Translate

Τρίτη 10 Σεπτεμβρίου 2019

BaiJiu increases nitric oxide bioactivity of Chinese herbs used to treat CAD via the NO3--NO2--NO pathway
BaiJiu (BJ) is a type of Chinese rice wine combined with the traditional Chinese herbs GuaLou (GL) and XieBai (XB), which have been used to treat and prevent coronary artery disease (CAD) for nearly 2,000 years in China. However, the mechanisms behind the compatibility of the components of this compound (GLXBBJ) have not been deeply investigated. In this study, the compatibility of the GLXBBJ compounds with nitric oxide (NO) bioactivity was evaluated in herbs, cells, and isolated aortic rings. Nitrate (NO3-) and nitrite (NO2-) concentrations were quantified by the Griess method. Nitric oxide (NO) was quantified by a multifunctional enzyme marker using a fluorescent probe. Qualitative analysis of L-arginine-endothelial NO synthase (eNOS) was performed by western blotting. The tension of aortic rings was measured by multi myograph system. The ability of BJ to reduce NO3- to NO2- and NO2- to NO was strongest under hypoxic conditions and was not affected by temperature. BJ-containing serum significantly decreased the NO3- content and increased the NO2- content in hypoxic cells. Combining BJ with GL, XB, or GLXB resulted in stronger vasodilation effects. These results demonstrate that BJ effectively reduces NO3-/NO2-, although only a small amount of NO3- is present. Once combined with GL, XB, or GLXB, which are rich in NO3-/NO2-, robust NO bioactivity was generated through the NO3--NO2--NO pathway. Therefore, this study supports the potential of using traditional Chinese herbs for promoting medical innovation and for future drug development. Corresponding author: (Yaoping Tang) E-mail: typqc@126.com; Tel: +86 18007802816; Fax: +86 7713946492 (Congxin Huang) E-mail: huangcongxin@vip.163.com; Tel: +86 13907131546 Conflict of interest: The authors declared that they have no conflicts of interest related to this work. Sources of support: This work was supported by the National Natural Science Foundation of China [grant # 81774115, 2018]; National Natural Science Foundation of China [grant # 81460721, 2015]; National Natural Science Foundation of China [grant # 30960487, 2010]; National Natural Science Foundation of China [grant # 31460243, 2015]; and Guangxi Autonomous Region Scientific Research and Technology Development and Chairman Technology Fund Project of China [No.1517-10].We thank Mr. Zhixiang Long and Mr. Xiaoguang Shi for their technical support of nitrate and nitrite measurements. We also appreciate Dr. Laijun Liu for the assistance in manuscript preparation. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Multiplicity of Nitric Oxide and Natriuretic Peptide Signalling in Heart Failure
Heart failure (HF) is a common consequence of several cardiovascular diseases, and is understood as a vicious cycle of cardiac and haemodynamic decline. The current inventory of treatments either alleviate the pathophysiological features (e.g., cardiac dysfunction, neurohumoral activation, ventricular remodelling) and/or target any underlying pathologies (e.g., hypertension, myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine 3’,5’-monophosphate (cGMP), synthesised by nitric oxide (NO)- and natriuretic peptide (NP)- responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signalling, which can occur following GC deactivation and the upregulation of cyclic nucleotide-hydrolysing phosphodiesterases (PDEs), promotes cardiac dysfunction. Herein we review the role that NO/cGMP and NP/cGMP signalling plays in HF. After considering disease aetiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from pre-clinical models and patients that compromised cGMP signalling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined. To whom correspondence should be addressed. Email: m.e.j.preedy@qmul.ac.uk. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Cardiovascular Pharmacology of the NLRP3 Inflammasome
No abstract available
cGMP Signaling and Modulation in Heart Failure
Cyclic GMP (cGMP) represents a classic intracellular second messenger molecule. Over the past two decades, important discoveries have identified that cGMP signaling becomes deranged in heart failure, and that cGMP and its main kinase effector, Protein Kinase G, generally oppose the biological abnormalities contributing to heart failure, in experimental studies. These findings have influenced the design of clinical trials of cGMP-augmenting drugs in heart failure patients. At present, the trial results of cGMP-augmenting therapies in heart failure remain mixed. As detailed in this review, strong evidence now exists that Protein Kinase G opposes pathologic cardiac remodeling through regulation of diverse biological processes and myocardial substrates. Potential reasons for the failures of cGMP-augmenting drugs in HF may be related to biological mechanisms opposing cGMP, or due to certain features of clinical trials, all of which are discussed. Correspondence: Robert M. Blanton, MD. Molecular Cardiology Research Institute, Tufts Medical Center. 800 Washington Street, #080. Boston, MA, USA 02111. Telephone +1 617 636 7678 Fax +1 617 636 1444 Email: rblanton@tuftsmedicalcenter.org Sources of Support: NIH R01HL131831 Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Research progress of mechanisms and drug therapy for atherosclerosis on TLR pathway
Recent reports have established atherosclerosis (AS) as a major factor in the pathogenetic process of cardiovascular diseases like ischemic stroke and coronary heart disease. Although the possible pathogenesis of AS remains to be elucidated, a large number of investigations strongly suggest that the inhibition of Toll-like receptors (TLRs) alleviates the severity of AS to some extent by suppressing vascular inflammation and the formation of atherosclerotic plaques. As pattern recognition receptors (PRRs), TLRs occupy a vital position in innate immunity, mediating various signaling pathways in infective and sterile inflammation. This review summarizes the available data on the research progress of AS and the latest antiatherosclerotic drugs associated with TLR pathway. Corresponding author. E-mail address: yunmanlicpu@hotmail.com (Y.-m. Li) E-mail address: huyahui324@163.com Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
FSTL3 induces lipid accumulation and inflammatory response in macrophages and associates with atherosclerosis
FSTL3 as adipokine take part in dyslipidemia and inflammatory response, but the association of FSTL3 with atherosclerosis is unclear. This study indicated that FSTL3 showed significantly higher level (Control: 7.68±3.10 ng/mL versus AS: 9.29±2.37 ng/mL; P<0.001) in atherosclerosis, and FSTL3 expressed higher in plaque of ApoE knockout mice and located in macrophages. OxLDL induced expression and secretion of FSTL3, meanwhile, FSTL3 promoted lipid accumulation in macrophages. The advanced study found that FSTL3 upregulated CD36 and LOX-1 expression with dose-dependent manner, however, FSTL3 also evoked interleukin 1-β (IL1-β), monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor-α (TNFα) and matrix metalloproteinase-9 (MMP-9) secretion in macrophages. On the contrary that downregulated FSTL3 attenuated expression of oxLDL induced CD36, LOX-1 and inflammatory cytokines expressing. All of these results demonstrated that FSTL3 as novelty cytokine take part in process of atherosclerosis through increasing lipid accumulation and inflammation via regulating CD36 and LOX-1 expression. Corresponding author: Ding Wenjun, MD, PhD Department of Cardiac Surgery, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital 180 Feng Lin road Shanghai 200025 People’s Republic of China Tel: +86 21 64041990 Fax: +86 21 64041990 Email address: doctor_dingwj@163.com Co-corresponding author: Wang Chunsheng, MD, PhD Department of Cardiac Surgery, Shanghai Institute of Cardiovascular Diseases Zhongshan Hospital 180 Feng Lin road Shanghai 200025 People’s Republic of China Tel: +86 21 64041990 Fax: +86 21 64041990 Email address: doctor_wangcs@163.com This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Long non-coding RNA NEAT1 promotes myocardiocyte apoptosis and suppresses proliferation via regulation of miR-129-5p
Recent studies have revealed the important role of long non-coding RNAs (lncRNAs) in heart development and pathogenesis. This study was aimed to investigate the role of NEAT1 in hypoxia-induced cardiac injury and explore its possible molecular mechanism. Real-time PCR (RT-PCR) was used to determine the relative RNA expression of NEAT1 and its potential target microRNA (miRNA), miR-129-5p, in the plasma of patients with acute myocardial infarction, heart failure, and angina, as well as in H2O2-treated H9c2 cells. The role of NEAT1 overexpression or inhibition in H9c2 cell migration and proliferation was assessed by transwell assay and Edu staining, respectively. Collagen deposition and apoptosis were evaluated by western blot detection of collagen and apoptotic proteins, including Capase3, Bax, and Bcl2. We showed that H2O2 treatment significantly decreased H9c2 cell migration and proliferation while increasing H9c2 cell apoptosis. Inhibition of NEAT1 attenuated the cell apoptosis and alleviated proliferation inhibition induced by hypoxia. Bioinformatics analysis showed that miR-129-5p was the direct target of NEAT1, which was confirmed by luciferase assay. NEAT1 upregulation aggravated apoptosis by downregulating miR-129-5p. In conclusion, we uncovered a novel NEAT1-miR129 axis and its implication in H2O2-induced heart failure. Correspondence author: Ling Qin, Department of Cardiology, The First Hospital of Jilin University, No.71 of Xinmin Street, Changchun City, Jilin Province, 130021, PR. China. Tel: 86 0431-84808227 Email: tx75452@163.com Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
Dietary apigenin reduces induction of LOX-1 and NLRP3 expression, leukocyte adhesion and acetylated low-density lipoprotein uptake in human endothelial cells exposed to trimethylamine-N-oxide
By inducing vascular inflammation, trimethylamine-N-oxide (TMAO) is associated with endothelial dysfunction, atherosclerosis, and enhanced risk of cardiovascular diseases in humans. However, the underlying mechanisms are unknown. Expression of several genes related to arteriosclerosis, inflammasomes, and endothelial dysfunction was quantified by PCR following exposure to TMAO. LOX-1, ICAM-1, and NLRP3 were also quantified by western blot, while leukocytic adhesion was examined using fluorescently labeled U937 cells. Scavenger receptors (SRs), adhesion molecules, and other genes associated with atherosclerosis were induced in endothelial cells exposed to TMAO. On the other hand, apigenin, a flavonoid that is abundant in parsley and celery, prevents initial arteriosclerosis events in endothelial cells. Apigenin reversed the effects of TMAO on mRNA expression of LOX-1, SREC, SR-PSOX, NLRP3, ASC, TXNIP, VCAM-1, ICAM-1, and MCP-1, as well as protein expression of LOX-1, the adhesion molecule ICAM-1, and the inflammasome protein NLRP3. Apigenin also suppressed leukocyte adhesion and uptake of acetylated low-density lipoprotein. The data indicate that expression of SRs and adhesion molecules in response to TMAO, along with formation of NLRP3 inflammasomes, may drive endothelial dysfunction via uptake of acetylated LDL and lymphocyte adhesion. Apigenin reverses these effects, implying that it may also prevent arteriosclerosis. Corresponding author: Prof. Kazuo Yamagata Ph.D. Laboratory of Molecular Health Science of Food, Department of Food Bioscience and Biotechnology, College of Bioresource Sciences (NUBS), 1866, Kameino, Fujisawa, Kanagawa 252-8510, Japan. Phone: +81-466-84-3986, Fax: +81-466-84-3986 E-mail: yamagata.kazuo@nihon-u.ac.jp Competing interests; The authors report no conflicts of interest. Kazuo Yamagata; yamagata.kazuo@nihon-u.ac.jp, Kazuki Hashiguchi;brka14091@g.nihon-u.ac.jp, Hiroaki Yamamoto;mt.hiro-7714@ezeb.ne.jp, Motoki Tagami; mtagami@sanraku.or.jp, Funding; Not Availability of data and materials; The data used and/or analyzed during the current study are available from the corresponding author on reasonable request. Author’s contributions; K.Y. and M.T conceived and designed the project; K.Y, H.K, S.K and H.Y. performed experiments; K.S and K.H. performed statistical analysis of the data with inputs from S.K. Consent for publication; Not applicable. Ethics approval and consent to participate; Not applicable. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
In vitro anticoagulant activity of Mikania laevigata: deepening the study of the possible interaction between guaco and anticoagulants
Mikania laevigata, popularly known in Brazil as guaco, is widely used for respiratory disorders. As this plant is rich in coumarins, there is evidence of indications that it may cause bleeding and therefore should not be used concomitantly with anticoagulants. The basis of this information is very theoretical, with no clinical evidence of such contraindication. Thus, the aim of this study was to evaluate the in vitro effect of M. laevigata extract on blood coagulation through prothrombin (PT) and activated partial thromboplastin (aPTT) tests, fibrinogen plasma concentration (FPC), and the new thrombin generation test (TGT), which investigate, with high sensibility, hemostatic changes (CAAE 60904316.6.0000.5149), besides evaluating its qualitative micromolecular composition, providing scientific evidence to support the management of patients taking warfarin. Ethanolic extracts of guaco leaves were incubated with a plasma pool of healthy individuals at concentrations of 1.67 mg/ml, 2.26 mg/ml, and 2.86 mg/ml. The presence of flavonoids, tannins, coumarins, and triterpenes was demonstrated by selective reagents in thin layer chromatography. Benzoylgrandifloric acid, cinnamoylgrandifloric acid, o-coumaric acid, coumarin, and quercetin-3-β-glucoside were identified by co-injection in ultra-performance liquid chromatography. The extract at all concentrations prolonged TP and aPPT, and reduced the potential for endogenous thrombin potential (ETP) by TGT. The control plasma had ETP=1465 nM/min and after the addition of M. laevigata extract (2.26 mg/mL), this value was reduced to 1087 nM/min, indicating a lower generation of thrombin. Related to FPC, concentrations of 2.26 mg/mL and 2.86 mg/mL were effective in reducing plasma fibrinogen levels. These results allow us to conclude that the guaco extract demonstrated an anticoagulant effect in vitro, possibly interfering with intrinsic, extrinsic, and common coagulation pathways. A discussion on the contribution of the identified substances to the activity is also present. Corresponding author: Paula Mendonça Leite. Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, Minas Gerais, Brazil. Tel.: +55 31 3409 6936; Fax: +55 31 3409 6935 E-mail address: paulamleite02@gmail.com (P. M. Leite) ORCIT ID 0000-0002-8499-5791 Disclosures: The authors state no conflict of interest. Funding and Acknowledgements: This work was supported by grants from Pró-Reitoria de Pesquisa from Universidade Federal de Minas Gerais, Fundação de Amparo a Pesquisa do Estado de Minas Gerais (FAPEMIG), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Brazil. Ethical Approval: All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee (Ethics Committee of the Federal University of Minas Gerais number CAAE 60904316.6.0000.5149) and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.
The role of YAP/TAZ in energy metabolism in the heart
The heart requires a high amount of energy, in the form of adenosine triphosphate (ATP), to maintain its viability and pump function. Anaerobic glycolysis and mitochondrial oxidative phosphorylation are the main metabolic pathways by which ATP is generated, utilizing fatty acids (FAs), glucose, lactate, and ketone bodies as primary substrates. Previous studies have demonstrated that, in response to stress, the heart undergoes alterations in metabolism, ranging from changes in substrate utilization to mitochondrial function, collectively called metabolic remodeling. However, the molecular mechanism mediating metabolic remodeling in the heart remains unclear. Yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which are major downstream effectors of the Hippo signaling pathway, play an important role in the regulation of heart size and cellular homeostasis of cardiomyocytes through the regulation of various transcriptional factors under both physiological and pathophysiological conditions. Recent findings in various organs and cell types have revealed that YAP and TAZ play an important role in energy metabolism. Here we summarize what is currently known about YAP/TAZ in the regulation of metabolism of various substrates and mitochondrial function in various organs and cell types and discuss the potential role of YAP/TAZ in mediating metabolic remodeling of the heart during stress and heart failure. Address correspondence to: Junichi Sadoshima, MD, PhD Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 S. Orange Ave., MSB G609, Newark, NJ 07103, USA Phone: +1-973-972-8916 E-mail: sadoshju@njms.rutgers.edu Sources of funding: This work was supported in part by U.S. Public Health Service Grants HL67724, HL91469, HL112330, HL138720, and AG23039 (J.S.), and by the Foundation of Leducq Transatlantic Network of Excellence 15CBD04 (J.S.). T.K. has been supported by a Postdoctoral Fellowship from the Uehara Memorial Foundation. Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.

Δεν υπάρχουν σχόλια:

Δημοσίευση σχολίου

Αρχειοθήκη ιστολογίου

Translate