1
|
Chen XJ, Liu SY, Li SM, Feng JK, Hu Y, Cheng XZ, Hou CZ, Xu Y, Hu M, Feng L, Xiao L. The recent advance and prospect of natural source compounds for the treatment of heart failure. Heliyon 2024; 10:e27110. [PMID: 38444481 PMCID: PMC10912389 DOI: 10.1016/j.heliyon.2024.e27110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Heart failure is a continuously developing syndrome of cardiac insufficiency caused by diseases, which becomes a major disease endangering human health as well as one of the main causes of death in patients with cardiovascular diseases. The occurrence of heart failure is related to hemodynamic abnormalities, neuroendocrine hormones, myocardial damage, myocardial remodeling etc, lead to the clinical manifestations including dyspnea, fatigue and fluid retention with complex pathophysiological mechanisms. Currently available drugs such as cardiac glycoside, diuretic, angiotensin-converting enzyme inhibitor, vasodilator and β receptor blocker etc are widely used for the treatment of heart failure. In particular, natural products and related active ingredients have the characteristics of mild efficacy, low toxicity, multi-target comprehensive efficacy, and have obvious advantages in restoring cardiac function, reducing energy disorder and improving quality of life. In this review, we mainly focus on the recent advance including mechanisms and active ingredients of natural products for the treatment of heart failure, which will provide the inspiration for the development of more potent clinical drugs against heart failure.
Collapse
Affiliation(s)
- Xing-Juan Chen
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | - Si-Yuan Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Si-Ming Li
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | | | - Ying Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Xiao-Zhen Cheng
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | - Cheng-Zhi Hou
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | - Yun Xu
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | - Mu Hu
- Peking University International Hospital, Beijing, 102206, China
| | - Ling Feng
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| | - Lu Xiao
- China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, 100053, China
| |
Collapse
|
2
|
Hu Y, Jin L, Wang Z. Genome-wide association study of dilated cardiomyopathy-induced heart failure associated with renal insufficiency in a Chinese population. BMC Cardiovasc Disord 2023; 23:335. [PMID: 37391705 PMCID: PMC10314512 DOI: 10.1186/s12872-023-03370-0] [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: 10/20/2022] [Accepted: 06/28/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND As it is unclear whether there is genetic susceptibility to cardiorenal syndrome (CRS), we conducted a genome-wide association study of dilated cardiomyopathy (DCM)-induced heart failure (HF) associated with renal insufficiency (RI) in a Chinese population to identify putative susceptibility variants and culprit genes. METHODS A total of 99 Han Chinese patients with DCM-induced chronic HF were selected and divided into one of three groups, namely, HF with normal renal function (Group 1), HF with mild RI (Group 2) and HF with moderate to severe RI (Group 3). Genomic DNA was extracted from each subject for genotyping. RESULTS According to Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, top 10 lists of molecular function, cell composition and biological process of differential target genes and 15 signalling pathways were discriminated among the three groups. Additionally, sequencing results identified 26 significantly different single-nucleotide polymorphisms (SNPs) in the 15 signalling pathways, including three SNPs (rs57938337, rs6683225 and rs6692782) in ryanodine receptor 2 (RYR2) and two SNPs (rs12439006 and rs16958069) in RYR3. The genotype and allele frequencies of the five SNPs in RYR2 and RYR3 were significantly differential between HF (Group 1) and CRS (Group 2 + 3) patients. CONCLUSION Twenty-six significantly different SNP loci in 17 genes of the 15 KEGG pathways were found in the three patient groups. Among these variants, rs57938337, rs6683225 and rs6692782 in RYR2 and rs12439006 and rs16958069 in RYR3 are associated with RI in Han Chinese patients with heart failure, suggesting that these variants may be used to identify patients susceptible to CRS in the future.
Collapse
Affiliation(s)
- Yuexin Hu
- Department of Cardiovascular Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 246 Guangzhou Road, Nanjing, Jiangsu, 210008, China
- Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| | - Liangli Jin
- Department of Cardiovascular Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 246 Guangzhou Road, Nanjing, Jiangsu, 210008, China
- Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China
| | - Zhi Wang
- Department of Cardiovascular Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, No. 246 Guangzhou Road, Nanjing, Jiangsu, 210008, China.
- Department of Cardiovascular Medicine, Nanjing Chest Hospital, Nanjing, China.
| |
Collapse
|
3
|
Zhang P, Huang L, Li X, Hu F, Niu X, Sun Y, Yao W, Tian W. NF1-Related MicroRNA Gene Polymorphisms and the Susceptibility to Soft Tissue Sarcomas: A Case-Control Study. DNA Cell Biol 2023; 42:229-238. [PMID: 36989515 DOI: 10.1089/dna.2022.0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Soft tissue sarcomas (STS) are rare malignant tumors of mesenchymal origin, which are easy to metastasize and relapse and are a great threat to human health. In our previous study, the abnormal expression of neurofibromin 1 (NF1) is observed in tumor tissue of STS, and the NF1 gene is regulated by miRNAs. The study aimed to assess the association between NF1-related miRNA gene polymorphisms and the risk of STS. In this case-control study, the information and peripheral blood were collected from 169 patients with STS and 170 healthy controls. Six single-nucleotide polymorphisms of the NF1-related miRNAs were investigated and genotyped using a Sequenom MassARRAY® matrix-assisted laser desorption/ionization time-of-flight mass spectrometry platform. The association between the polymorphisms and the risk of STS was estimated using unconditional logistic regression analysis. There was a significant statistical difference on genotype distribution of miR-199a2 rs12139213 between the case group and the control group (p = 0.026). Comparing with individuals with wild-type AA, individuals with the AT/TT genotype had a 1.753-fold (odds ratio [OR] = 1.753, 95% confidence interval [CI] = 1.090-2.819, p = 0.021) increased risk of STS and 1.907-fold (OR = 1.907, 95% CI = 1.173-3.102, p = 0.009) increased risk of STS adjusted for age and smoking status. Individuals with the AG/GG genotype for miR24-3p rs4743988 displayed a significantly reduced risk of STS compared with individuals with homozygous mutations AA (OR = 0.605, 95% CI = 0.376-0.973, p = 0.038). Individuals carrying the AT/TT genotype for miR-199a2 rs12139213 or the AA genotype for miR24-3p rs4743988 may be susceptible to STS, which could be potential biomarkers for the diagnosis of STS.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Lingling Huang
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Xinling Li
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoying Niu
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Yang Sun
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Weitao Yao
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Wen Tian
- Department of Bone and Soft Tissue Cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| |
Collapse
|
4
|
Zhang J, Zhang Z, Nie X, Liu Y, Qi Y, Wang J. Deregulated RNAs involved in sympathetic regulation of sepsis-induced acute lung injury based on whole transcriptome sequencing. BMC Genomics 2022; 23:836. [PMID: 36526959 PMCID: PMC9758828 DOI: 10.1186/s12864-022-09073-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Sympathetic nerves play essential roles in the regulation of lung inflammation, and we investigated the effect of sympathetic denervation (SD) on sepsis-induced acute lung injury (ALI) in mice. Mice were randomized to the control, SD, ALI and SD + ALI, groups. SD and ALI were established through intratracheal 6-hydroxydopamine and intraperitoneal lipopolysaccharide, respectively. Models and gene expressions levels were evaluated by HE staining, ELISA, Western blotting and RT-qPCR. RNA extraction, whole transcriptome sequencing and subsequent biostatistical analysis were performed. Sympathetic denervation in the lungs significantly attenuated lung TNF-ɑ and norepinephrine expression, alleviated sepsis-induced acute lung injury and inhibited NF-κB signaling. Compared with the ALI group, the SD + ALI group exhibited 629 DE circRNAs, 269 DE lncRNAs,7 DE miRNAs and 186 DE mRNAs, respectively. Some DE RNAs were validated by RT-qPCR. CircRNA-miRNA-mRNA regulatory networks in the SD + ALI group revealed enrichment of the B-cell receptor signaling pathway, IL-17 signaling pathway, neuroactive ligand-receptor interaction, CAM, primary immunodeficiency, and cytokine-cytokine receptor interaction terms. The lncRNA-miRNA-mRNA network also revealed inflammation-related signaling pathways. Taken together, based on the successfully established models of SD and ALI, we show here that sympathetic nerves may regulate sepsis-induced ALI supposedly by affecting the expression of circRNAs, lncRNAs, miRNAs, and mRNAs in the lungs. These results may allow for further exploration of the roles of pulmonary sympathetic nerves in sepsis-induced ALI.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Zhao Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinran Nie
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Yingli Liu
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| | - Jing Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
5
|
Nwokafor C, Serova LI, Tanelian A, Nahvi RJ, Sabban EL. Variable Response of Norepinephrine Transporter to Traumatic Stress and Relationship to Hyperarousal. Front Behav Neurosci 2021; 15:725091. [PMID: 34650410 PMCID: PMC8507558 DOI: 10.3389/fnbeh.2021.725091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
The noradrenergic systems play a key role in stress triggered disorders such as post-traumatic stress disorder (PTSD). We hypothesized that traumatic stress will alter expression of norepinephrine transporter (NET) in locus coeruleus (LC) and its target brain regions which could be related to hyperarousal. Male Sprague-Dawley rats were subjected to single prolonged stress (SPS) and several weeks later the LC was isolated. NET mRNA levels in LC, determined by RT-PCR, displayed variable response with high and low responsive subgroups. In different cohort, acoustic startle response (ASR) was measured 2 weeks after SPS and levels of NET mRNA and protein in LC determined. The high NET responsive subgroup had greater hyperarousal. Nevertheless, NET protein levels, as determined by western blots, were lower than unstressed controls in LC, ventral hippocampus and medial prefrontal cortex and displayed considerable variability. Hypermethylation of specific CpG region in promoter of SLC6A2 gene, encoding NET, was present in the low, but not high, NET mRNA responsive subgroup. Taken together, the results demonstrate variability in stress elicited changes in NET gene expression and involvement of epigenetic changes. This may underlie mechanisms of susceptibility and resilience to traumatic stress triggered neuropsychiatric symptoms, especially hyperarousal.
Collapse
Affiliation(s)
- Chiso Nwokafor
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Arax Tanelian
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Roxanna J Nahvi
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
6
|
Scherf-Clavel M, Weber H, Deckert J, Erhardt-Lehmann A. The role of pharmacogenetics in the treatment of anxiety disorders and the future potential for targeted therapeutics. Expert Opin Drug Metab Toxicol 2021; 17:1249-1260. [PMID: 34643143 DOI: 10.1080/17425255.2021.1991912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Anxiety disorders (AD) are among the most common mental disorders worldwide. Pharmacotherapy, including benzodiazepines, selective serotonin reuptake inhibitors, serotonin-norepinephrine reuptake inhibitors, and tricyclic antidepressants is currently based on 'trial-and-error,' and is effective in a subset of patients or produces partial response only. Recent research proposes that treatment response and tolerability of the drugs are associated with genetic factors. AREAS COVERED In the present review, we provide information on pharmacogenetics (PGx) in AD, including pharmacokinetic and pharmacodynamic genes. Moreover, we discuss the future potential of PGx for personalized treatment. EXPERT OPINION In psychiatry, PGx testing is still in its infancy, especially in the treatment of AD. As of today, implementation in clinical routine is recommended only for CYP2D6 and CYP2C19, mainly in terms of safety of treatment and potentially of treatment outcome in general. However, the evidence for PGx testing addressing pharmacodynamics for specific AD is limited to date. Nevertheless, PGx may develop into a valuable and promising tool to improve therapy in AD, but there is a need for more research to fully exploit its possibilities. Future perspectives include research into single genes, polygenic risk scores, and pharmacoepigenetics to provide targeted therapy.
Collapse
Affiliation(s)
- Maike Scherf-Clavel
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Heike Weber
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Angelika Erhardt-Lehmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany.,Translational Department, Max Planck Institute for Psychiatry, München, Germany
| |
Collapse
|
7
|
Eikelis N, Dixon JB, Lambert EA, Hanin G, Tzur Y, Greenberg DS, Soreq H, Marques FZ, Fahey MT, Head GA, Schlaich MP, Lambert GW. MicroRNA-132 may be associated with blood pressure and liver steatosis-preliminary observations in obese individuals. J Hum Hypertens 2021; 36:911-916. [PMID: 34453104 DOI: 10.1038/s41371-021-00597-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 11/09/2022]
Abstract
Recent findings in experimental models have shown that the microRNA miR-132 (mir-132) is an important regulator of liver homeostasis and lipid metabolism. We aimed to assess miR-132 expression in liver and fat tissues of obese individuals and examine its association with blood pressure (BP) and hepatic steatosis. We examined obese individuals undergoing bariatric surgery for weight loss (n = 19). Clinical and demographic information was obtained. Quantitative PCR was performed to determine tissue expression of miR-132 in liver and subcutaneous and visceral fat biopsies obtained during bariatric surgery. Liver biopsies were read by a single liver pathologist and graded for steatosis, inflammation and fibrosis. Participants (aged 39 ± 8.1 years) had a body mass index (BMI) of 42 ± 4.5 kg/m2 and presented with 2.2 ± 1.2 metabolic abnormalities. Supine BP was 127 ± 16/74 ± 11 mmHg. Hepatic and visceral fat expression of miR-132 were correlated (r = 0.59, P = 0.033). There was no correlation between subcutaneous and visceral expression of miR-132 (r = -0.31, P = 0.20). Hepatic and visceral fat miR-132 expression were associated with BMI (r = 0.62 and r = 0.68, P = 0.049 respectively) and degree of liver steatosis (r = 0.60 and r = 0.55, P < 0.05, respectively). Subcutaneous fat miRNA-132 expression was correlated to office systolic BP (r = 0.46, P < 0.05), several aspects of 24 h BP (24 h systolic BP: r = 0.52; day systolic BP: r = 0.59, P < 0.05 for all), plasma triglycerides (r = 0.51, P < 0.01) and liver enzymes (ALT: r = -0.52; AST: r = -0.48, P < 0.05 for all). We found an association between miR-132 and markers of cardiovascular and metabolic disease. Reduction of miR-132 may be a target for the regulation of liver lipid homeostasis and control of obesity-related blood pressure.
Collapse
Affiliation(s)
- Nina Eikelis
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - John B Dixon
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Elisabeth A Lambert
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Geula Hanin
- Department of Genetics, University of Cambridge, Cambridge, UK.,The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonat Tzur
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David S Greenberg
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, VIC, Australia
| | - Michael T Fahey
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Geoffrey A Head
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Markus P Schlaich
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.,Dobney Hypertension Centre, School of Medicine-Royal Perth Hospital Unit, University of Western Australia, Perth, WA, Australia
| | - Gavin W Lambert
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia. .,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
Fichna JP, Humińska-Lisowska K, Safranow K, Adamczyk JG, Cięszczyk P, Żekanowski C, Berdyński M. Rare Variant in the SLC6A2 Encoding a Norepinephrine Transporter Is Associated with Elite Athletic Performance in the Polish Population. Genes (Basel) 2021; 12:genes12060919. [PMID: 34203885 PMCID: PMC8232774 DOI: 10.3390/genes12060919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Numerous genetic factors have been shown to influence athletic performance, but the list is far from comprehensive. In this study, we analyzed genetic variants in two genes related to mental abilities, SLC6A2 (rs1805065) and SYNE1 (rs2635438) in a group of 890 athletes (320 endurance, 265 power, and 305 combat athletes) vs. 1009 sedentary controls. Genotyping of selected SNPs was performed using TaqMan SNP genotyping assays. SLC6A2 codes for norepinephrine transporter, a protein involved in modulating mood, arousal, memory, learning, and pain perception, while SYNE1 encodes protein important for the maintenance of the cerebellum—the part of the brain that coordinates complex body movements. Both SNPs (rs2635438 and rs1805065) showed no statistically significant differences between the frequencies of variants in the athletes and the sedentary controls (athletes vs. control group) or in the athlete subgroups (martial vs. control, endurance vs. control, and power vs. control). The rs1805065 T variant of SLC6A2 was found to be overrepresented in male high-elite martial sports athletes when compared to sedentary controls (OR = 6.56, 95%CI = 1.82–23.59, p = 0.010). This supports the hypothesis that genetic variants potentially affecting brain functioning can influence elite athletic performance and indicate the need for further genetic association studies, as well as functional analyses.
Collapse
Affiliation(s)
- Jakub P. Fichna
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.P.F.); (C.Ż.)
| | - Kinga Humińska-Lisowska
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (K.H.-L.); (P.C.)
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-204 Szczecin, Poland;
| | - Jakub G. Adamczyk
- Department of Theory of Sport, Józef Piłsudski University of Physical Education, 00-968 Warsaw, Poland;
| | - Paweł Cięszczyk
- Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland; (K.H.-L.); (P.C.)
| | - Cezary Żekanowski
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.P.F.); (C.Ż.)
| | - Mariusz Berdyński
- Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.P.F.); (C.Ż.)
- Correspondence: ; Tel.: +48-226-086-485
| |
Collapse
|
9
|
Martins HC, Schratt G. MicroRNA-dependent control of neuroplasticity in affective disorders. Transl Psychiatry 2021; 11:263. [PMID: 33941769 PMCID: PMC8093191 DOI: 10.1038/s41398-021-01379-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/17/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Affective disorders are a group of neuropsychiatric disorders characterized by severe mood dysregulations accompanied by sleep, eating, cognitive, and attention disturbances, as well as recurring thoughts of suicide. Clinical studies consistently show that affective disorders are associated with reduced size of brain regions critical for mood and cognition, neuronal atrophy, and synaptic loss in these regions. However, the molecular mechanisms that mediate these changes and thereby increase the susceptibility to develop affective disorders remain poorly understood. MicroRNAs (miRNAs or miRs) are small regulatory RNAs that repress gene expression by binding to the 3'UTR of mRNAs. They have the ability to bind to hundreds of target mRNAs and to regulate entire gene networks and cellular pathways implicated in brain function and plasticity, many of them conserved in humans and other animals. In rodents, miRNAs regulate synaptic plasticity by controlling the morphology of dendrites and spines and the expression of neurotransmitter receptors. Furthermore, dysregulated miRNA expression is frequently observed in patients suffering from affective disorders. Together, multiple lines of evidence suggest a link between miRNA dysfunction and affective disorder pathology, providing a rationale to consider miRNAs as therapeutic tools or molecular biomarkers. This review aims to highlight the most recent and functionally relevant studies that contributed to a better understanding of miRNA function in the development and pathogenesis of affective disorders. We focused on in vivo functional studies, which demonstrate that miRNAs control higher brain functions, including mood and cognition, in rodents, and that their dysregulation causes disease-related behaviors.
Collapse
Affiliation(s)
- Helena Caria Martins
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, 8057, Zurich, Switzerland
| | - Gerhard Schratt
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, 8057, Zurich, Switzerland.
| |
Collapse
|
10
|
Nuclear functions of microRNAs relevant to the cardiovascular system. Transl Res 2021; 230:151-163. [PMID: 33186782 DOI: 10.1016/j.trsl.2020.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022]
Abstract
A fraction of the transcriptome is translated into proteins. The rest is classified as non-protein coding RNA (Ribonucleic Acid) but has gained increased attention as functional and regulatory group of transcripts. The gene regulatory role of non-coding RNAs (ncRNAs) has now been widely accepted in diverse biological processes in both physiology and disease. MicroRNAs fall into this latter group and are widely known for their diverse post-transcriptional regulatory role. MicroRNA sequences are embedded in the long ncRNAs, known as primary microRNAs, are processed into precursor microRNAs and are typically transported out of the nucleus for maturation and loading into a protein complex forming RNA-induced silencing complex (RISC) that either drives the degradation of messenger RNA (mRNA) or blocks its translation. A new phenomenon is emerging where microRNAs have active roles within the nucleus. The presence of RISC components including microRNAs in the nucleus supports this notion. They may integrate with chromatin modifiers, microprocessing machinery and mRNA stabilizing transcripts to play a multifunctional role in the nucleus. Although a limited number of studies appreciate this novel activity of microRNAs relevant to the cardiovascular system, they provide proof-of-concept that requires consideration while targeting miRNAs with therapeutic potential.
Collapse
|
11
|
Wadhawan A, Reynolds MA, Makkar H, Scott AJ, Potocki E, Hoisington AJ, Brenner LA, Dagdag A, Lowry CA, Dwivedi Y, Postolache TT. Periodontal Pathogens and Neuropsychiatric Health. Curr Top Med Chem 2021; 20:1353-1397. [PMID: 31924157 DOI: 10.2174/1568026620666200110161105] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 02/08/2023]
Abstract
Increasing evidence incriminates low-grade inflammation in cardiovascular, metabolic diseases, and neuropsychiatric clinical conditions, all important causes of morbidity and mortality. One of the upstream and modifiable precipitants and perpetrators of inflammation is chronic periodontitis, a polymicrobial infection with Porphyromonas gingivalis (P. gingivalis) playing a central role in the disease pathogenesis. We review the association between P. gingivalis and cardiovascular, metabolic, and neuropsychiatric illness, and the molecular mechanisms potentially implicated in immune upregulation as well as downregulation induced by the pathogen. In addition to inflammation, translocation of the pathogens to the coronary and peripheral arteries, including brain vasculature, and gut and liver vasculature has important pathophysiological consequences. Distant effects via translocation rely on virulence factors of P. gingivalis such as gingipains, on its synergistic interactions with other pathogens, and on its capability to manipulate the immune system via several mechanisms, including its capacity to induce production of immune-downregulating micro-RNAs. Possible targets for intervention and drug development to manage distal consequences of infection with P. gingivalis are also reviewed.
Collapse
Affiliation(s)
- Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Department of Psychiatry, Saint Elizabeths Hospital, Washington, D.C. 20032, United States
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, United States
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, United States
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, United States
| | - Andrew J Hoisington
- Air Force Institute of Technology, Wright-Patterson Air Force Base, United States
| | - Lisa A Brenner
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Christopher A Lowry
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Department of Integrative Physiology, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, United States
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Alabama, United States
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, United States
| |
Collapse
|
12
|
Warren WG, Papagianni EP, Stevenson CW, Stubbendorff C. In it together? The case for endocannabinoid-noradrenergic interactions in fear extinction. Eur J Neurosci 2021; 55:952-970. [PMID: 33759226 DOI: 10.1111/ejn.15200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/26/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022]
Abstract
Anxiety and trauma-related disorders, such as post-traumatic stress disorder (PTSD), are debilitating mental illnesses with great personal and socioeconomic costs. Examining memory formation and relevant behavioural responding associated with aversive stimuli may improve our understanding of the neurobiology underlying fear memory processing and PTSD treatment. The neurocircuitry underpinning learned fear and its inhibition through extinction is complex, involving synergistic interactions between different neurotransmitter systems in inter-connected brain areas. Endocannabinoid and noradrenergic transmission have both been implicated separately in fear memory processing and PTSD, but potential interactions between these systems in relation to fear extinction have received little attention to date. Their receptors are expressed together in brain areas crucial for fear extinction, which is enhanced by both cannabinoid and noradrenergic receptor activation in these areas. Moreover, cannabinoid signalling modulates the activity of locus coeruleus noradrenaline (NA) neurons and the release of NA in the medial prefrontal cortex, a brain area that is crucial for fear extinction. Interestingly, endocannabinoid-noradrenergic system interactions have been shown to regulate the encoding and retrieval of fear memory. Thus, noradrenergic regulation of fear extinction may also be driven indirectly in part via cannabinoid receptor signalling. In this perspective paper, we collate the available relevant literature and propose a synergistic role for the endocannabinoid and noradrenergic systems in regulating fear extinction, the study of which may further our understanding of the neurobiological substrates of PTSD and its treatment.
Collapse
Affiliation(s)
- William G Warren
- School of Biosciences, University of Nottingham, Loughborough, UK
| | | | - Carl W Stevenson
- School of Biosciences, University of Nottingham, Loughborough, UK
| | - Christine Stubbendorff
- School of Biosciences, University of Nottingham, Loughborough, UK.,Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
13
|
Trait anxiety, a personality risk factor associated with Alzheimer's Disease. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110124. [PMID: 33035604 DOI: 10.1016/j.pnpbp.2020.110124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in elderly population and the leading cause of dementia worldwide. While senile plaques and neurofibrillary tangles have been proposed as the principal histopathologic hallmarks of AD, the exact etiology of this disease is still far from being clearly understood. AD has been recognized as pathological consequences of complex interactions among genetic, aging, medical, life style and psychosocial factors. Recently, the roles of neuroticism personality traits in AD incidence and progression have come into focus. More specifically, increasing evidence has further shown that the trait anxiety, one major component of neuroticism predicting the individual vulnerability in response to stress, is a risk factor for AD and may correlated with various AD pathologies. In this review, we summarized recent literature on the association of trait anxiety with AD. We also discussed the possible neuroendocrinological and neurochemical mechanisms of this association, which may provide clinical implications for AD diagnosis and therapy.
Collapse
|
14
|
Yuan H, Jiao L, Yu N, Duan H, Yu Y, Bai Y. Histone Deacetylase 3-Mediated Inhibition of microRNA-19a-3p Facilitates the Development of Rheumatoid Arthritis-Associated Interstitial Lung Disease. Front Physiol 2020; 11:549656. [PMID: 33343379 PMCID: PMC7746846 DOI: 10.3389/fphys.2020.549656] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/02/2020] [Indexed: 11/22/2022] Open
Abstract
Histone deacetylase (HDAC) has been implicated in rheumatoid arthritis (RA) progression. We investigated the roles of histone deacetylase 3 (HDAC3) involved in RA-associated interstitial lung disease (ILD) fibrosis. Firstly, we measured the expression of HDAC3 and interleukin 17 receptor A (IL17RA) in lung tissue samples from normal controls, idiopathic pulmonary fibrosis (IPF) patients, and RA-ILD patients. Next, chromatin immunoprecipitation (ChIP) and dual luciferase reporter assay were employed to detect the interaction between HDAC3 and microRNA-19a-3p (miR-19a-3p) and between miR-19a-3p and IL17RA. Further, immunohistochemistry was used to localize HDAC3 and IL17RA expression in lung tissues. Additionally, functional assays were conducted followed by expression determination of HDAC3, miR-19a-3p, and IL17RA with reverse transcription quantitative PCR (RT-qPCR) and Western blot analysis. The effect of HDAC3 on RA-ILD in the constructed RA-ILD mouse model was also studied based on arthritis assessment. We found overexpressed HDAC3 and IL17RA as well as silenced miR-19a-3p in RA-ILD mouse model and RA-ILD patients. In the mouse model, HDAC3 downregulated miR-19a-3p in lung fibroblasts to promote the progression of RA-ILD fibrosis. In lung fibroblasts of RA-ILD mice, IL17RA was a target gene of miR-19a-3p. miR-19a-3p negatively regulated IL17RA, thereby increasing the expression of fibrosis markers, COL1A1, COL3A1, and FN, in lung fibroblasts of mice. Taken together, HDAC3 upregulated IL17RA expression by targeting miR-19a-3p to facilitate the RA-ILD fibrosis development, which sheds light on a new HDAC3/miR-19a-3p/IL17RA axis functioning in RA-ILD fibrosis.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Jiao
- Yanching Institute of Technology, Langfang, China
| | - Nan Yu
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Haifeng Duan
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yong Yu
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yanrong Bai
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
15
|
Muir J, Tse YC, Iyer ES, Biris J, Cvetkovska V, Lopez J, Bagot RC. Ventral Hippocampal Afferents to Nucleus Accumbens Encode Both Latent Vulnerability and Stress-Induced Susceptibility. Biol Psychiatry 2020; 88:843-854. [PMID: 32682566 DOI: 10.1016/j.biopsych.2020.05.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/25/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Stress is a major risk factor for depression, but not everyone responds to stress in the same way. Identifying why certain individuals are more susceptible is essential for targeted treatment and prevention. In rodents, nucleus accumbens (NAc) afferents from the ventral hippocampus (vHIP) are implicated in stress-induced susceptibility, but little is known about how this pathway might encode future vulnerability or specific behavioral phenotypes. METHODS We used fiber photometry to record in vivo activity in vHIP-NAc afferents during tests of depressive- and anxiety-like behavior in male and female mice, both before and after a sex-specific chronic variable stress protocol, to probe relationships between prestress neural activity and behavior and potential predictors of poststress behavioral adaptation. Furthermore, we examined chronic variable stress-induced alterations in vHIP-NAc activity in vivo and used ex vivo slice electrophysiology to identify the mechanism of this change. RESULTS We identified behavioral specificity of the vHIP-NAc pathway to anxiety-like and social interaction behavior. We also showed that this activity is broadly predictive of stress-induced susceptibility in both sexes, while prestress behavior is predictive only of anxiety-like behavior. We observed a stress-induced increase in in vivo vHIP-NAc activity coincident with an increase in spontaneous excitatory postsynaptic current frequency. CONCLUSIONS We implicate vHIP-NAc in social interaction and anxiety-like behavior and identify markers of vulnerability in this neural signal, with elevated prestress vHIP-NAc activity predicting increased susceptibility across behavioral domains. Our findings indicate that individual differences in neural activity and behavior play a role in predetermining susceptibility to later stress, providing insight into mechanisms of vulnerability.
Collapse
Affiliation(s)
- Jessie Muir
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Yiu Chung Tse
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | - Eshaan S Iyer
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Julia Biris
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | | | - Joëlle Lopez
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montréal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Montréal, Quebec, Canada.
| |
Collapse
|
16
|
Kucharik AH, Chang C. The Relationship Between Hypermobile Ehlers-Danlos Syndrome (hEDS), Postural Orthostatic Tachycardia Syndrome (POTS), and Mast Cell Activation Syndrome (MCAS). Clin Rev Allergy Immunol 2020; 58:273-297. [PMID: 31267471 DOI: 10.1007/s12016-019-08755-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, an association between hypermobile Ehlers-Danlos syndrome (hEDS), mast cell activation syndrome (MCAS), and postural orthostatic tachycardia syndrome (POTS) has garnered attention and patients are increasingly presenting with this triad. However, a real relationship between these entities is unclear due to a lack of scientific validity. We conducted an extensive review of the literature using two different search strategies. A narrower strategy included 88 searches of various combinations of terms for each of the three conditions, yielding 19 unique papers. A broader search included 136 searches of various combinations of terms but included all forms of EDS and yielded 40 unique papers. Of these, only four and nine papers from the narrower and broader search strategies were original research articles. None of these papers resulted from a combination of the search terms for the three conditions. All three clinical entities are controversial in either existence or pathogenesis. MCAS is a poorly defined clinical entity, and many studies do not adhere to the proposed criteria when establishing the diagnosis. Patients previously diagnosed with EDS hypermobility type may not meet the new, stricter criteria for hEDS but may for a less severe hypermobility spectrum disorder (HSD). The pathophysiology of POTS is still unclear. An evidence-based, common pathophysiologic mechanism between any of the two, much less all three conditions, has yet to be described. Our review of the literature shows that current evidence is lacking on the existence of MCAS or hEDS as separate or significant clinical entities. Studies proposing a relationship between the three clinical entities are either biased or based on outdated criteria. The reason behind the purported association of these entities stems from an overlapping pool of vague, subjective symptoms, which is inadequate evidence to conclude that any such relationship exists.
Collapse
Affiliation(s)
| | - Christopher Chang
- Florida Atlantic University, Boca Raton, FL, USA.
- Joe DiMaggio Children's Hospital, 1131 North 35th Avenue, Suite 220, Hollywood, FL, 33021, USA.
- University of California, Davis, CA, USA.
- Florida International University, Miami, FL, USA.
| |
Collapse
|
17
|
Jackson KL, Gueguen C, Lim K, Eikelis N, Stevenson ER, Charchar FJ, Lambert GW, Burke SL, Paterson MR, Marques FZ, Head GA. Neural suppression of miRNA-181a in the kidney elevates renin expression and exacerbates hypertension in Schlager mice. Hypertens Res 2020; 43:1152-1164. [PMID: 32427944 DOI: 10.1038/s41440-020-0453-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/05/2020] [Accepted: 04/14/2020] [Indexed: 11/09/2022]
Abstract
BPH/2J mice are a genetic model of hypertension with overactivity of the sympathetic nervous system (SNS) and renin-angiotensin system (RAS). BPH/2J display higher renal renin mRNA and low levels of its negative regulator microRNA-181a (miR-181a). We hypothesise that high renal SNS activity may reduce miR-181a expression, which contributes to elevated RAS activity and hypertension in BPH/2J. Our aim was to determine whether in vivo administration of a renal-specific miR-181a mimic or whether renal denervation could increase renal miR-181a abundance to reduce renal renin mRNA, RAS activity and hypertension in BPH/2J mice. Blood pressure (BP) in BPH/2J and normotensive BPN/3J mice was measured via radiotelemetry probes. Mice were administered miR-181a mimic or a negative control (1-25 nmol, i.v., n = 6-10) with BP measured for 48 h after each dose or they underwent renal denervation or sham surgery (n = 7-9). Injection of 5-25 nmol miR-181a mimic reduced BP in BPH/2J mice after 36-48 h (-5.3 ± 1.8, -6.1 ± 1.9 mmHg, respectively, P < 0.016). Treatment resulted in lower renal renin and inflammatory marker (TLR4) mRNA levels in BPH/2J. The mimic abolished the hypotensive effect of blocking the RAS with enalaprilat (P < 0.01). No differences between mimic or vehicle were observed in BPN/3J mice except for a higher level of renal angiotensinogen in the mimic-treated mice. Renal miR-181a levels that were lower in sham BPH/2J mice were greater following renal denervation and were thus similar to those of BPN/3J. Our findings suggest that the reduced renal miR-181a may partially contribute to the elevated BP in BPH/2J mice, through an interaction between the renal sympathetic nerves and miR-181a regulation of the RAS.
Collapse
Affiliation(s)
- Kristy L Jackson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Cindy Gueguen
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kyungjoon Lim
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Physiology, Anatomy & Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Nina Eikelis
- Human Neurotransmitters Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Emily R Stevenson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Fadi J Charchar
- Faculty of Science and Technology, Federation University Australia, Ballarat, VIC, Australia
| | - Gavin W Lambert
- Human Neurotransmitters Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Sandra L Burke
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Madeleine R Paterson
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC, Australia
| | - Francine Z Marques
- Faculty of Science and Technology, Federation University Australia, Ballarat, VIC, Australia.,Hypertension Research Laboratory, School of Biological Sciences, Monash University, Clayton, VIC, Australia.,Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Pharmacology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
18
|
Kaye DM, Shihata WA, Jama HA, Tsyganov K, Ziemann M, Kiriazis H, Horlock D, Vijay A, Giam B, Vinh A, Johnson C, Fiedler A, Donner D, Snelson M, Coughlan MT, Phillips S, Du XJ, El-Osta A, Drummond G, Lambert GW, Spector TD, Valdes AM, Mackay CR, Marques FZ. Deficiency of Prebiotic Fiber and Insufficient Signaling Through Gut Metabolite-Sensing Receptors Leads to Cardiovascular Disease. Circulation 2020; 141:1393-1403. [PMID: 32093510 DOI: 10.1161/circulationaha.119.043081] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND High blood pressure (BP) continues to be a major, poorly controlled but modifiable risk factor for cardiovascular death. Among key Western lifestyle factors, a diet poor in fiber is associated with prevalence of high BP. The impact of lack of prebiotic fiber and the associated mechanisms that lead to higher BP are unknown. Here we show that lack of prebiotic dietary fiber leads to the development of a hypertensinogenic gut microbiota, hypertension and its complications, and demonstrate a role for G-protein coupled-receptors (GPCRs) that sense gut metabolites. METHODS One hundred seventy-nine mice including C57BL/6J, gnotobiotic C57BL/6J, and knockout strains for GPR41, GPR43, GPR109A, and GPR43/109A were included. C57BL/6J mice were implanted with minipumps containing saline or a slow-pressor dose of angiotensin II (0.25 mg·kg-1·d-1). Mice were fed diets lacking prebiotic fiber with or without addition of gut metabolites called short-chain fatty acids ([SCFA)] produced during fermentation of prebiotic fiber in the large intestine), or high prebiotic fiber diets. Cardiac histology and function, BP, sodium and potassium excretion, gut microbiome, flow cytometry, catecholamines and methylation-wide changes were determined. RESULTS Lack of prebiotic fiber predisposed mice to hypertension in the presence of a mild hypertensive stimulus, with resultant pathological cardiac remodeling. Transfer of a hypertensinogenic microbiota to gnotobiotic mice recapitulated the prebiotic-deprived hypertensive phenotype, including cardiac manifestations. Reintroduction of SCFAs to fiber-depleted mice had protective effects on the development of hypertension, cardiac hypertrophy, and fibrosis. The cardioprotective effect of SCFAs were mediated via the cognate SCFA receptors GPR43/GPR109A, and modulated L-3,4-dihydroxyphenylalanine levels and the abundance of T regulatory cells regulated by DNA methylation. CONCLUSIONS The detrimental effects of low fiber Westernized diets may underlie hypertension, through deficient SCFA production and GPR43/109A signaling. Maintaining a healthy, SCFA-producing microbiota is important for cardiovascular health.
Collapse
Affiliation(s)
- David M Kaye
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences (D.M.K.).,Department of Cardiology, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Waled A Shihata
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Hamdi A Jama
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia.,Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (H.A.J., K.T., F.Z.M.)
| | - Kirill Tsyganov
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (H.A.J., K.T., F.Z.M.).,Monash Bioinformatics Platform (K.T.)
| | - Mark Ziemann
- Epigenetics in Human Health and Disease (M.Z., A.E-O.).,School of Life and Environmental Sciences, Deakin University, Geelong, Australia (M.Z.)
| | - Helen Kiriazis
- Mouse Cardiology Research Platform (H.K., D.D., X-J.D.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Duncan Horlock
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Amrita Vijay
- Department for Twin Research and Genetic Epidemiology, King's College London, UK (A.Vijay, T.D.S., A.M.V.)
| | - Beverly Giam
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, and Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia (A.Vinh, G.D.)
| | | | - April Fiedler
- Centre for Cardiovascular Biology and Disease Research, and Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia (A.Vinh, G.D.)
| | - Daniel Donner
- Mouse Cardiology Research Platform (H.K., D.D., X-J.D.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School (M.S., M.T.C.)
| | | | | | - Xiao-Jun Du
- Mouse Cardiology Research Platform (H.K., D.D., X-J.D.), Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease (M.Z., A.E-O.).,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, New Territories (A.E-O.)
| | - Grant Drummond
- Centre for Cardiovascular Biology and Disease Research, and Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia (A.Vinh, G.D.)
| | - Gavin W Lambert
- Iverson Health Innovation Research Institute and School of Health Sciences, Swinburne University of Technology, Melbourne, Australia (G.W.L.)
| | - Tim D Spector
- Department for Twin Research and Genetic Epidemiology, King's College London, UK (A.Vijay, T.D.S., A.M.V.)
| | - Ana M Valdes
- Department for Twin Research and Genetic Epidemiology, King's College London, UK (A.Vijay, T.D.S., A.M.V.).,School of Medicine, University of Nottingham, UK; NIHR Nottingham Biomedical Research Centre, UK (A.M.V.)
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute (C.R.M.).,Department of Biochemistry and Molecular Biology (C.R.M.), Monash University, Melbourne, Australia
| | - Francine Z Marques
- Heart Failure Research Group (D.M.K., W.A.S., H.A.J., D.H., B.G., A.F., F.Z.M.), Baker Heart and Diabetes Institute, Melbourne, Australia.,Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science (H.A.J., K.T., F.Z.M.)
| |
Collapse
|
19
|
Wilson MA, Liberzon I, Lindsey ML, Lokshina Y, Risbrough VB, Sah R, Wood SK, Williamson JB, Spinale FG. Common pathways and communication between the brain and heart: connecting post-traumatic stress disorder and heart failure. Stress 2019; 22:530-547. [PMID: 31161843 PMCID: PMC6690762 DOI: 10.1080/10253890.2019.1621283] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Psychiatric illnesses and cardiovascular disease (CVD) contribute to significant overall morbidity, mortality, and health care costs, and are predicted to reach epidemic proportions with the aging population. Within the Veterans Administration (VA) health care system, psychiatric illnesses such as post-traumatic stress disorder (PTSD) and CVD such as heart failure (HF), are leading causes of hospital admissions, prolonged hospital stays, and resource utilization. Numerous studies have demonstrated associations between PTSD symptoms and CVD endpoints, particularly in the Veteran population. Not only does PTSD increase the risk of HF, but this relationship is bi-directional. Accordingly, a VA-sponsored conference entitled "Cardiovascular Comorbidities in PTSD: The Brain-Heart Consortium" was convened to explore potential relationships and common biological pathways between PTSD and HF. The conference was framed around the hypothesis that specific common systems are dysregulated in both PTSD and HF, resulting in a synergistic acceleration and amplification of both disease processes. The conference was not intended to identify all independent pathways that give rise to PTSD and HF, but rather identify shared systems, pathways, and biological mediators that would be modifiable in both disease processes. The results from this conference identified specific endocrine, autonomic, immune, structural, genetic, and physiological changes that may contribute to shared PTSD-CVD pathophysiology and could represent unique opportunities to develop therapies for both PTSD and HF. Some recommendations from the group for future research opportunities are provided.
Collapse
Affiliation(s)
- Marlene A. Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System, Columbia SC
- Corresponding author information: Marlene A. Wilson, Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia SC 29208, Research Service, Columbia VA Health Care System, Columbia SC 29209, ; 803-216-3507
| | - Israel Liberzon
- Department of Psychiatry, Texas A&M College of Medicine, Bryan, TX
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, and Research Service, Omaha VA Medical Center, Omaha NE
| | - Yana Lokshina
- Department of Psychiatry, Texas A&M College of Medicine, Bryan, TX
| | - Victoria B. Risbrough
- VA Center of Excellence for Stress and Mental Health, La Jolla CA, Dept. of Psychiatry, University of California San Diego
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Susan K. Wood
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System, Columbia SC
| | - John B. Williamson
- Department of Neurology, University of Florida College of Medicine, Gainesville FL
| | - Francis G. Spinale
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine and Research Service, Columbia VA Health Care System., Columbia SC
| |
Collapse
|
20
|
Rizvi S, Raza ST, Rahman Q, Eba A, Zaidi ZH, Mahdi F. Association of endothelial nitric oxide synthase (eNOS) and norepinephrine transporter (NET) genes polymorphism with type 2 diabetes mellitus. Mol Biol Rep 2019; 46:5433-5441. [PMID: 31377977 DOI: 10.1007/s11033-019-04998-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/23/2019] [Indexed: 11/25/2022]
Abstract
Genetic factors in combination with environmental factors play a critical role in the development type 2 diabetes mellitus (T2DM) which is growing as an epidemic globally. In present study we aim to assess the association of eNOS (G894T, rs1799983) and NET (G1287A, rs5569) genes polymorphism with T2DM. A case-control study including a total of 400 North Indian subjects (200 T2DM cases and 200 controls) was performed using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) approach to analyze genetic polymorphism. Alleles/genotype frequencies between cases and controls were compared using χ2 and Student's t-tests. Odds ratios and 95% confidence intervals were calculated by logistic regression to assess the relative association between disease and genotypes. In case of NET gene, GG (P = 0.002 in T2DM males, 0.053 in overall T2DM cases) genotype and G allele (P = 0.003 in T2DM males, 0.027 in overall T2DM cases) were found to be a positive risk factors and AG genotype (P = 0.012 in T2DM males) and A allele (P = 0.003 in T2DM males, P = 0.027 in overall T2DM cases) as negative risk factor for T2DM. No association of eNOS gene polymorphism was found with T2DM (P values of all genotypes and alleles were greater than 0.05). NET gene polymorphism might be associated with the risk of T2DM whereas; eNOS gene polymorphism do not confer any risk of T2DM in North Indian Ethnic group. It is hoped that understanding genetic causes of T2DM will lead to earlier diagnosis, preventive measures and more effective and specific treatment.
Collapse
Affiliation(s)
- Saliha Rizvi
- Molecular Biology Lab, Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, 226003, India
| | - Syed Tasleem Raza
- Molecular Biology Lab, Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, 226003, India.
| | - Qamar Rahman
- Amity University, Lucknow Campus, Lucknow, India
| | - Ale Eba
- Molecular Biology Lab, Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, 226003, India
| | - Zeashan H Zaidi
- Department of Statistics, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Farzana Mahdi
- Molecular Biology Lab, Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, Uttar Pradesh, 226003, India
| |
Collapse
|
21
|
Gruzdev SK, Yakovlev AA, Druzhkova TA, Guekht AB, Gulyaeva NV. The Missing Link: How Exosomes and miRNAs can Help in Bridging Psychiatry and Molecular Biology in the Context of Depression, Bipolar Disorder and Schizophrenia. Cell Mol Neurobiol 2019; 39:729-750. [PMID: 31089834 DOI: 10.1007/s10571-019-00684-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/03/2019] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) only recently have been recognized as promising molecules for both fundamental and clinical neuroscience. We provide a literature review of miRNA biomarker studies in three most prominent psychiatric disorders (depression, bipolar disorder and schizophrenia) with the particular focus on depression due to its social and healthcare importance. Our search resulted in 191 unique miRNAs across 35 human studies measuring miRNA levels in blood, serum or plasma. 30 miRNAs replicated in more than one study. Most miRNAs targeted neuroplasticity and neurodevelopment pathways. Various limitations do not allow us to make firm conclusions on clinical potential of studied miRNAs. Based on our results we discuss the rationale for future research investigations of exosomal mechanisms to overcome methodological caveats both in studying etiology and pathogenesis, and providing an objective back-up for clinical decisions.
Collapse
Affiliation(s)
- S K Gruzdev
- Institute of Medicine, RUDN University, Miklukho-Maklaya Str. 6, Moscow, Russia, 117198.
| | - A A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5A, Moscow, Russia, 117485
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| | - T A Druzhkova
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| | - A B Guekht
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
- Russian National Research Medical University, Ostrovitianov Str. 1, Moscow, Russia, 117997
| | - N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova Str., 5A, Moscow, Russia, 117485
- Moscow Research & Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Donskaya Str., 43, Moscow, Russia, 115419
| |
Collapse
|
22
|
Lambert GW, Schlaich MP, Eikelis N, Lambert EA. Sympathetic activity in obesity: a brief review of methods and supportive data. Ann N Y Acad Sci 2019; 1454:56-67. [PMID: 31268175 DOI: 10.1111/nyas.14140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/11/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
The increase in the prevalence of obesity and the concomitant rise in obesity-related illness have led to substantial pressure on health care systems throughout the world. While the combination of reduced exercise, increased sedentary time, poor diet, and genetic predisposition is undoubtedly pivotal in generating obesity and increasing disease risk, a large body of work indicates that the sympathetic nervous system (SNS) contributes to obesity-related disease development and progression. In obesity, sympathetic nervous activity is regionalized, with activity in some outflows being particularly sensitive to the obese state, whereas other outflows, or responses to stimuli, may be blunted, thereby making the assessment of sympathetic nervous activation in the clinical setting difficult. Isotope dilution methods and direct nerve recording techniques have been developed and utilized in clinical research, demonstrating that in obesity there is preferential activation of the muscle vasoconstrictor and renal sympathetic outflows. With weight loss, sympathetic activity is reduced. Importantly, sympathetic nervous activity is associated with end-organ dysfunction and changes in sympathetic activation that accompany weight loss are often reflected in an improvement of end-organ function. Whether targeting the SNS directly improves obesity-related illness remains unknown, but merits further attention.
Collapse
Affiliation(s)
- Gavin W Lambert
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, University of Western Australia, Perth, Western Australia, Australia
| | - Nina Eikelis
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Elisabeth A Lambert
- The Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, Victoria, Australia.,The School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
23
|
Guilherme JPLF, Bigliassi M, Lancha Junior AH. Association study of SLC6A2 gene Thr99Ile variant (rs1805065) with athletic status in the Brazilian population. Gene 2019; 707:53-57. [PMID: 31075414 DOI: 10.1016/j.gene.2019.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/24/2019] [Accepted: 05/06/2019] [Indexed: 10/26/2022]
Abstract
Genetic variants in monoamine neurotransmitter genes have been recurrently associated with panic disorder, addiction and mood disorders. Recent evidence also indicates that norepinephrine neurotransmission can influence a series of psychophysical and psychobiological parameters related to athletic performance, and the presence of variants in the SLC6A2 (solute carrier family 6 member 2) gene, which encodes the norepinephrine transporter, can be detrimental to an adequate noradrenergic signaling. Accordingly, the objective of the present study was to explore the SLC6A2 Thr99Ile variant (rs1805065) in a cohort composed of highly-trained individuals and non-trained individuals. A total of 1556 Brazilians: 926 non-athletes and 630 athletes (322 endurance athletes and 308 power athletes) were compared in this case-control association study. The Thr99Ile variant showed only two genotypes (C/C or C/T), and a low minor allele frequency of ≈1%. However, none of the power athletes had the mutant T-allele (i.e., the C/T genotype), which may be related to decreased norepinephrine transporter activity. The genotype distribution and allele frequency observed in power athletes were significantly different when compared to non-athletes or endurance athletes. Therefore, the presence of the T-allele may decrease the chance of belonging to the group of athletes involved in explosive physical tasks. These results still need to be replicated in independent cohorts. However, it appears reasonable to assume that there is an association between the SLC6A2 gene variant and power athletic status.
Collapse
Affiliation(s)
- João Paulo L F Guilherme
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil; Endurance Performance Research Group, School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil.
| | - Marcelo Bigliassi
- Endurance Performance Research Group, School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Antonio H Lancha Junior
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
24
|
Meng L, Bai X, Zheng Y, Chen D, Zheng Y. Altered expression of norepinephrine transporter participate in hypertension and depression through regulated TNF-α and IL-6. Clin Exp Hypertens 2019; 42:181-189. [PMID: 30957546 DOI: 10.1080/10641963.2019.1601205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aim: We explored the role of histone modification in the association of depression-hypertension by comparing norepinephrine transporter (NET) gene levels in different depression-hypertensive patients. Then, we analyzed the expression of NET correlation with inflammatory cytokines to provide a new direction for detecting the association mechanism between depression and hypertension.Methods: NE expression levels in serum of diverse groups were detected by enzyme-linked immunosorbent assay. Then histone acetyltransferase (HAT), histone deacetylase (HDAC), H3K27ac, NET, TNF-α, and interleukin-6 (IL-6) were detected by western blot in nine female subjects in different depression and hypertension groups, and Chromatin immunoprecipitation-polymerase chain reaction (Chip-PCR) were used to confirm the degree of acetylation affecting on the transcription level of NET gene. Meanwhile, correlation between NET with TNF/IL-6 was analyzed by SPSS19.0 software program. Finally, Quantitative real-time polymerase chain reaction (qPCR) and western blot were used to detect TNF-α and IL-6 expression levels after NET overexpression or interference treatment in human umbilical vein endothelial cells and Neuro-2a cells.Results: The expression of HAT and H3K27ac had lower levels in D-H and nonD-H group than nonD-nonH group. The results showed that higher acetylation could promote expression of NET genes. Meanwhile, the expression of NET had a significant negative correlation with IL-6 (R = -0.933, p < 0.01) and tumor necrosis factor (TNF) (R = -0.817, p < 0.01) in subjects. In addition, the results confirmed that TNF-α and IL-6 mRNA and protein partial expressions could be inhibited by NET in both HUVECs and Neuronal cells (p < 0.01).Conclusion: In conclusion, differential expression of NET gene might function as an important factor in interaction between depression and hypertension by partially targeting TNF-α and IL-6.
Collapse
Affiliation(s)
- Lin Meng
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Xiaoxue Bai
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Yan Zheng
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Dongmei Chen
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| | - Yang Zheng
- Department of Cardiology, the First Hospital of Jilin University, Changchun, Jilin Province, P. R. China
| |
Collapse
|
25
|
Arnold AC, Ng J, Raj SR. Postural tachycardia syndrome - Diagnosis, physiology, and prognosis. Auton Neurosci 2018; 215:3-11. [PMID: 29523389 PMCID: PMC6113123 DOI: 10.1016/j.autneu.2018.02.005] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/22/2018] [Accepted: 02/25/2018] [Indexed: 11/29/2022]
Abstract
Postural tachycardia syndrome (POTS) is a heterogeneous clinical syndrome that has gained increasing interest over the past few decades due to its increasing prevalence and clinical impact on health-related quality of life. POTS is clinically characterized by sustained excessive tachycardia upon standing that occurs in the absence of significant orthostatic hypotension and other medical conditions and or medications, and with chronic symptoms of orthostatic intolerance. POTS represents one of the most common presentations of syncope and presyncope secondary to autonomic dysfunction in emergency rooms and in cardiology, neurology, and primary care clinics. The most sensitive method to detect POTS is a detailed medical history, physical examination with orthostatic vital signs or brief tilt table test, and a resting 12-lead electrocardiogram. Additional diagnostic testing may be warranted in selected patients based on clinical signs. While the precise etiology remains unknown, the orthostatic tachycardia in POTS is thought to reflect convergence of multiple pathophysiological processes, as a final common pathway. Based on this, POTS is often described as a clinical syndrome consisting of multiple heterogeneous disorders, with several underlying pathophysiological processes proposed in the literature including partial sympathetic neuropathy, hyperadrenergic state, hypovolemia, mast cell activation, deconditioning, and immune-mediated. These clinical features often overlap, however, making it difficult to categorize individual patients. Importantly, POTS is not associated with mortality, with many patients improving to some degree over time after diagnosis and proper treatment. This review will outline the current understanding of diagnosis, pathophysiology, and prognosis in POTS.
Collapse
Affiliation(s)
- Amy C Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Mail Code H109, Hershey, PA, USA; Autonomic Dysfunction Center, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jessica Ng
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Satish R Raj
- Autonomic Dysfunction Center, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
26
|
A functional genetic variation of SLC6A2 repressor hsa-miR-579-3p upregulates sympathetic noradrenergic processes of fear and anxiety. Transl Psychiatry 2018; 8:226. [PMID: 30341278 PMCID: PMC6195525 DOI: 10.1038/s41398-018-0278-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 01/05/2023] Open
Abstract
Increased sympathetic noradrenergic signaling is crucially involved in fear and anxiety as defensive states. MicroRNAs regulate dynamic gene expression during synaptic plasticity and genetic variation of microRNAs modulating noradrenaline transporter gene (SLC6A2) expression may thus lead to altered central and peripheral processing of fear and anxiety. In silico prediction of microRNA regulation of SLC6A2 was confirmed by luciferase reporter assays and identified hsa-miR-579-3p as a regulating microRNA. The minor (T)-allele of rs2910931 (MAFcases = 0.431, MAFcontrols = 0.368) upstream of MIR579 was associated with panic disorder in patients (pallelic = 0.004, ncases = 506, ncontrols = 506) and with higher trait anxiety in healthy individuals (pASI = 0.029, pACQ = 0.047, n = 3112). Compared to the major (A)-allele, increased promoter activity was observed in luciferase reporter assays in vitro suggesting more effective MIR579 expression and SLC6A2 repression in vivo (p = 0.041). Healthy individuals carrying at least one (T)-allele showed a brain activation pattern suggesting increased defensive responding and sympathetic noradrenergic activation in midbrain and limbic areas during the extinction of conditioned fear. Panic disorder patients carrying two (T)-alleles showed elevated heart rates in an anxiety-provoking behavioral avoidance test (F(2, 270) = 5.47, p = 0.005). Fine-tuning of noradrenaline homeostasis by a MIR579 genetic variation modulated central and peripheral sympathetic noradrenergic activation during fear processing and anxiety. This study opens new perspectives on the role of microRNAs in the etiopathogenesis of anxiety disorders, particularly their cardiovascular symptoms and comorbidities.
Collapse
|
27
|
A polymorphism in the noradrenaline transporter gene is associated with increased blood pressure in patients with resistant hypertension. J Hypertens 2018; 36:1571-1577. [DOI: 10.1097/hjh.0000000000001736] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Angyal N, Horvath EZ, Tarnok Z, Richman MJ, Bognar E, Lakatos K, Sasvari-Szekely M, Nemoda Z. Association analysis of norepinephrine transporter polymorphisms and methylphenidate response in ADHD patients. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:122-128. [PMID: 29374517 DOI: 10.1016/j.pnpbp.2018.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/27/2017] [Accepted: 01/17/2018] [Indexed: 10/18/2022]
Abstract
AIMS Methylphenidate (MPH) is the most frequently prescribed drug in Attention Deficit Hyperactivity Disorder (ADHD). Hitherto mostly the dopamine transporter gene has been studied in MPH-response and only a few studies analyzed the norepinephrine transporter (NET, SLC6A2) gene, although MPH is a potent inhibitor of both dopamine and norepinephrine transporters. We aimed to analyze this monoamine transporter gene in relation to ADHD per se and MPH-response in particular to gain further knowledge in ADHD pharmacogenetics using a Caucasian sample. METHODS Six single nucleotide polymorphisms (rs28386840, rs2242446, rs3785143, rs3785157, rs5569, rs7194256 SNP) were studied across the NET gene in 163 ADHD children (age: 9.3±2.6; 86.5% male) using ADHD-RS hyperactivity-impulsivity and inattention scales. For case-control analysis 486 control subjects were also genotyped. At the MPH-response analysis responders had minimum 25% decrease of ADHD-RS total score after 2months of treatment, and chi-square test compared 90 responders and 32 non-responders, whereas ANOVA was used to assess symptom improvement after the first month among the 122 ADHD patients. RESULTS The classical case-control analysis did not yield any association with ADHD diagnosis, which was supported by meta-analysis conducted on the available genetic data (combining previously published and the present studies). On the other hand, the intronic rs3785143 showed nominal association with inattention symptoms (p=0.01). The haplotype analysis supported this association, and indicated the importance of the first haploblock encompassing the intronic and 2 promoter SNPs. With MPH-response only the promoter rs28386840 showed nominal association: Those with at least one T-allele were overrepresented in the responder group (42% vs 19%, p=0.08), and they had better improvement on the hyperactivity-impulsivity scale compared to the AA genotype (p=0.04). CONCLUSION Although none of our single SNP findings remained significant after correcting for multiple testing, our results from the MPH-response analysis indicate the potential importance of promoter variants in the NET gene.
Collapse
Affiliation(s)
- Nora Angyal
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Erzsebet Zsofia Horvath
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Zsanett Tarnok
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Mara J Richman
- Department of Clinical Psychology and Addiction, Eötvös Loránd University, Budapest, Hungary
| | - Emese Bognar
- Vadaskert Child and Adolescent Psychiatric Hospital, Budapest, Hungary
| | - Krisztina Lakatos
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Maria Sasvari-Szekely
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Zsofia Nemoda
- Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
29
|
Yuan H, Mischoulon D, Fava M, Otto MW. Circulating microRNAs as biomarkers for depression: Many candidates, few finalists. J Affect Disord 2018; 233:68-78. [PMID: 28673667 DOI: 10.1016/j.jad.2017.06.058] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/14/2017] [Accepted: 06/26/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Recent research has highlighted the potential of microRNAs to serve as physiological indicators of disease process among clinically depressed patients. METHODS In a comprehensive literature search through PubMed, we identified 23 articles comparing circulating (blood, plasma, or serum) microRNA expression levels in depressed versus healthy human subjects. Six studies examining circulatory microRNA expression through animal models of depression were also identified through the search and details of each study were outlined. A meta-analytic evaluation of these studies was not considered feasible, given the absence of concordance in the literature to date. RESULTS A total of 178 specific microRNA candidates were identified in the human studies as significantly expressed among depressed samples. Ninety-seven of these microRNAs were upregulated, 75 were downregulated, and 6 showed mixed expression in depressed samples. Few microRNAs were consistently expressed across studies; the most consistent evidence was for microRNA-132, with replication in 4 different studies. Among animal studies, 2 studies investigated microRNA-16 through distinct stress-induced depression models. LIMITATIONS Structural variations in microRNA sequences, methodological inconsistencies in technology used among studies to measure microRNA expression levels, differences in the clinical severity and forms of depression among subjects, and the overall paucity of studies make it difficult to ascertain any robust, preliminary targets deserving of biomarker potential. CONCLUSIONS Ongoing research needs to address this high rate of non-replication as well as the methodological and reporting challenges of microRNA experimentation in order to determine valid effect sizes for the more proliferative candidates associated with depression.
Collapse
Affiliation(s)
- Heidi Yuan
- Department of Psychological and Brain Sciences, Boston University, United States; Pomona College, Claremont, CA, United States
| | - David Mischoulon
- Depression Clinical and Research Program, Massachusetts General Hospital and Harvard Medical School, United States
| | - Maurizio Fava
- Depression Clinical and Research Program, Massachusetts General Hospital and Harvard Medical School, United States
| | - Michael W Otto
- Department of Psychological and Brain Sciences, Boston University, United States.
| |
Collapse
|
30
|
Modulation of sympathetic preganglionic neuron activity via adrenergic receptors. Hypertens Res 2018; 41:499-505. [PMID: 29748580 DOI: 10.1038/s41440-018-0049-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 11/08/2022]
Abstract
The sympathetic preganglionic neurons (SPNs) play a key role in the sympathetic nervous system. Previous reports have suggested that norepinephrine (NE) directly affects SPNs via both inhibitory hyperpolarization interactions mediated by α2 receptors and excitatory depolarization interactions mediated by α1 receptors. It remains poorly understood, however, whether the excitability of SPNs can be inhibited indirectly (presynaptically) as well as directly (postsynaptically). We intracellularly recorded 41 SPNs using the whole-cell patch-clamp technique in spinal cord slice preparations of neonatal rats. We examined the effects of NE or dexmedetomidine hydrochloride (Dxm) (α2-adrenergic receptor agonist) on SPNs by analyzing the excitatory postsynaptic potentials (EPSPs) and inhibitory postsynaptic potentials (IPSPs). EPSPs were dominant in 15 SPNs (EPSP-SPNs) and IPSPs were dominant in 7 SPNs (IPSP-SPNs) at baseline. We were unable to analyze the postsynaptic potentials in the other 19 SPNs, due to high frequency of action potential firings (firing-SPNs). At baseline, the membrane potentials and resistances of each type of SPN were similar. NE (1 μM) gradually depolarized the EPSP-SPNs and IPSP-SPNs (P < 0.001) and NE significantly increased the EPSP frequency of the EPSP-SPNs (P < 0.05). Dxm (10 nM) after application of NE decreased the EPSP frequency of the EPSP-SPNs (P < 0.001) and the EPSP voltage and IPSP voltage of the IPSP-SPNs (P < 0.05). In 5 of the 19 firing-SPNs, NE induced membrane hyperpolarization (P < 0.05) and completely inhibited firings. Dxm had no effect in these neurons. The SPNs received inhibitory modulation through α2-adrenergic receptors. Some SPNs can be directly inhibited via effects independent of the α2 receptors.
Collapse
|
31
|
Weger M, Sandi C. High anxiety trait: A vulnerable phenotype for stress-induced depression. Neurosci Biobehav Rev 2018; 87:27-37. [DOI: 10.1016/j.neubiorev.2018.01.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/14/2018] [Accepted: 01/21/2018] [Indexed: 11/25/2022]
|
32
|
Esler M, Lambert G, Schlaich M, Dixon J, Sari CI, Lambert E. Obesity Paradox in Hypertension: Is This Because Sympathetic Activation in Obesity-Hypertension Takes a Benign Form? Hypertension 2017; 71:22-33. [PMID: 29158358 DOI: 10.1161/hypertensionaha.117.09790] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Murray Esler
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.).
| | - Gavin Lambert
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.)
| | - Markus Schlaich
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.)
| | - John Dixon
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.)
| | - Carolina Ika Sari
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.)
| | - Elisabeth Lambert
- From the Baker Heart and Diabetes Institute, Melbourne, Australia (M.E., J.D., C.I.S.); Swinburne University of Technology, Melbourne, Australia (G.L., E.L.); and School of Medicine, Royal Perth Hospital Unit, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, Perth (M.S.)
| |
Collapse
|
33
|
Meuret AE, Kroll J, Ritz T. Panic Disorder Comorbidity with Medical Conditions and Treatment Implications. Annu Rev Clin Psychol 2017; 13:209-240. [PMID: 28375724 DOI: 10.1146/annurev-clinpsy-021815-093044] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Panic disorder (PD) is unique among the anxiety disorders in that panic symptoms are primarily of a physical nature. Consequently, comorbidity with medical illness is significant. This review examines the association between PD and medical illness. We identify shared pathophysiological and psychological correlates and illustrate how physiological activation in panic sufferers underlies their symptom experience in the context of the fight-or-flight response and beyond a situation-specific response pattern. We then review evidence for bodily symptom perception accuracy in PD. Prevalence of comorbidity for PD and medical illness is presented, with a focus on respiratory and cardiovascular illness, irritable bowel syndrome, and diabetes, followed by an outline for potential pathways of a bidirectional association. We conclude by illustrating commonalities in mediating mechanistic pathways and moderating risk factors across medical illnesses, and we discuss implications for diagnosis and treatment of both types of conditions.
Collapse
Affiliation(s)
- Alicia E Meuret
- Department of Psychology, Southern Methodist University, Dallas, Texas 75275;
| | - Juliet Kroll
- Department of Psychology, Southern Methodist University, Dallas, Texas 75275;
| | - Thomas Ritz
- Department of Psychology, Southern Methodist University, Dallas, Texas 75275;
| |
Collapse
|