- d Department of Botany , University of Kerala , Trivandrum , India
| | - G M Nair
- a Inter-University Centre for Genomics and Gene Technology, Department of Biotechnology , University of Kerala , Trivandrum , India
| | - P R Sudhakaran
- a Inter-University Centre for Genomics and Gene Technology, Department of Biotechnology , University of Kerala , Trivandrum , India
| |
Collapse
|
22
|
Decoding resistant hypertension signalling pathways. Clin Sci (Lond) 2017; 131:2813-2834. [PMID: 29184046 DOI: 10.1042/cs20171398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
Abstract
Resistant hypertension (RH) is a clinical condition in which the hypertensive patient has become resistant to drug therapy and is often associated with increased cardiovascular morbidity and mortality. Several signalling pathways have been studied and related to the development and progression of RH: modulation of sympathetic activity by leptin and aldosterone, primary aldosteronism, arterial stiffness, endothelial dysfunction and variations in the renin-angiotensin-aldosterone system (RAAS). miRNAs comprise a family of small non-coding RNAs that participate in the regulation of gene expression at post-transcriptional level. miRNAs are involved in the development of both cardiovascular damage and hypertension. Little is known of the molecular mechanisms that lead to development and progression of this condition. This review aims to cover the potential roles of miRNAs in the mechanisms associated with the development and consequences of RH, and explore the current state of the art of diagnostic and therapeutic tools based on miRNA approaches.
Collapse
|
23
|
HPA Axis Genes, and Their Interaction with Childhood Maltreatment, are Related to Cortisol Levels and Stress-Related Phenotypes. Neuropsychopharmacology 2017; 42:2446-2455. [PMID: 28589964 PMCID: PMC5645736 DOI: 10.1038/npp.2017.118] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/12/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
Stress responses are controlled by the hypothalamus pituitary adrenal (HPA)-axis and maladaptive stress responses are associated with the onset and maintenance of stress-related disorders such as major depressive disorder (MDD). Genes that play a role in the HPA-axis regulation may likely contribute to the relation between relevant neurobiological substrates and stress-related disorders. Therefore, we performed gene-wide analyses for 30 a priori literature-based genes involved in HPA-axis regulation in 2014 subjects (34% male; mean age: 42.5) to study the relations with lifetime MDD diagnosis, cortisol awakening response, and dexamethasone suppression test (DST) levels (subsample N=1472) and hippocampal and amygdala volume (3T MR images; subsample N=225). Additionally, gene by childhood maltreatment (CM) interactions were investigated. Gene-wide significant results were found for dexamethasone suppression (CYP11A1, CYP17A1, POU1F1, AKR1D1), hippocampal volume (CYP17A1, CYP11A1, HSD3B2, PROP1, AVPRA1, SRD5A1), amygdala volume (POMC, CRH, HSD3B2), and lifetime MDD diagnosis (FKBP5 and CRH), all permutation p-values<0.05. Interactions with CM were found for several genes; the strongest interactions were found for NR3C2, where the minor allele of SNP rs17581262 was related to smaller hippocampal volume, smaller amygdala volume, higher DST levels, and higher odds of MDD diagnosis only in participants with CM. As hypothesized, several HPA-axis genes are associated with stress-related endophenotypes including cortisol response and reduced brain volumes. Furthermore, we found a pleiotropic interaction between CM and the mineralocorticoid receptor gene, suggesting that this gene plays an important moderating role in stress and stress-related disorders.
Collapse
|
24
|
HDL-cholesterol concentration in pregnant Chinese Han women of late second trimester associated with genetic variants in CETP, ABCA1, APOC3, and GALNT2. Oncotarget 2017; 8:56737-56746. [PMID: 28915626 PMCID: PMC5593597 DOI: 10.18632/oncotarget.18128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/25/2017] [Indexed: 11/25/2022] Open
Abstract
Objective To investigate whether HDL-C level in pregnant Chinese Han women of late second trimester correlated with loci in high-density lipoprotein-cholesterol (HDL-C)-related genes found in genome-wide association studies (GWAS). Methods Seven single-nucleotide polymorphisms (rs3764261 in CETP, rs1532085 in LIPC, rs7241918 in LIPG, rs1883025 in ABCA1, rs4225 in APOC3, rs1059611 in LPL, and rs16851339 in GALNT2) were genotyped using the Sequenom MassArray system for 1,884 pregnant women. Results The following polymorphisms were statistically associated with HDL-C level after adjusting for age, gestational week, pre-pregnancy BMI and state of GDM or HOMAIR: (i) rs3764261 (b = -0.055 mmol/L, 95% CI -0.101 to -0.008, p = 0.021), (ii) rs1883025 (b = -0.054 mmol/L, 95% CI -0.097 to -0.012, p = 0.013), (iii) rs4225 (b = -0.071 mmol/L, 95% CI -0.116 to -0.027, p = 1.79E-3) and (iv) rs16851339 (b = -0.064 mmol/L, 95% CI -0.120 to -0.008, p = 0.025). The more risk alleles the pregnant women have, the lower the plasma HDL-C levels of the subjects are. Conclusions Several risk alleles found to be related to HDL-C in GWAS are also associated with HDL-C levels in pregnant Chinese Han women and these risk loci contribute additively to low HDL-C levels.
Collapse
|
25
|
Randesi M, Levran O, Correa da Rosa J, Hankins J, Rule J, Kreek MJ, Lee WM. Association of Variants of Arginine Vasopressin and Arginine Vasopressin Receptor 1A With Severe Acetaminophen Liver Injury. Cell Mol Gastroenterol Hepatol 2017; 3:500-505. [PMID: 28462386 PMCID: PMC5404026 DOI: 10.1016/j.jcmgh.2017.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/16/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Acetaminophen-related acute liver injury and liver failure (ALF) result from ingestion of supratherapeutic quantities of this analgesic, frequently in association with other forms of substance abuse including alcohol, opioids, and cocaine. Thus, overdosing represents a unique high-risk behavior associated with other forms of drug use disorder. METHODS We examined a series of 21 single nucleotide polymorphisms (SNPs) in 9 genes related to impulsivity and/or stress responsivity that may modify response to stress. Study subjects were 229 white patients admitted to tertiary care liver centers for ALF that was determined to be due to acetaminophen toxicity after careful review of historical and biochemical data. Identification of relevant SNPs used Sanger sequencing, TaqMan, or custom microarray. Association tests were carried out to compare genotype frequencies between patients and healthy white controls. RESULTS The mean age was 37 years, and 75.6% were female, with similar numbers classified as intentional overdose or unintentional (without suicidal intent, occurring for a period of several days, usually due to pain). There was concomitant alcohol abuse in 30%, opioid use in 33.6%, and use of other drugs of abuse in 30.6%. The genotype frequencies of 2 SNPs were found to be significantly different between the cases and controls, specifically SNP rs2282018 in the arginine vasopressin gene (AVP, odds ratio 1.64) and SNP rs11174811 in the AVP receptor 1A gene (AVPR1A, odds ratio 1.89), both of which have been previously linked to a drug use disorder diagnosis. CONCLUSIONS Patients who develop acetaminophen-related ALF have increased frequency of gene variants that may cause altered stress responsivity, which has been shown to be associated with other unrelated substance use disorders.
Collapse
Affiliation(s)
- Matthew Randesi
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Orna Levran
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Joel Correa da Rosa
- Center for Clinical and Translational Science, Rockefeller University, New York, New York
| | - Julia Hankins
- Center for the Genetics of Host Defense, Department of Internal Medicine, UT Southwestern Medical Center at Dallas, Dallas, Texas
| | - Jody Rule
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center at Dallas, Dallas, Texas
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - William M. Lee
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center at Dallas, Dallas, Texas
- Correspondence Address correspondence to: William M. Lee, MD, FACP, FAASLD, Division of Digestive and Liver Diseases, UT Southwestern Medical Center at Dallas, 5959 Harry Hines Boulevard, Suite 420, Dallas, Texas 75390-8887. fax: (214) 645–6114.Division of Digestive and Liver DiseasesUT Southwestern Medical Center at Dallas5959 Harry Hines Boulevard, Suite 420DallasTexas 75390-8887
| | | |
Collapse
|
26
|
Cornejo-García JA, Perkins JR, Jurado-Escobar R, García-Martín E, Agúndez JA, Viguera E, Pérez-Sánchez N, Blanca-López N. Pharmacogenomics of Prostaglandin and Leukotriene Receptors. Front Pharmacol 2016; 7:316. [PMID: 27708579 PMCID: PMC5030812 DOI: 10.3389/fphar.2016.00316] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/02/2016] [Indexed: 01/15/2023] Open
Abstract
Individual genetic background together with environmental effects are thought to be behind many human complex diseases. A number of genetic variants, mainly single nucleotide polymorphisms (SNPs), have been shown to be associated with various pathological and inflammatory conditions, representing potential therapeutic targets. Prostaglandins (PTGs) and leukotrienes (LTs) are eicosanoids derived from arachidonic acid and related polyunsaturated fatty acids that participate in both normal homeostasis and inflammatory conditions. These bioactive lipid mediators are synthesized through two major multistep enzymatic pathways: PTGs by cyclooxygenase and LTs by 5-lipoxygenase. The main physiological effects of PTGs include vasodilation and vascular leakage (PTGE2); mast cell maturation, eosinophil recruitment, and allergic responses (PTGD2); vascular and respiratory smooth muscle contraction (PTGF2), and inhibition of platelet aggregation (PTGI2). LTB4 is mainly involved in neutrophil recruitment, vascular leakage, and epithelial barrier function, whereas cysteinyl LTs (CysLTs) (LTC4, LTD4, and LTE4) induce bronchoconstriction and neutrophil extravasation, and also participate in vascular leakage. PTGs and LTs exert their biological functions by binding to cognate receptors, which belong to the seven transmembrane, G protein-coupled receptor superfamily. SNPs in genes encoding these receptors may influence their functionality and have a role in disease susceptibility and drug treatment response. In this review we summarize SNPs in PTGs and LTs receptors and their relevance in human diseases. We also provide information on gene expression. Finally, we speculate on future directions for this topic.
Collapse
Affiliation(s)
- José A Cornejo-García
- Research Laboratory, International Business Information Management Association (IBIMA)-Regional University Hospital of Malaga, University of Málaga (UMA)Malaga, Spain; Allergy Unit, International Business Information Management Association (IBIMA)-Regional University Hospital of Malaga, University of Málaga (UMA)Malaga, Spain
| | - James R Perkins
- Research Laboratory, International Business Information Management Association (IBIMA)-Regional University Hospital of Malaga, University of Málaga (UMA) Malaga, Spain
| | - Raquel Jurado-Escobar
- Research Laboratory, International Business Information Management Association (IBIMA)-Regional University Hospital of Malaga, University of Málaga (UMA) Malaga, Spain
| | | | - José A Agúndez
- Department of Pharmacology, University of Extremadura Caceres, Spain
| | - Enrique Viguera
- Genetics Unit, Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga Malaga, Spain
| | - Natalia Pérez-Sánchez
- Allergy Unit, International Business Information Management Association (IBIMA)-Regional University Hospital of Malaga, University of Málaga (UMA) Malaga, Spain
| | | |
Collapse
|
27
|
Jaisser F, Farman N. Emerging Roles of the Mineralocorticoid Receptor in Pathology: Toward New Paradigms in Clinical Pharmacology. Pharmacol Rev 2016; 68:49-75. [PMID: 26668301 DOI: 10.1124/pr.115.011106] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mineralocorticoid receptor (MR) and its ligand aldosterone are the principal modulators of hormone-regulated renal sodium reabsorption. In addition to the kidney, there are several other cells and organs expressing MR, in which its activation mediates pathologic changes, indicating potential therapeutic applications of pharmacological MR antagonism. Steroidal MR antagonists have been used for decades to fight hypertension and more recently heart failure. New therapeutic indications are now arising, and nonsteroidal MR antagonists are currently under development. This review is focused on nonclassic MR targets in cardiac, vascular, renal, metabolic, ocular, and cutaneous diseases. The MR, associated with other risk factors, is involved in organ fibrosis, inflammation, oxidative stress, and aging; for example, in the kidney and heart MR mediates hormonal tissue-specific ion channel regulation. Genetic and epigenetic modifications of MR expression/activity that have been documented in hypertension may also present significant risk factors in other diseases and be susceptible to MR antagonism. Excess mineralocorticoid signaling, mediated by aldosterone or glucocorticoids binding, now appears deleterious in the progression of pathologies that may lead to end-stage organ failure and could therefore benefit from the repositioning of pharmacological MR antagonists.
Collapse
Affiliation(s)
- F Jaisser
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| | - N Farman
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| |
Collapse
|
28
|
de Kloet ER, Otte C, Kumsta R, Kok L, Hillegers MHJ, Hasselmann H, Kliegel D, Joëls M. Stress and Depression: a Crucial Role of the Mineralocorticoid Receptor. J Neuroendocrinol 2016; 28. [PMID: 26970338 DOI: 10.1111/jne.12379] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/30/2016] [Accepted: 02/23/2016] [Indexed: 12/27/2022]
Abstract
Cortisol and corticosterone act on the appraisal process, which comprises the selection of an appropriate coping style and the encoding of the experience for storage in the memory. This action exerted by the stress hormones is mediated by mineralocorticoid receptors (MRs), which are expressed abundantly in the limbic circuitry, particularly in the hippocampus. Limbic MR is down-regulated by chronic stress and during depression but induced by antidepressants. Increased MR activity inhibits hypothalamic-pituitary-adrenal axis activity, promotes slow wave sleep, reduces anxiety and switches circuit connectivity to support coping. Cortisol and emotion-cognition are affected by MR gene haplotypes based on rs5522 and rs2070951. Haplotype 1 (GA) moderates the effects of (early) life stressors, reproductive cycle and oral contraceptives. MR haplotype 2 (CA) is a gain of function variant that protects females against depression by association with an optimistic, resilient phenotype. Activation of MR therefore may offer a target for alleviating depression and cognitive dysfunction. Accordingly, the MR agonist fludrocortisone was found to enhance the efficacy of antidepressants and to improve memory and executive functions in young depressed patients. In conclusion, CORT coordinates via MR the networks underlying how an individual copes with stress, and this action is complemented by the widely distributed lower affinity glucocorticoid receptor (GR) involved in the subsequent management of stress adaptation. In this MR:GR regulation, the MR is an important target for promoting resilience.
Collapse
MESH Headings
- Adaptation, Psychological
- Animals
- Antidepressive Agents/therapeutic use
- Brain/metabolism
- Brain/physiopathology
- Corticosterone/metabolism
- Corticosterone/physiology
- Depression/metabolism
- Depression/physiopathology
- Fludrocortisone/therapeutic use
- Humans
- Polymorphism, Single Nucleotide
- Receptors, Glucocorticoid/metabolism
- Receptors, Glucocorticoid/physiology
- Receptors, Mineralocorticoid/agonists
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Receptors, Mineralocorticoid/physiology
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- E R de Kloet
- Division of Internal Medicine, Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - C Otte
- Klinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - R Kumsta
- Genetic Psychology, Fakultät für Psychologie, Ruhr-Universität Bochum, Bochum, Germany
| | - L Kok
- Department of Anesthesiology and Intensive Care, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M H J Hillegers
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Hasselmann
- Klinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - D Kliegel
- Department of Biological und Clinical Psychology, University of Trier, Trier, Germany
| | - M Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
29
|
Bretschneider M, Busch B, Mueller D, Nolze A, Schreier B, Gekle M, Grossmann C. Activated mineralocorticoid receptor regulates micro-RNA-29b in vascular smooth muscle cells. FASEB J 2016; 30:1610-22. [PMID: 26728178 DOI: 10.1096/fj.15-271254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/11/2015] [Indexed: 12/17/2022]
Abstract
Inappropriately activated mineralocorticoid receptor (MR) is a risk factor for vascular remodeling with unclear molecular mechanism. Recent findings suggest that post-transcriptional regulation by micro-RNAs (miRs) may be involved. Our aim was to search for MR-dependent miRs in vascular smooth muscle cells (VSMCs) and to explore the underlying molecular mechanism and the pathologic relevance. We detected that aldosteroneviathe MR reduces miR-29bin vivoin murine aorta and in human primary and cultured VSMCs (ED50= 0.07 nM) but not in endothelial cells [quantitative PCR (qPCR), luciferase assays]. This effect was mediated by an increased decay of miR-29b in the cytoplasm with unchanged miR-29 family member or primary-miR levels. Decreased miR-29b led to an increase in extracellular matrix measured by ELISA and qPCR and enhanced VSMC migration in single cell-tracking experiments. Additionally, cell proliferation and the apoptosis/necrosis ratio (caspase/lactate dehydrogenase assay) was modulated by miR-29b. Enhanced VSMC migration by aldosterone required miR-29b regulation. Control experiments were performed with scrambled RNA and empty plasmids, by comparing aldosterone-stimulated with vehicle-incubated cells. Overall, our findings provide novel insights into the molecular mechanism of aldosterone-mediated vascular pathogenesis by identifying miR-29b as a pathophysiologic relevant target of activated MR in VSMCs and by highlighting the importance of miR processing for miR regulation.-Bretschneider, M., Busch, B., Mueller, D., Nolze, A., Schreier, B., Gekle, M., Grossmann, C. Activated mineralocorticoid receptor regulates micro-RNA-29b in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Maria Bretschneider
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Bianca Busch
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Daniel Mueller
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Nolze
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Barbara Schreier
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Michael Gekle
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Grossmann
- *Julius Bernstein Institute of Physiology and Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
30
|
Santulli G. MicroRNAs and Endothelial (Dys) Function. J Cell Physiol 2015; 231:1638-44. [PMID: 26627535 DOI: 10.1002/jcp.25276] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that microRNAs (miRs)-non-coding RNAs that can regulate gene expression via translational repression and/or post-transcriptional degradation-are becoming one of the most fascinating areas of physiology, given their fundamental roles in countless pathophysiological processes. The relative roles of different miRs in vascular biology as direct or indirect post-transcriptional regulators of fundamental genes implied in vascular remodeling designate miRs as potential biomarkers and/or promising drug targets. The mechanistic importance of miRs in modulating endothelial cell (EC) function in physiology and in disease is addressed here. Drawbacks of currently available therapeutic options are also discussed, pointing at the challenges and clinical opportunities provided by miR-based treatments. J. Cell. Physiol. 231: 1638-1644, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gaetano Santulli
- Columbia University Medical Center, New York Presbyterian Hospital-Manhattan, New York, New York
| |
Collapse
|
31
|
Bailey EL, McBride MW, Beattie W, McClure JD, Graham D, Dominiczak AF, Sudlow CLM, Smith C, Wardlaw JM. Differential gene expression in multiple neurological, inflammatory and connective tissue pathways in a spontaneous model of human small vessel stroke. Neuropathol Appl Neurobiol 2015; 40:855-72. [PMID: 24417612 PMCID: PMC4260148 DOI: 10.1111/nan.12116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/08/2014] [Indexed: 02/03/2023]
Abstract
AIMS Cerebral small vessel disease (SVD) causes a fifth of all strokes plus diffuse brain damage leading to cognitive decline, physical disabilities and dementia. The aetiology and pathogenesis of SVD are unknown, but largely attributed to hypertension or microatheroma. METHODS We used the spontaneously hypertensive stroke-prone rat (SHRSP), the closest spontaneous experimental model of human SVD, and age-matched control rats kept under identical, non-salt-loaded conditions, to perform a blinded analysis of mRNA microarray, qRT-PCR and pathway analysis in two brain regions (frontal and mid-coronal) commonly affected by SVD in the SHRSP at age five, 16 and 21 weeks. RESULTS We found gene expression abnormalities, with fold changes ranging from 2.5 to 59 for the 10 most differentially expressed genes, related to endothelial tight junctions (reduced), nitric oxide bioavailability (reduced), myelination (impaired), glial and microglial activity (increased), matrix proteins (impaired), vascular reactivity (impaired) and albumin (reduced), consistent with protein expression defects in the same rats. All were present at age 5 weeks thus predating blood pressure elevation. 'Neurological' and 'inflammatory' pathways were more affected than 'vascular' functional pathways. CONCLUSIONS This set of defects, although individually modest, when acting in combination could explain the SHRSP's susceptibility to microvascular and brain injury, compared with control rats. Similar combined, individually modest, but multiple neurovascular unit defects, could explain susceptibility to spontaneous human SVD.
Collapse
Affiliation(s)
- Emma L Bailey
- Centre for Clinical Brain Sciences, University of Edinburgh, Western General Hospital, Edinburgh; Department of Bioengineering, Imperial College London, London
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wang X, Li W, Ma L, Gao J, Liu J, Ping F, Nie M. Association study of the miRNA-binding site polymorphisms of CDKN2A/B genes with gestational diabetes mellitus susceptibility. Acta Diabetol 2015; 52:951-8. [PMID: 25990668 DOI: 10.1007/s00592-015-0768-2] [Citation(s) in RCA: 19] [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: 11/03/2014] [Accepted: 04/26/2015] [Indexed: 10/23/2022]
Abstract
AIMS Gestational diabetes mellitus (GDM) is a complex disease induced by a combination of genetic factors and environmental exposures. Growing evidence suggests that common single nucleotide polymorphisms within miRNA-binding sites (miR-binding SNPs) contribute to the development of various diseases. However, the roles of miR-binding SNPs in GDM have not been fully elucidated. The CDKN2A/B genes have been identified as two of the strongest genetic determinants for diabetes risk. The aim of the study was to first investigate the associations between miR-binding SNPs of CDKN2A/B, GDM susceptibility, and quantitative metabolism traits. METHODS Three miR-binding SNPs of CDKN2A/B gene (rs1063192, rs3217992, and rs3088440) were selected and genotyped using TaqMan allelic discrimination assays in 839 cases of GDM and 900 controls. RESULTS The CC genotype of CDKN2B rs1063192, which is located in the hsa-miR-323b-5p binding site, was significantly associated with GDM [OR 1.418 (1.143, 1.908); p = 0.003]. The C allele of rs1063192 occurred with significantly higher frequency in GDM [OR 1.22 (1.03, 1.44); p = 0.021]. The rs1063192 genotype CC exhibited increased glucose levels at 1 h and 3 h, as well as higher insulin levels at 3 h during an OGTT compared with the control TT genotype (p < 0.05). We also found that the rs1063192 CC genotype was associated with lower total cholesterol and LDL cholesterol levels (p < 0.05). CONCLUSIONS The CC genotype of CDKN2B rs1063192 in the hsa-miR-323b-5p binding site increased the risk of GDM in pregnant Chinese Han women. Importantly, our study provides evidence that miR-binding SNPs are a novel source of GDM susceptibility loci.
Collapse
Affiliation(s)
- Xiaojing Wang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Shuai Fu Yuan No. 1, Dongcheng District, Beijing, 100730, China.
| | - Wei Li
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Shuai Fu Yuan No. 1, Dongcheng District, Beijing, 100730, China.
| | - Liangkun Ma
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jinsong Gao
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Juntao Liu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Fan Ping
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Shuai Fu Yuan No. 1, Dongcheng District, Beijing, 100730, China.
| | - Min Nie
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Shuai Fu Yuan No. 1, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
33
|
Seeger T, Boon RA. MicroRNAs in cardiovascular ageing. J Physiol 2015; 594:2085-94. [PMID: 26040259 DOI: 10.1113/jp270557] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRs) have emerged as potent regulators of pathways in physiological and disease contexts. This review focuses on the role of miRs in ageing of the cardiovascular system. Several miRs have been described to be regulated during ageing and some of these miRs are involved in the regulation of ageing-related processes. We discuss the roles of miR-34, miR-217 and miR-29, which are induced during ageing in the vasculature. The roles of miR-34, miR-29 (age-induced) and miR-18/19, which are decreased during ageing in the heart, are discussed as well. Furthermore, numerous miRs that play a role in diseases associated with ageing, like diabetes, atherosclerosis, hypertension, cardiac hypertrophy and atrial fibrillation, are also briefly discussed. miRs also serve as circulating biomarkers for cardiovascular ageing or ageing-associated diseases. Finally, pharmacological modulation of ageing-related miRs might become a promising strategy to combat cardiovascular ageing in a clinical setting.
Collapse
Affiliation(s)
- Timon Seeger
- Department of Medicine (Division of Cardiology), Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany
| |
Collapse
|
34
|
Shi L, Liao J, Liu B, Zeng F, Zhang L. Mechanisms and therapeutic potential of microRNAs in hypertension. Drug Discov Today 2015; 20:1188-204. [PMID: 26004493 DOI: 10.1016/j.drudis.2015.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 01/08/2023]
Abstract
Hypertension is the major risk factor for the development of stroke, coronary artery disease, heart failure and renal disease. The underlying cellular and molecular mechanisms of hypertension are complex and remain largely elusive. MicroRNAs (miRNAs) are short, noncoding RNA fragments of 22-26 nucleotides and regulate protein expression post-transcriptionally by targeting the 3'-untranslated region of mRNA. A growing body of recent research indicates that miRNAs are important in the pathogenesis of arterial hypertension. Herein, we summarize the current knowledge regarding the mechanisms of miRNAs in cardiovascular remodeling, focusing specifically on hypertension. We also review recent progress of the miRNA-based therapeutics including pharmacological and nonpharmacological therapies (such as exercise training) and their potential applications in the management of hypertension.
Collapse
Affiliation(s)
- Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China.
| | - Jingwen Liao
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Bailin Liu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Fanxing Zeng
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
35
|
Are microRNAs the Molecular Link Between Metabolic Syndrome and Alzheimer's Disease? Mol Neurobiol 2015; 53:2320-38. [PMID: 25976367 DOI: 10.1007/s12035-015-9201-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/29/2015] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in people over 65 years of age. At present, treatment options for AD address only its symptoms, and there are no available treatments for the prevention or delay of the disease process. Several preclinical and epidemiological studies have linked metabolic risk factors such as hypertension, obesity, dyslipidemia, and diabetes to the pathogenesis of AD. However, the molecular mechanisms that underlie this relationship are not fully understood. Considering that less than 1% of cases of AD are attributable to genetic factors, the identification of new molecular targets linking metabolic risk factors to neuropathological processes is necessary for improving the diagnosis and treatment of AD. The dysregulation of microRNAs (miRNAs), small non-coding RNAs that regulate several biological processes, has been implicated in the development of different pathologies. In this review, we summarize some of the relevant evidence that points to the role of miRNAs in metabolic syndrome (MetS) and AD and propose that miRNAs may be a molecular link in the complex relationship between both diseases.
Collapse
|
36
|
Reuter M, Cooper AJ, Smillie LD, Markett S, Montag C. A new measure for the revised reinforcement sensitivity theory: psychometric criteria and genetic validation. Front Syst Neurosci 2015; 9:38. [PMID: 25852497 PMCID: PMC4360558 DOI: 10.3389/fnsys.2015.00038] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 02/23/2015] [Indexed: 11/13/2022] Open
Abstract
Jeffrey Gray's Reinforcement Sensitivity Theory (RST) represents one of the most influential biologically-based personality theories describing individual differences in approach and avoidance tendencies. The most prominent self-report inventory to measure individual differences in approach and avoidance behavior to date is the BIS/BAS scale by Carver and White (1994). As Gray and McNaughton (2000) revised the RST after its initial formulation in the 1970/80s, and given the Carver and White measure is based on the initial conceptualization of RST, there is a growing need for self-report inventories measuring individual differences in the revised behavioral inhibition system (BIS), behavioral activation system (BAS) and the fight, flight, freezing system (FFFS). Therefore, in this paper we present a new questionnaire measuring individual differences in the revised constructs of the BIS, BAS and FFFS in N = 1814 participants (German sample). An English translated version of the new measure is also presented and tested in N = 299 English language participants. A large number of German participants (N = 1090) also filled in the BIS/BAS scales by Carver and White (1994) and the correlations between these measures are presented. Finally, this same subgroup of participants provided buccal swaps for the investigation of the arginine vasopressin receptor 1a (AVPR1a) gene. Here, a functional genetic polymorphism (rs11174811) on the AVPR1a gene was shown to be associated with individual differences in both the revised BIS and classic BIS dimensions.
Collapse
Affiliation(s)
- Martin Reuter
- Department of Psychology, University of BonnBonn, Germany
- Laboratory of Neurogenetics, University of BonnBonn, Germany
- Center of Economics and Neuroscience, University of BonnBonn, Germany
| | - Andrew J. Cooper
- Department of Psychology, Goldsmiths, University of LondonLondon, UK
| | - Luke D. Smillie
- Melbounre School of Psychological Sciences, The University of MelbourneMelbourne, VIC, Australia
| | | | | |
Collapse
|
37
|
Butterworth MB. MicroRNAs and the regulation of aldosterone signaling in the kidney. Am J Physiol Cell Physiol 2015; 308:C521-7. [PMID: 25673770 DOI: 10.1152/ajpcell.00026.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 01/22/2023]
Abstract
The role of small noncoding RNAs, termed microRNAs (miRs), in development and disease has been recognized for many years. The number of miRs and regulated targets that reinforce a role for miRs in human disease and disease progression is ever-increasing. However, less is known about the involvement of miRs in steady-state, nondisease homeostatic pathways. In the kidney, much of the regulated ion transport is under the control of hormonal signaling. Evidence is emerging that miRs are involved in the hormonal regulation of kidney function and, particularly, in ion transport. In this short review, the production and intra- and extracellular signaling of miRs and the involvement of miRs in kidney disease are discussed. The discussion also focuses on the role of these small biological molecules in the homeostatic control of ion transport in the kidney. MiR regulation of and by corticosteroid hormones, in particular the mineralocorticoid hormone aldosterone, is considered. While information about the role of aldosterone-regulated miRs in the kidney is limited, an increase in the research in this area will undoubtedly highlight the involvement of miRs as central mediators of hormonal signaling in normal physiology.
Collapse
Affiliation(s)
- Michael B Butterworth
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Xu R, Bi C, Song J, Wang L, Ge C, Liu X, Zhang M. Upregulation of miR-142-5p in atherosclerotic plaques and regulation of oxidized low-density lipoprotein-induced apoptosis in macrophages. Mol Med Rep 2015; 11:3229-34. [PMID: 25586666 PMCID: PMC4368070 DOI: 10.3892/mmr.2015.3191] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 09/18/2014] [Indexed: 01/25/2023] Open
Abstract
MicroRNA (miR)-142-5p is a member of the miR-142 family, which have been shown to be associated with tumors, stem cells and disorders of the immune system. However, the role of miR-142-5p in atherosclerosis has yet to be investigated. In the present study, an atherosclerotic apolipoprotein E-deficient (apoE−/−) mouse model was constructed and fed a high-fat diet. The expression levels of miR-142-5p in the murine atherosclerotic plaques were detected by gene microarray analysis. In addition, an in vitro assay was used to determine the expression levels of miR-142-5p in human endothelial cells, smooth muscle cells and macrophages, which were treated with oxidized low-density lipoprotein (ox-LDL). Furthermore, a miR-142-5p inhibitor and mimic was transfected into cultured human macrophages, in order to observe the effects on transforming growth factor-β2 (TGF-β2) expression. The effects of co-transfection of the miR-142-5p inhibitor or mimic with TGF-β2, in human macrophages, on the rate of apoptosis was analyzed. The expression levels of miR-142-5p were 6.84-fold higher in mice with stable atherosclerotic plaques, and 2.69-fold higher in mice with vulnerable atherosclerotic plaques, as compared with the controls. Furthermore, the expression levels of miR-142-5p were upregulated in the cultured human macrophages. The percentage of apoptotic cells was lowest in the macrophages transfected with both TGF-β2 and miR-142-5p inhibitors and treated with ox-LDL. The expression levels of miR-142-5p were upregulated in the atherosclerotic plaques of the apoE−/− mice. The findings of the present study have shown that the upregulation of miR-142-5p expression may regulate apoptosis in human macrophages by targeting TGF-β2. This effect may have an important role in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Ruijin Xu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chenglong Bi
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jiantao Song
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lin Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Cheng Ge
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinxin Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mei Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministries of Education and Public Health, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
39
|
Wronska A, Kurkowska-Jastrzebska I, Santulli G. Application of microRNAs in diagnosis and treatment of cardiovascular disease. Acta Physiol (Oxf) 2015; 213:60-83. [PMID: 25362848 DOI: 10.1111/apha.12416] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/08/2014] [Accepted: 10/24/2014] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Innovative, more stringent diagnostic and prognostic biomarkers and effective treatment options are needed to lessen its burden. In recent years, microRNAs have emerged as master regulators of gene expression - they bind to complementary sequences within the mRNAs of their target genes and inhibit their expression by either mRNA degradation or translational repression. microRNAs have been implicated in all major cellular processes, including cell cycle, differentiation and metabolism. Their unique mode of action, fine-tuning gene expression rather than turning genes on/off, and their ability to simultaneously regulate multiple elements of relevant pathways makes them enticing potential biomarkers and therapeutic targets. Indeed, cardiovascular patients have specific patterns of circulating microRNA levels, often early in the disease process. This article provides a systematic overview of the role of microRNAs in the pathophysiology, diagnosis and treatment of CVD.
Collapse
Affiliation(s)
- A. Wronska
- Helen and Clyde Wu Center for Molecular Cardiology; Department of Physiology and Cellular Biophysics; College of Physicians and Surgeons of Columbia University; New York NY USA
| | - I. Kurkowska-Jastrzebska
- Department of Experimental and Clinical Pharmacology; Medical University of Warsaw; Warsaw Poland
- 2nd Department of Neurology; National Institute of Psychiatry and Neurology; Warsaw Poland
| | - G. Santulli
- Helen and Clyde Wu Center for Molecular Cardiology; Department of Physiology and Cellular Biophysics; College of Physicians and Surgeons of Columbia University; New York NY USA
| |
Collapse
|
40
|
Murakami K. Non-coding RNAs and hypertension-unveiling unexpected mechanisms of hypertension by the dark matter of the genome. Curr Hypertens Rev 2015; 11:80-90. [PMID: 25828869 PMCID: PMC5384352 DOI: 10.2174/1573402111666150401105317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/05/2015] [Accepted: 03/30/2015] [Indexed: 12/23/2022]
Abstract
Hypertension is a major risk factor of cardiovascular diseases and a most important health problem in developed countries. Investigations on pathophysiology of hypertension have been based on gene products from coding region that occupies only about 1% of total genome region. On the other hand, non-coding region that occupies almost 99% of human genome has been regarded as "junk" for a long time and went unnoticed until these days. But recently, it turned out that noncoding region is extensively transcribed to non-coding RNAs and has various functions. This review highlights recent updates on the significance of non-coding RNAs such as micro RNAs and long non-coding RNAs (lncRNAs) on the pathogenesis of hypertension, also providing an introduction to basic biology of noncoding RNAs. For example, microRNAs are associated with hypertension via neuro-fumoral factor, sympathetic nerve activity, ion transporters in kidneys, endothelial function, vascular smooth muscle phenotype transformation, or communication between cells. Although reports of lncRNAs on pathogenesis of hypertension are scarce at the moment, new lncRNAs in relation to hypertension are being discovered at a rapid pace owing to novel techniques such as microarray or next-generation sequencing. In the clinical settings, clinical use of non-coding RNAs in identifying cardiovascular risks or developing novel tools for treating hypertension such as molecular decoy or mimicks is promising, although improvement in chemical modification or drug delivery system is necessary.
Collapse
Affiliation(s)
- Kazuo Murakami
- Department of Health Care and Preventive Medicine, Matsuyama Red Cross Hospital, 1 Bunkyo-cho, Matsuyama, Ehime, 790-8524, Japan.
| |
Collapse
|
41
|
Dalila N, Brockmöller J, Tzvetkov MV, Schirmer M, Haubrock M, Vormfelde SV. Impact of mineralocorticoid receptor polymorphisms on urinary electrolyte excretion with and without diuretic drugs. Pharmacogenomics 2015; 16:115-27. [DOI: 10.2217/pgs.14.163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Polymorphisms in the mineralocorticoid receptor may affect urinary sodium and potassium excretion. We investigated polymorphisms in the MR gene in relation to urinary electrolyte excretion in two separate studies. Patients & methods: The genotype–phenotype association was studied in healthy volunteers after single doses of bumetanide, furosemide, torsemide, hydrochlorothiazide, triamterene and after NaCl restriction. Results: High potassium excretion under all conditions except torsemide, and high NaCl excretion after bumetanide and furosemide were associated with the A allele of the intron-3 polymorphism (rs3857080). This polymorphism explained 5–10% of the functional variation and in vitro, rs3857080 affected DNA binding of the transcription factor LHX4. Conclusion: rs3857080 may be a promising new candidate for research in cardiac and renal disorders and on antialdosteronergic drugs like spironolactone. Original submitted 23 June 2014; Revision submitted 5 November 2014
Collapse
Affiliation(s)
- Nawar Dalila
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Brockmöller
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Markus Schirmer
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Haubrock
- Institute of Bioinformatics, University Medical Center Göttingen, Göttingen, Germany
| | | |
Collapse
|
42
|
Marques FZ, Charchar FJ. microRNAs in Essential Hypertension and Blood Pressure Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:215-35. [PMID: 26663185 DOI: 10.1007/978-3-319-22671-2_11] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Unravelling the complete genetic predisposition to high blood pressure (BP) has proven to be challenging. This puzzle and the fact that coding regions of the genome account for less than 2 % of the entire human DNA support the hypothesis that mechanisms besides coding genes are likely to contribute to BP regulation. Non-coding RNAs, especially microRNAs, are emerging as key players of transcription regulation in both health and disease states. They control basic functions in virtually all cell types relevant to the cardiovascular system and, thus, a direct involvement with BP regulation is highly probable. Here we review the literature about microRNAs associated with regulation of BP and hypertension, highlighting investigations, methodology and difficulties arising in the field. These molecules are being studied for exploitation in diagnostics, prognostics and therapeutics in many diseases. There have been some studies that examined biological fluid microRNAs as biomarkers for hypertension, but most remain inconclusive due to the small sample sizes and differences in methodological standardisation. Fewer studies have analysed tissue microRNA levels in vascular smooth muscle cells and the kidney. Others focused on the interaction between single nucleotide polymorphisms and microRNA binding sites. Studies in animals have shown that angiotensin II, high-salt diet and exercise change microRNA levels in hypertension. Treatment of spontaneously hypertensive rats with a miR-22 inhibitor and treatment of hypertensive Schlager BPH/2J mice with a miR-181a mimic decreased their BP. This supports the use of microRNAs as therapeutic targets in hypertension, and future studies should test the use of other microRNAs found in human association studies. In conclusion, there is a clear need of increased pace of human, animal and functional studies to help us understand the multifaceted roles of microRNAs as critical regulators of the development and physiology of BP.
Collapse
Affiliation(s)
- Francine Z Marques
- School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia.,Heart Failure Research Group, Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Fadi J Charchar
- School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia.
| |
Collapse
|
43
|
Abstract
Cardiovascular disease remains the most prevalent cause of human morbidity and mortality in ageing Western societies. Basic and translational scientific efforts have focused on the development and improvement of diagnostic and therapeutic strategies to limit the burden of associated diseases, such as stroke and myocardial infarction, and diabetes mellitus and arterial hypertension. Progress in molecular medicine and biology has unravelled a complex epigenetic and post-transcriptional gene-regulating machinery in humans which may limit disease development. An increasing number of attractive molecular strategies, which use the potential of modulating noncoding RNAs, have surfaced over the last decade. Currently, the most extensively studied gene-regulating RNA subspecies are microRNAs, which have been shown to adjust the translational output of coding transcripts by enforcing their degradation and inhibiting their translation into protein. Key findings indicate that microRNAs act as crucial regulators in the majority of human pathologies. Thus, recent research has focused on detecting and modulating microRNAs for therapeutic and biomarker purposes. This review focuses on main and repeated discoveries regarding the role and the therapeutic and biomarker feasibility of microRNAs during cardiovascular disease development and exacerbation.
Collapse
Affiliation(s)
- L Maegdefessel
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
The emerging role of non-coding RNA in essential hypertension and blood pressure regulation. J Hum Hypertens 2014; 29:459-67. [PMID: 25391760 DOI: 10.1038/jhh.2014.99] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 09/19/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022]
Abstract
Unravelling the complete genetic predisposition to high blood pressure (BP) has proven to be challenging. This puzzle and the fact that coding regions of the genome account for less than 2% of the entire human DNA support the hypothesis that genetic mechanism besides coding genes are likely to contribute to BP regulation. Non-coding RNAs (ncRNAs) are emerging as key players of transcription regulation in both health and disease states. They control basic functions in virtually all cell types relevant to the cardiovascular system and, thus, a direct involvement with BP regulation is highly probable. Here, we review the literature about ncRNAs associated with human BP and essential hypertension, highlighting investigations, methodology and difficulties arising in the field. The most investigated ncRNAs so far are microRNAs (miRNAs), small ncRNAs that modulate gene expression by posttranscriptional mechanisms. We discuss studies that have examined miRNAs associated with BP in biological fluids, such as blood and urine, and tissues, such as vascular smooth muscle cells and the kidney. Furthermore, we review the interaction between miRNA binding sites and single nucleotide polymorphisms in genes associated with BP. In conclusion, there is a clear need for more human and functional studies to help elucidate the multifaceted roles of ncRNAs, in particular mid- and long ncRNAs in BP regulation.
Collapse
|
45
|
A Common Polymorphism Within MSLN Affects miR-611 Binding Site and Soluble Mesothelin Levels in Healthy People. J Thorac Oncol 2014; 9:1662-8. [DOI: 10.1097/jto.0000000000000322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Edinger RS, Coronnello C, Bodnar AJ, Labarca M, Bhalla V, LaFramboise WA, Benos PV, Ho J, Johnson JP, Butterworth MB. Aldosterone regulates microRNAs in the cortical collecting duct to alter sodium transport. J Am Soc Nephrol 2014; 25:2445-57. [PMID: 24744440 PMCID: PMC4214524 DOI: 10.1681/asn.2013090931] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 02/05/2014] [Indexed: 12/26/2022] Open
Abstract
A role for microRNAs (miRs) in the physiologic regulation of sodium transport in the kidney has not been established. In this study, we investigated the potential of aldosterone to alter miR expression in mouse cortical collecting duct (mCCD) epithelial cells. Microarray studies demonstrated the regulation of miR expression by aldosterone in both cultured mCCD and isolated primary distal nephron principal cells. Aldosterone regulation of the most significantly downregulated miRs, mmu-miR-335-3p, mmu-miR-290-5p, and mmu-miR-1983 was confirmed by quantitative RT-PCR. Reducing the expression of these miRs separately or in combination increased epithelial sodium channel (ENaC)-mediated sodium transport in mCCD cells, without mineralocorticoid supplementation. Artificially increasing the expression of these miRs by transfection with plasmid precursors or miR mimic constructs blunted aldosterone stimulation of ENaC transport. Using a newly developed computational approach, termed ComiR, we predicted potential gene targets for the aldosterone-regulated miRs and confirmed ankyrin 3 (Ank3) as a novel aldosterone and miR-regulated protein. A dual-luciferase assay demonstrated direct binding of the miRs with the Ank3-3' untranslated region. Overexpression of Ank3 increased and depletion of Ank3 decreased ENaC-mediated sodium transport in mCCD cells. These findings implicate miRs as intermediaries in aldosterone signaling in principal cells of the distal kidney nephron.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - John P Johnson
- Renal-Electrolyte Division, Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Michael B Butterworth
- Renal-Electrolyte Division, Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
47
|
Welten SM, Bastiaansen AJ, de Jong RC, de Vries MR, Peters EA, Boonstra MC, Sheikh SP, La Monica N, Kandimalla ER, Quax PH, Nossent AY. Inhibition of 14q32 MicroRNAs miR-329, miR-487b, miR-494, and miR-495 Increases Neovascularization and Blood Flow Recovery After Ischemia. Circ Res 2014; 115:696-708. [DOI: 10.1161/circresaha.114.304747] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
Effective neovascularization is crucial for recovery after cardiovascular events.
Objective:
Because microRNAs regulate expression of up to several hundred target genes, we set out to identify microRNAs that target genes in all pathways of the multifactorial neovascularization process. Using
www.targetscan.org
, we performed a reverse target prediction analysis on a set of 197 genes involved in neovascularization. We found enrichment of binding sites for 27 microRNAs in a single microRNA gene cluster. Microarray analyses showed upregulation of 14q32 microRNAs during neovascularization in mice after single femoral artery ligation.
Methods and Results:
Gene silencing oligonucleotides (GSOs) were used to inhibit 4 14q32 microRNAs, miR-329, miR-487b, miR-494, and miR-495, 1 day before double femoral artery ligation. Blood flow recovery was followed by laser Doppler perfusion imaging. All 4 GSOs clearly improved blood flow recovery after ischemia. Mice treated with GSO-495 or GSO-329 showed increased perfusion already after 3 days (30% perfusion versus 15% in control), and those treated with GSO-329 showed a full recovery of perfusion after 7 days (versus 60% in control). Increased collateral artery diameters (arteriogenesis) were observed in adductor muscles of GSO-treated mice, as well as increased capillary densities (angiogenesis) in the ischemic soleus muscle. In vitro, treatment with GSOs led to increased sprout formation and increased arterial endothelial cell proliferation, as well as to increased arterial myofibroblast proliferation.
Conclusions:
The 14q32 microRNA gene cluster is highly involved in neovascularization. Inhibition of 14q32 microRNAs miR-329, miR-487b, miR-494, and miR-495 provides a promising tool for future therapeutic neovascularization.
Collapse
Affiliation(s)
- Sabine M.J. Welten
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Antonius J.N.M. Bastiaansen
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Rob C.M. de Jong
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Margreet R. de Vries
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Erna A.B. Peters
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Martin C. Boonstra
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Søren P. Sheikh
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Nicola La Monica
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Ekambar R. Kandimalla
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - Paul H.A. Quax
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| | - A. Yaël Nossent
- From the Department of Surgery (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., M.C.B., P.H.A.Q., A.Y.N.) and Einthoven Laboratory for Experimental Vascular Medicine (S.M.J.W., A.J.N.M.B., R.C.M.d.J., M.R.d.V., E.A.B.P., P.H.A.Q., A.Y.N.), Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark (S.P.S.); and Idera Pharmaceuticals, Cambridge, MA (N.L.M., E.R.K.)
| |
Collapse
|
48
|
Boštjančič E, Glavač D. miRNome in myocardial infarction: Future directions and perspective. World J Cardiol 2014; 6:939-958. [PMID: 25276296 PMCID: PMC4176804 DOI: 10.4330/wjc.v6.i9.939] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/28/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs), which are small and non-coding RNAs, are genome encoded from viruses to humans. They contribute to various developmental, physiological and pathological processes in living organisms. A huge amount of research results revealed that miRNAs regulate these processes also in the heart. miRNAs may have cell-type-specific or tissue-specific expression patterns or may be expressed ubiquitously. Primary studies of miRNA involvement in hypertrophy, heart failure and myocardial infarction analyzed miRNAs that are enriched in or specific for cardiomyocytes; however, growing evidence suggest that other miRNAs, not cardiac or muscle-specific, play a significant role in cardiovascular disease. Abnormal miRNA regulation has been shown to be involved in cardiac diseases, suggesting that miRNAs might affect cardiac structure and function. In this review, we focus on miRNAs that have been found to contribute to the pathogenesis of myocardial infarction (MI) and the response post-MI and characterized as diagnostic, prognostic and therapeutic targets. The majority of these studies were performed using mouse and rat models of MI, with a focus on the identification of basic cellular and molecular pathways involved in MI and in the response post-MI. Much research has also been performed on animal and human plasma samples from MI individuals to identify miRNAs that are possible prognostic and/or diagnostic targets of MI and other MI-related diseases. A large proportion of research is focused on miRNAs as promising therapeutic targets and biomarkers of drug responses and/or stem cell treatment approaches. However, only a few studies have described miRNA expression in human heart tissue following MI.
Collapse
|
49
|
Pivarcsi A, Ståhle M, Sonkoly E. Genetic polymorphisms altering microRNA activity in psoriasis - a key to solve the puzzle of missing heritability? Exp Dermatol 2014; 23:620-4. [DOI: 10.1111/exd.12469] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2014] [Indexed: 12/15/2022]
Affiliation(s)
- Andor Pivarcsi
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| | - Mona Ståhle
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| | - Enikő Sonkoly
- Unit of Dermatology and Venerology; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
50
|
Abstract
According to the World Health Organization, cardiovascular disease accounts for approximately 30% of all deaths in the United States, and is the worldwide leading cause of morbidity and mortality. Over the last several years, microRNAs have emerged as critical regulators of physiological homeostasis in multiple organ systems, including the cardiovascular system. The focus of this review is to provide an overview of the current state of knowledge of the molecular mechanisms contributing to the multiple causes of cardiovascular disease with respect to regulation by microRNAs. A major challenge in understanding the roles of microRNAs in the pathophysiology of cardiovascular disease is that cardiovascular disease may arise from perturbations in intracellular signaling in multiple cell types including vascular smooth muscle and endothelial cells, cardiac myocytes and fibroblasts, as well as hepatocytes, pancreatic β-cells, and others. Additionally, perturbations in intracellular signaling cascades may also have profound effects on heterocellular communication via secreted cytokines and growth factors. There has been much progress in recent years to identify the microRNAs that are both dysregulated under pathological conditions, as well as the signaling pathway(s) regulated by an individual microRNA. The goal of this review is to summarize what is currently known about the mechanisms whereby microRNAs maintain cardiovascular homeostasis and to attempt to identify some key unresolved questions that require further study.
Collapse
Affiliation(s)
- Ronald L Neppl
- Boston Children's Hospital, Department of Cardiology ; Harvard Medical School, Department of Pediatrics Boston MA, 02115
| | - Da-Zhi Wang
- Boston Children's Hospital, Department of Cardiology ; Harvard Medical School, Department of Pediatrics Boston MA, 02115
| |
Collapse
|