1
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
2
|
Hu W, Liu Y, Lian C, Lu H. Genetic insight into putative causes of xanthelasma palpebrarum: a Mendelian randomization study. Front Immunol 2024; 15:1347112. [PMID: 38601164 PMCID: PMC11004296 DOI: 10.3389/fimmu.2024.1347112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Xanthelasma palpebrarum (XP) is the most common form of cutaneous xanthoma, with a prevalence of 1.1%~4.4% in the population. However, the cause of XP remains largely unknown. In the present study, we used Mendelian randomization to assess the genetic association between plasma lipids, metabolic traits, and circulating protein with XP, leveraging summary statistics from large genome-wide association studies (GWAS). Genetically predicted plasma cholesterol and LDL-C, but not HDL-C or triglyceride, were significantly associated with XP. Metabolic traits, including BMI, fasting glucose, type 2 diabetes, systolic and diastolic blood pressure, were not significantly associated with XP. Furthermore, we found genetically predicted 12 circulating proteins were associated with XP, including FN1, NTM, FCN2, GOLM1, ICAM5, PDE5A, C5, CLEC11A, CXCL1, CCL2, CCL11, CCL13. In conclusion, this study identified plasma cholesterol, LDL-C, and 12 circulating proteins to be putative causal factors for XP, highlighting the role of plasma cholesterol and inflammatory response in XP development.
Collapse
Affiliation(s)
- Wenting Hu
- Department of Dermatology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Yaozhong Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, Ann Arbor, MI, United States
| | - Cuihong Lian
- Department of Dermatology, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Haocheng Lu
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Pușcașu C, Zanfirescu A, Negreș S, Șeremet OC. Exploring the Multifaceted Potential of Sildenafil in Medicine. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2190. [PMID: 38138293 PMCID: PMC10744870 DOI: 10.3390/medicina59122190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Phosphodiesterase type 5 (PDE5) is pivotal in cellular signalling, regulating cyclic guanosine monophosphate (cGMP) levels crucial for smooth muscle relaxation and vasodilation. By targeting cGMP for degradation, PDE5 inhibits sustained vasodilation. PDE5 operates in diverse anatomical regions, with its upregulation linked to various pathologies, including cancer and neurodegenerative diseases. Sildenafil, a selective PDE5 inhibitor, is prescribed for erectile dysfunction and pulmonary arterial hypertension. However, considering the extensive roles of PDE5, sildenafil might be useful in other pathologies. This review aims to comprehensively explore sildenafil's therapeutic potential across medicine, addressing a gap in the current literature. Recognising sildenafil's broader potential may unveil new treatment avenues, optimising existing approaches and broadening its clinical application.
Collapse
Affiliation(s)
| | - Anca Zanfirescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (S.N.); (O.C.Ș.)
| | | | | |
Collapse
|
4
|
Bork NI, Subramanian H, Kurelic R, Nikolaev VO, Rybalkin SD. Role of Phosphodiesterase 1 in the Regulation of Real-Time cGMP Levels and Contractility in Adult Mouse Cardiomyocytes. Cells 2023; 12:2759. [PMID: 38067187 PMCID: PMC10706287 DOI: 10.3390/cells12232759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/19/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
In mouse cardiomyocytes, the expression of two subfamilies of the calcium/calmodulin-regulated cyclic nucleotide phosphodiesterase 1 (PDE1)-PDE1A and PDE1C-has been reported. PDE1C was found to be the major subfamily in the human heart. It is a dual substrate PDE and can hydrolyze both 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP). Previously, it has been reported that the PDE1 inhibitor ITI-214 shows positive inotropic effects in heart failure patients which were largely attributed to the cAMP-dependent protein kinase (PKA) signaling. However, the role of PDE1 in the regulation of cardiac cGMP has not been directly addressed. Here, we studied the effect of PDE1 inhibition on cGMP levels in adult mouse ventricular cardiomyocytes using a highly sensitive fluorescent biosensor based on Förster resonance energy transfer (FRET). Live-cell imaging in paced and resting cardiomyocytes showed an increase in cGMP after PDE1 inhibition with ITI-214. Furthermore, PDE1 inhibition and PDE1A knockdown amplified the cGMP-FRET responses to the nitric oxide (NO)-donor sodium nitroprusside (SNP) but not to the C-type natriuretic peptide (CNP), indicating a specific role of PDE1 in the regulation of the NO-sensitive guanylyl cyclase (NO-GC)-regulated cGMP microdomain. ITI-214, in combination with CNP or SNP, showed a positive lusitropic effect, improving the relaxation of isolated myocytes. Immunoblot analysis revealed increased phospholamban (PLN) phosphorylation at Ser-16 in cells treated with a combination of SNP and PDE1 inhibitor but not with SNP alone. Our findings reveal a previously unreported role of PDE1 in the regulation of the NO-GC/cGMP microdomain and mouse ventricular myocyte contractility. Since PDE1 serves as a cGMP degrading PDE in cardiomyocytes and has the highest hydrolytic activities, it can be expected that PDE1 inhibition might be beneficial in combination with cGMP-elevating drugs for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Nadja I. Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.I.B.); (H.S.); (R.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Hariharan Subramanian
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.I.B.); (H.S.); (R.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Roberta Kurelic
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.I.B.); (H.S.); (R.K.)
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.I.B.); (H.S.); (R.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Sergei D. Rybalkin
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.I.B.); (H.S.); (R.K.)
| |
Collapse
|
5
|
Vishwakarma VK, Shah S, Kaur T, Singh AP, Arava SK, Kumar N, Yadav RK, Yadav S, Arora T, Yadav HN. Effect of vinpocetine alone and in combination with enalapril in experimental model of diabetic cardiomyopathy in rats: possible involvement of PDE-1/TGF-β/ Smad 2/3 signalling pathways. J Pharm Pharmacol 2023; 75:1198-1211. [PMID: 37229596 DOI: 10.1093/jpp/rgad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DC) is one of the severe secondary complications of diabetes mellitus in humans. Vinpocetine is an alkaloid having pleiotropic pharmacological effects. The present study is designed to investigate the effect of vinpocetine in DC in rats. METHODS Rats were fed a high-fat diet for nine weeks along with single dose of streptozotocin after the second week to induce DC. The haemodynamic evaluation was performed to assess the functional status of rats using the Biopac system. Cardiac echocardiography, biochemical, oxidative stress parameters and inflammatory cytokine level were analysed in addition to haematoxylin-eosin and Masson's trichome staining to study histological changes, cardiomyocyte diameter and fibrosis, respectively. Phosphodiesterase-1 (PDE-1), transforming growth factor-β (TGF-β) and p-Smad 2/3 expression in cardiac tissues were quantified using western blot/RT-PCR. KEY FINDING Vinpocetine treatment and its combination with enalapril decreased the glucose levels compared to diabetic rats. Vinpocetine improved the echocardiographic parameters and cardiac functional status of rats. Vinpocetine decreased the cardiac biochemical parameters, oxidative stress, inflammatory cytokine levels, cardiomyocyte diameter and fibrosis in rats. Interestingly, expressions of PDE-1, TGF-β and p-Smad 2/3 were ameliorated by vinpocetine alone and in combination with enalapril. CONCLUSIONS Vinpocetine is a well-known inhibitor of PDE-1 and the protective effect of vinpocetine in DC is exerted by inhibition of PDE-1 and subsequent inhibition of the expression of TGF-β/Smad 2/3.
Collapse
Affiliation(s)
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Tajpreet Kaur
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sudheer Kumar Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Niraj Kumar
- Department of Neuroanesthesiogy and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Raj Kanwar Yadav
- Department Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sushma Yadav
- Department of Obstetrics and Gynaecology, SHKM Government Medical College, Nuh, Haryana, India
| | - Taruna Arora
- RBMCH, ICMR-Head Quarter's Ansari Nagar, New Delhi, India
| | | |
Collapse
|
6
|
Roy S, Kloner RA, Salloum FN, Jovin IS. Cardiac Effects of Phosphodiesterase-5 Inhibitors: Efficacy and Safety. Cardiovasc Drugs Ther 2023; 37:793-806. [PMID: 34652581 PMCID: PMC9010479 DOI: 10.1007/s10557-021-07275-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 01/23/2023]
Abstract
The coexistence of cardiovascular disease and erectile dysfunction is widespread, possibly owing to underlying endothelial dysfunction in both diseases. Millions of patients with cardiovascular disease are prescribed phosphodiesterase-5 (PDE5) inhibitors for the management of erectile dysfunction. Although the role of PDE5 inhibitors in erectile dysfunction therapy is well established, their effects on the cardiovascular system are unclear. Preclinical studies investigating the effect of PDE5 inhibitors on ischemia-reperfusion injury, pressure overload-induced hypertrophy, and chemotoxicity suggested a possible clinical role for each of these medications; however, attempts to translate these findings to the bedside have resulted in mixed outcomes. In this review, we explore the biologic preclinical effects of PDE5 inhibitors in mediating cardioprotection. We then examine clinical trials investigating PDE5 inhibition in patients with heart failure, coronary artery disease, and ventricular arrhythmias and discuss why the studies likely have yet to show positive results and efficacy with PDE5 inhibition despite no safety concerns.
Collapse
Affiliation(s)
- Sumon Roy
- Pauley Heart Center, Virginia Commonwealth University Medical Center, McGuire VAMC, 1201 Broad Rock Boulevard, 111J, Richmond, VA, 23249, USA
| | - Robert A Kloner
- Huntington Medical Research Institute, Pasadena, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fadi N Salloum
- Pauley Heart Center, Virginia Commonwealth University Medical Center, McGuire VAMC, 1201 Broad Rock Boulevard, 111J, Richmond, VA, 23249, USA
| | - Ion S Jovin
- Pauley Heart Center, Virginia Commonwealth University Medical Center, McGuire VAMC, 1201 Broad Rock Boulevard, 111J, Richmond, VA, 23249, USA.
- McGuire Veterans Affairs Medical Center, Richmond, VA, USA.
| |
Collapse
|
7
|
Metwally E, Mak V, Soriano A, Zebisch M, Silvestre HL, McEwan PA, Ermakov G, Beaumont M, Tawa P, Barker JJ, Yen R, Patel A, Lim YH, Healy D, Hanisak J, Cheng AC, Greshock T, Fischmann TO. Structural insights into selective small molecule activation of PKG1α. Commun Biol 2023; 6:798. [PMID: 37524852 PMCID: PMC10390508 DOI: 10.1038/s42003-023-05095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
cGMP-dependent protein kinase I-α (PKG1α) is a target for pulmonary arterial hypertension due to its role in the regulation of smooth muscle function. While most work has focused on regulation of cGMP turnover, we recently described several small molecule tool compounds which were capable of activating PKG1α via a cGMP independent pathway. Selected molecules were crystallized in the presence of PKG1α and were found to bind to an allosteric site proximal to the low-affinity nucleotide binding domain. These molecules act to displace the switch helix and cause activation of PKG1α representing a new mechanism for the activation and control of this critical therapeutic path. The described structures are vital to understanding the function and control of this key regulatory pathway.
Collapse
Affiliation(s)
- Essam Metwally
- Modeling and Informatics, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA.
| | - Victor Mak
- Discovery Chemistry, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Aileen Soriano
- Quantitative Biosciences, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Matthias Zebisch
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RZ, UK
| | - H Leonardo Silvestre
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RZ, UK
| | - Paul A McEwan
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RZ, UK
| | - Grigori Ermakov
- Discovery Bioanalytics, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Maribel Beaumont
- Discovery Bioanalytics, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Paul Tawa
- Quantitative Biosciences, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - John J Barker
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RZ, UK
| | - Rose Yen
- Discovery Chemistry, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Akash Patel
- Discovery Chemistry, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Yeon-Hee Lim
- Discovery Chemistry, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - David Healy
- Discovery Biology, MRL, Merck & Co., Inc., Boston, MA, USA
| | - Jennifer Hanisak
- Discovery Chemistry, MRL, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Alan C Cheng
- Modeling and Informatics, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Tom Greshock
- Discovery Chemistry, MRL, Merck & Co., Inc., 213 E. Grand Avenue, South San Francisco, CA, USA
| | - Thierry O Fischmann
- Protein and Structural Chemistry, MRL, Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
8
|
Chang P, Zhang X, Zhang J, Wang J, Wang X, Li M, Wang R, Yu J, Fu F. BNP protects against diabetic cardiomyopathy by promoting Opa1-mediated mitochondrial fusion via activating the PKG-STAT3 pathway. Redox Biol 2023; 62:102702. [PMID: 37116257 PMCID: PMC10165144 DOI: 10.1016/j.redox.2023.102702] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/25/2023] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
Brain natriuretic peptide (BNP) belongs to the family of natriuretic peptides, which are responsible for a wide range of actions. Diabetic cardiomyopathy (DCM) is often associated with increased BNP levels. This present research intends to explore the role of BNP in the development of DCM and the underlying mechanisms. Diabetes was induced in mice using streptozotocin (STZ). Primary neonatal cardiomyocytes were treated with high glucose. It was found that the levels of plasma BNP started to increase at 8 weeks after diabetes, which preceded the development of DCM. Addition of exogenous BNP promoted Opa1-mediated mitochondrial fusion, inhibited mitochondrial oxidative stress, preserved mitochondrial respiratory capacity and prevented the development of DCM, while knockdown of endogenous BNP exacerbated mitochondrial dysfunction and accelerated DCM. Opa1 knockdown attenuated the aforementioned protective action of BNP both in vivo and in vitro. BNP-induced mitochondrial fusion requires the activation of STAT3, which facilitated Opa1 transcription by binding to its promoter regions. PKG, a crucial signaling biomolecule in the BNP signaling pathway, interacted with STAT3 and induced its activation. Knockdown of NPRA (the receptor of BNP) or PKG blunted the promoting effect of BNP on STAT3 phosphorylation and Opa1-mediated mitochondrial fusion. The results of this study demonstrate for the first time that there is a rise in BNP during the early stages of DCM as a compensatory protection mechanism. BNP is a novel mitochondrial fusion activator in protecting against hyperglycemia-induced mitochondrial oxidative injury and DCM through the activation of NPRA-PKG-STAT3-Opa1 signaling pathway.
Collapse
Affiliation(s)
- Pan Chang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China; Clinical Experimental Center, The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710100, China
| | - Xiaomeng Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Jing Zhang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Jianbang Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Xihui Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Man Li
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China; Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, Xi'an, 710032, China
| | - Rui Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Jun Yu
- Clinical Experimental Center, The Affiliated Xi'an International Medical Center Hospital, Northwest University, Xi'an, 710100, China.
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, Xi'an, 710032, China; Department of Cardiology, Tangdu Hospital, Airforce Medical University, Xi'an, 710038, China.
| |
Collapse
|
9
|
Huang S, Chen J, Song M, Yu Y, Geng J, Lin D, Yang J, Wu J, Li K, Yu Y, Wang J, Hu L, Shan Q, Wang J, Chen P, Chen F. Whole-exome sequencing and electrophysiological study reveal a novel loss-of-function mutation of KCNA10 in epinephrine provoked long QT syndrome with familial history of sudden cardiac death. Leg Med (Tokyo) 2023; 62:102245. [PMID: 36965351 DOI: 10.1016/j.legalmed.2023.102245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023]
Abstract
Congenital long QT syndrome (LQTS) is one type of inherited fatal cardiac arrhythmia that may lead to sudden cardiac death (SCD). Mutations in more than 16 genes have been reported to be associated with LQTS, whereas the genetic causes of about 20% of cases remain unknown. In the present study, we investigated a four-generation pedigree with familial history of syncope and SCD. The proband was a 33-year-old young woman who experienced 3 episodes of syncope when walking at night. The electrocardiogram revealed a markedly epinephrine-provoked prolonged QT interval (QT = 468 ms, QTc = 651 ms) but no obvious arrhythmia in the resting state. Three family members have died of suspected SCD. Whole-exome sequencing and bioinformatic analysis based on pedigree revealed that a novel missense mutation KCNA10 (c.1397G>A/Arg466Gln) was the potential genetic lesion. Sanger sequencing was performed to confirm the whole-exome sequencing results. This mutation resulted in the KV1.8 channel amino acid residue 466 changing from arginine to glutamine, and the electrophysiological experiments verified it as a loss-of-function mutation of KV1.8, which reduced the K+ currents of KV1.8 and might result in the prolonged QT interval. These findings suggested that KCNA10 (c.1397G>A) mutation was possibly pathogenic in this enrolled LQTS family, and may provide a new potential genetic target for diagnosis and counseling of stress-related LQTS families as well as the postmortem diagnosis of SCD.
Collapse
Affiliation(s)
- Shuainan Huang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Ji Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Miaomiao Song
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Youjia Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jie Geng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Donghai Lin
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jiawen Yang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jiayi Wu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Kai Li
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yanfang Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Jie Wang
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Li Hu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Qijun Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| | - Peng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China.
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
10
|
Swiecicka A. The efficacy of PDE5 inhibitors in diabetic patients. Andrology 2023; 11:245-256. [PMID: 36367281 PMCID: PMC10107754 DOI: 10.1111/andr.13328] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/27/2022] [Accepted: 10/14/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Phosphodiesterase 5 inhibitors (PDE5i), since their introduction in the late 1990s, have proven their efficacy in treating several conditions, predominantly pulmonary hypertension and erectile dysfunction where they remain the first-line therapeutic option. However, in the recent years, growing evidence from both animal and human studies has emerged to suggest the additional benefits of PDE5i in cardiovascular and metabolic disorders. This is of specific interest to the diabetes population where prevalent cardiovascular disease and metabolic dysregulation significantly contribute to the increased morbidity and mortality. OBJECTIVES To examine the available data on the non-standard, pleiotropic effects of PDE5i in patients with diabetes mellitus. MATERIALS AND METHODS The review of the published background research, preclinical studies and clinical trials. RESULTS In human studies, PDE5 inhibition appeared to be associated with reduced cardiovascular mortality and overall improved clinical outcomes in those with established cardiovascular disease. PDE5i were also consistently found to reduce albuminuria in subjects with diabetic nephropathy. Furthermore, animal data suggest a plausible effect of this group of medication on sensory function and neuropathic symptoms in diabetic neuropathy as well as improved wound healing. A decrease in insulin resistance and augmentation of beta cell function seen in preclinical studies has not been consistently demonstrated in human trials. DISCUSSION AND CONCLUSION In animal models, PDE5 inhibition appears to decrease oxidative stress and reduce some of the micro- and macrovascular complications associated with diabetes. However, data from human trials are limited and largely inconsistent, highlighting the need for adequately powered, randomised-controlled trials in diabetic cohorts in order to fully assess the benefits of PDE5i in this group of patients.
Collapse
Affiliation(s)
- Agnieszka Swiecicka
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| |
Collapse
|
11
|
Samidurai A, Xi L, Das A, Kukreja RC. Beyond Erectile Dysfunction: cGMP-Specific Phosphodiesterase 5 Inhibitors for Other Clinical Disorders. Annu Rev Pharmacol Toxicol 2023; 63:585-615. [PMID: 36206989 DOI: 10.1146/annurev-pharmtox-040122-034745] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cyclic guanosine monophosphate (cGMP), an important intracellular second messenger, mediates cellular functional responses in all vital organs. Phosphodiesterase 5 (PDE5) is one of the 11 members of the cyclic nucleotide phosphodiesterase (PDE) family that specifically targets cGMP generated by nitric oxide-driven activation of the soluble guanylyl cyclase. PDE5 inhibitors, including sildenafil and tadalafil, are widely used for the treatment of erectile dysfunction, pulmonary arterial hypertension, and certain urological disorders. Preclinical studies have shown promising effects of PDE5 inhibitors in the treatment of myocardial infarction, cardiac hypertrophy, heart failure, cancer and anticancer-drug-associated cardiotoxicity, diabetes, Duchenne muscular dystrophy, Alzheimer's disease, and other aging-related conditions. Many clinical trials with PDE5 inhibitors have focused on the potential cardiovascular, anticancer, and neurological benefits. In this review, we provide an overview of the current state of knowledge on PDE5 inhibitors and their potential therapeutic indications for various clinical disorders beyond erectile dysfunction.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA;
| |
Collapse
|
12
|
Jing G, Xia Z, Lei Q. Co-expression of soluble guanylyl cyclase subunits and PDE5A shRNA to elevate cellular cGMP level: A potential gene therapy for myocardial cell death. Technol Health Care 2022; 31:901-910. [PMID: 36442224 DOI: 10.3233/thc-220333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND: Genetic manipulation on the NO-sGC-cGMP pathway has been rarely achieved, partially due to complexity of the soluble guanylyl cyclase (sGC) enzyme. OBJECTIVE: We aim to develop gene therapy directly targeting the pathway to circumvent cytotoxicity and tolerance after prolonged use of NO-donors and the insufficiency of PDE inhibitors. METHODS: In this study, we constructed lentivirus vectors expressing GUCY1A3 and GUCY1B3 genes, which encoded the α1 and β1 subunits of soluble guanylyl cyclase (sGC), respectively, to enhance cGMP synthesis. We also constructed lentiviral vector harboring PDE5A shRNA to alleviate phosphodiesterase activity and cGMP degradation. RESULTS: Transductions of human HEK293 cells with the constructs were successful, as indicated by the fluorescent signal and altered gene expression produced by each vector. Overexpression of GUCY1A3 and GUCY1B3 resulted in increased sGC enzyme activity and elevated cGMP level in the cells. Expression of PDE5A shRNA resulted in decreased PDE5A expression and elevated cGMP level. Co-transduction of the three lentiviral vectors resulted in a more significant elevation of cGMP in HEK293 cells without obvious cytotoxicity. CONCLUSION: To the best of our knowledge, this is the first study to show that co-expression of exogenous subunits of the soluble guanylyl cyclase could form functional enzyme and increase cellular cGMP level in mammalian cells. Simultaneous expression of PDE5A shRNA could alleviate feedback up-regulation on PDE5A caused by cGMP elevation. Further studies are required to evaluate the effects of these constructs in vivo.
Collapse
Affiliation(s)
- Gao Jing
- Tianjin Key Laboratory of Exercise Physiology and Sport Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, China
- Family Medicine Clinic, Tianjin United Family Healthcare, Tianjin, China
| | - Zhang Xia
- Tianjin Key Laboratory of Exercise Physiology and Sport Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Quan Lei
- Tianjin Key Laboratory of Exercise Physiology and Sport Medicine, Institute of Exercise and Health, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
13
|
Saikia Q, Hazarika A, Mishra R. A Review on the Pharmacological Importance of PDE5 and Its Inhibition to Manage Biomedical Conditions. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x221129008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Phosphodiesterase type 5 (PDE5) is a cyclic GMP (cGMP) specific protein. It hydrolyzes the phosphodiesterase linkage and catalyzes the conversion of cGMP to 5’ GMP, which controls different physiological activities of the body. PDE5 is associated with biomedical conditions like neurological disorders, pulmonary arterial hypertension, cardiomyopathy, cancer, erectile dysfunction, and lower urinary tract syndrome. Inhibition of PDE5 has now been proven pharmaceutically effective in a variety of therapeutic conditions. Avanafil, tadalafil, sildenafil, and vardenafil are the most commonly used PDE5 inhibitors (PDE5i) today which are often used for the management of erectile dysfunction, lower urinary tract syndromes, malignancy, and pulmonary arterial hypertension. However, these synthetic PDE5i come with a slew of negative effects. Some of the most common side effects include mild headaches, flushing, dyspepsia, altered color vision, back discomfort, priapism, melanoma, hypotension and dizziness, non-arteritic anterior ischemic optic neuropathy (NAION), and hearing loss. In light of the potential negative effects of this class of medications, there is a lot of room for new, selective PDE5 inhibitors to be discovered. We have found 25 plant botanical compounds effectively inhibiting PDE5 which might be useful in treating a variety of disorders with minimal or no adverse effects.
Collapse
Affiliation(s)
- Queen Saikia
- Department of Zoology, Gauhati University, Guwahati, Assam, India
| | - Ajit Hazarika
- Tyagbir Hem Baruah College, Jamugurihat, Sonitpur, Assam, India
| | - Ritu Mishra
- Department of Zoology, Gauhati University, Guwahati, Assam, India
| |
Collapse
|
14
|
Chen JY, Xie ZX, Dai JZ, Han JY, Wang K, Lu LH, Jin JJ, Xue SJ. Reconstruction and analysis of potential biomarkers for hypertrophic cardiomyopathy based on a competing endogenous RNA network. BMC Cardiovasc Disord 2022; 22:422. [PMID: 36138345 PMCID: PMC9503253 DOI: 10.1186/s12872-022-02862-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/15/2022] [Indexed: 11/26/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common heritable cardiomyopath. Although considerable effort has been made to understand the pathogenesis of HCM, the mechanism of how long noncoding RNA (lncRNA)-associated competing endogenous RNA (ceRNA) network result in HCM remains unknown. In this study, we acquired a total of 520 different expression profiles of lncRNAs (DElncRNAs) and 371 messenger RNAs (mRNA, DEGs) by microarray and 33 microRNAs (DEmiRNAs) by sequencing in plasma of patients with HCM and healthy controls. Then lncRNA–miRNA pairs were predicted using miRcode and starBase and crossed with DEmiRNAs. MiRNA–mRNA pairs were retrieved from miRanda and TargetScan and crossed with DEGs. Combined with these pairs, the ceRNA network with eight lncRNAs, three miRNAs, and 22 mRNAs was constructed. lncRNA RP11-66N24.4 and LINC00310 were among the top 10% nodes. The hub nodes were analyzed to reconstruct a subnetwork. Furthermore, quantitative real-time polymerase chain reaction results showed that LINC00310 was significantly decreased in patients with HCM. For LINC00310, GO analysis revealed that biological processes were enriched in cardiovascular system development, sprouting angiogenesis, circulatory system development, and pathway analysis in the cGMP-PKG signaling pathway. These results indicate that the novel lncRNA-related ceRNA network in HCM and LINC00310 may play a role in the mechanism of HCM pathogenesis, which could provide insight into the pathogenesis of HCM.
Collapse
Affiliation(s)
- Jin-Yan Chen
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China. .,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China.
| | - Zhang-Xin Xie
- Department of Emergency, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jia-Zhen Dai
- Department of Cardiology, Zhangzhou Affilated Hospital, Zhangzhou, China
| | - Jun-Yong Han
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Kun Wang
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Li-Hong Lu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
| | - Jing-Jun Jin
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| | - Shi-Jie Xue
- Institute for Immunology, Fujian Academy of Medical Sciences, No. 7 Wusi Road, Fuzhou, 350001, China.,Fujian Provincial Key Laboratory of Medical Analysis, Fuzhou, 350001, China
| |
Collapse
|
15
|
Protein tyrosine phosphatase PTPN22 negatively modulates platelet function and thrombus formation. Blood 2022; 140:1038-1051. [PMID: 35767715 DOI: 10.1182/blood.2022015554] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is a protein tyrosine phosphatase that negatively regulates T-cell signaling. However, whether it is expressed and functions in platelets remains unknown. Here we investigated the expression and role of PTPN22 in platelet function. We reported PTPN22 expression in both human and mouse platelets. Using PTPN22-/- mice, we showed that PTPN22 deficiency significantly shortened tail-bleeding time and accelerated arterial thrombus formation without affecting venous thrombosis and the coagulation factors VIII and IX. Consistently, PTPN22-deficient platelets exhibited enhanced platelet aggregation, granule secretion, calcium mobilization, lamellipodia formation, spreading, and clot retraction. Quantitative phosphoproteomic analysis revealed the significant difference of phosphodiesterase 5A (PDE5A) phosphorylation in PTPN22-deficient platelets compared with wild-type platelets after collagen-related peptide stimulation, which was confirmed by increased PDE5A phosphorylation (Ser92) in collagen-related peptide-treated PTPN22-deficient platelets, concomitant with reduced level and vasodilator-stimulated phosphoprotein phosphorylation (Ser157/239). In addition, PTPN22 interacted with phosphorylated PDE5A (Ser92) and dephosphorylated it in activated platelets. Moreover, purified PTPN22 but not the mutant form (C227S) possesses intrinsic serine phosphatase activity. Furthermore, inhibition of PTPN22 enhanced human platelet aggregation, spreading, clot retraction, and increased PDE5A phosphorylation (Ser92). In conclusion, our study shows a novel role of PTPN22 in platelet function and arterial thrombosis, identifying new potential targets for future prevention of thrombotic or cardiovascular diseases.
Collapse
|
16
|
Ovchinnikov A, Potekhina A, Belyavskiy E, Ageev F. Heart Failure with Preserved Ejection Fraction and Pulmonary Hypertension: Focus on Phosphodiesterase Inhibitors. Pharmaceuticals (Basel) 2022; 15:ph15081024. [PMID: 36015172 PMCID: PMC9414416 DOI: 10.3390/ph15081024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary hypertension (PH) is common in patients with heart failure with preserved ejection fraction (HFpEF). A chronic increase in mean left atrial pressure leads to passive remodeling in pulmonary veins and capillaries and modest PH (isolated postcapillary PH, Ipc-PH) and is not associated with significant right ventricular dysfunction. In approximately 20% of patients with HFpEF, "precapillary" alterations of pulmonary vasculature occur with the development of the combined pre- and post-capillary PH (Cpc-PH), pertaining to a poor prognosis. Current data indicate that pulmonary vasculopathy may be at least partially reversible and thus serves as a therapeutic target in HFpEF. Pulmonary vascular targeted therapies, including phosphodiesterase (PDE) inhibitors, may have a valuable role in the management of patients with PH-HFpEF. In studies of Cpc-PH and HFpEF, PDE type 5 inhibitors were effective in long-term follow-up, decreasing pulmonary artery pressure and improving RV contractility, whereas studies of Ipc-PH did not show any benefit. Randomized trials are essential to elucidate the actual value of PDE inhibition in selected patients with PH-HFpEF, especially in those with invasively confirmed Cpc-PH who are most likely to benefit from such treatment.
Collapse
Affiliation(s)
- Artem Ovchinnikov
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
- Department of Clinical Functional Diagnostics, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, 127473 Moscow, Russia
- Correspondence: ; Tel.: +7-(495)-414-66-12 or +7-(916)-505-79-58; Fax: +7-(495)-414-66-12
| | - Alexandra Potekhina
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
| | - Evgeny Belyavskiy
- Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité Universitätsmedizin Berlin, Augustenburger Platz, 13353 Berlin, Germany
| | - Fail Ageev
- Out-Patient Department, Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, 3-d Cherepkovskaya St., 15a, 121552 Moscow, Russia
| |
Collapse
|
17
|
Mak VW, Patel AM, Yen R, Hanisak J, Lim YH, Bao J, Zheng R, Seganish WM, Yu Y, Healy DR, Ogawa A, Ren Z, Soriano A, Ermakov GP, Beaumont M, Metwally E, Cheng AC, Verras A, Fischmann T, Zebisch M, Silvestre HL, McEwan PA, Barker J, Rearden P, Greshock TJ. Optimization and Mechanistic Investigations of Novel Allosteric Activators of PKG1α. J Med Chem 2022; 65:10318-10340. [PMID: 35878399 DOI: 10.1021/acs.jmedchem.1c02109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Activation of PKG1α is a compelling strategy for the treatment of cardiovascular diseases. As the main effector of cyclic guanosine monophosphate (cGMP), activation of PKG1α induces smooth muscle relaxation in blood vessels, lowers pulmonary blood pressure, prevents platelet aggregation, and protects against cardiac stress. The development of activators has been mostly limited to cGMP mimetics and synthetic peptides. Described herein is the optimization of a piperidine series of small molecules to yield activators that demonstrate in vitro phosphorylation of vasodilator-stimulated phosphoprotein as well as antiproliferative effects in human pulmonary arterial smooth muscle cells. Hydrogen/deuterium exchange mass spectrometry experiments with the small molecule activators revealed a mechanism of action consistent with cGMP-induced activation, and an X-ray co-crystal structure with a construct encompassing the regulatory domains illustrated a binding mode in an allosteric pocket proximal to the low-affinity cyclic nucleotide-binding domain.
Collapse
Affiliation(s)
- Victor W Mak
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Akash M Patel
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Rose Yen
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jennifer Hanisak
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yeon-Hee Lim
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Jianming Bao
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States.,Ionova Life Science, Shenzhen 518122, Guangdong, China
| | - Rong Zheng
- IDSU, Wuxi AppTec Co., Ltd, Shanghai 200131, China
| | - W Michael Seganish
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Yang Yu
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David R Healy
- Discovery Biology, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Aimie Ogawa
- Quantitative Biosciences, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Zhao Ren
- Quantitative Biosciences, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Aileen Soriano
- Mass Spectrometry and Biophysics, Computation and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Grigori P Ermakov
- PPDM Discovery Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Maribel Beaumont
- PPDM Discovery Bioanalytics, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Essam Metwally
- Computational and Structural Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Alan C Cheng
- Computational and Structural Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Andreas Verras
- Schrodinger Inc., 120 West 45th Street, 17th Floor, New York, New York 10036-4041, United States.,Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Thierry Fischmann
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Matthias Zebisch
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - H Leonardo Silvestre
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Paul A McEwan
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - John Barker
- Evotec (UK) Ltd, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K
| | - Paul Rearden
- DMPK, Recursion Pharmaceuticals, Salt Lake City, Utah 84101, United States.,PPDM, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Thomas J Greshock
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| |
Collapse
|
18
|
Nik-Ahd F, Shindel AW. Pharmacotherapy for Erectile Dysfunction in 2021 and Beyond. Urol Clin North Am 2022; 49:209-217. [DOI: 10.1016/j.ucl.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
19
|
The sGC-cGMP Signaling Pathway as a Potential Therapeutic Target in Doxorubicin-Induced Heart Failure: A Narrative Review. Am J Cardiovasc Drugs 2022; 22:117-125. [PMID: 34151411 DOI: 10.1007/s40256-021-00487-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/05/2021] [Indexed: 01/01/2023]
Abstract
The anti-cancer agent doxorubicin (DOX) has high cardiotoxicity that is linked to DOX-mediated increase in oxidative stress, mitochondrial iron overload, DNA damage, autophagy, necrosis, and apoptosis, all of which are also associated with secondary tumorigenicity. This limits the clinical application of DOX therapies. Previous studies have attributed DOX-mediated cardiotoxicity to mitochondrial iron accumulation and the production of reactive oxygen species (ROS), which seem to be independent of its anti-tumor DNA damaging effects. Chemo-sensitization of soluble guanylate cyclase (sGC) in the cyclic guanosine monophosphate (cGMP) pathway induces tumor cell death despite the cardiotoxicity associated with DOX treatment. However, sGC-cGMP signaling must be activated during heart failure to facilitate myocardial cell survival. The sGC pathway is dependent on nitric oxide and signal transduction via the nitric oxide-sGC-cGMP pathway and is attenuated in various cardiovascular diseases. Additionally, cGMP signaling is regulated by the action of certain phosphodiesterases (PDEs) that protect the heart by inhibiting PDE, an enzyme that hydrolyses cGMP to GMP activity. In this review, we discuss the studies describing the interactions between cGMP regulation and DOX-mediated cardiotoxicity and their application in improving DOX therapeutic outcomes. The results provide novel avenues for the reduction of DOX-induced secondary tumorigenicity and improve cellular autonomy during DOX-mediated cardiotoxicity.
Collapse
|
20
|
Gu M, Zhou X, Zhu L, Gao Y, Gao L, Bai C, Yang L, Li G. Myostatin Mutation Promotes Glycolysis by Increasing Phosphorylation of Phosphofructokinase via Activation of PDE5A-cGMP-PKG in Cattle Heart. Front Cell Dev Biol 2022; 9:774185. [PMID: 35155444 PMCID: PMC8831326 DOI: 10.3389/fcell.2021.774185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/22/2021] [Indexed: 12/28/2022] Open
Abstract
Myostatin (MSTN) is a primary negative regulator of skeletal muscle mass and causes multiple metabolic changes. However, whether MSTN mutation affects heart morphology and physiology remains unclear. Myostatin mutation (MT) had no effect on cattle cardiac muscle in histological examination, but in biochemical assays, glycolysis increased in cattle hearts with MT. Compared with wild-type cattle, there were no differences in mRNA and protein levels of rate-limiting enzymes, but phosphofructokinase (PFK) phosphorylation increased in cattle hearts with MT. Transcriptome analysis showed that phosphodiesterase-5A (PDE5A), a target for inhibiting cGMP-PKG signaling, was downregulated. For the mechanism, chromatin immunoprecipitation qPCR showed that the SMAD2/SMAD3 complex in the canonical downstream pathway for MSTN combined with the promoter of PDE5A. The cGMP-PKG pathway was activated, and PKG increased phosphorylation of PFK in cattle hearts with MT. In addition, activation of PKG and the increase in PFK phosphorylation promoted glycolysis. Knockdown of PKG resulted in the opposite phenomena. The results indicated that MT potentiated PFK phosphorylation via the PDE5A-cGMP-PKG pathway and thereby promoted glycolysis in the heart.
Collapse
Affiliation(s)
- Mingjuan Gu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Xinyu Zhou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Lin Zhu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Yajie Gao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Li Gao
- Baotou Teachers’ College, Baotou, China
| | - Chunling Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
| | - Lei Yang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
- *Correspondence: Lei Yang, ; Guangpeng Li,
| | - Guangpeng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot, China
- School of Life Science, Inner Mongolia University, Hohhot, China
- *Correspondence: Lei Yang, ; Guangpeng Li,
| |
Collapse
|
21
|
Ceddia RP, Liu D, Shi F, Crowder MK, Mishra S, Kass DA, Collins S. Increased Energy Expenditure and Protection From Diet-Induced Obesity in Mice Lacking the cGMP-Specific Phosphodiesterase PDE9. Diabetes 2021; 70:2823-2836. [PMID: 34620617 PMCID: PMC8660992 DOI: 10.2337/db21-0100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022]
Abstract
Cyclic nucleotides cAMP and cGMP are important second messengers for the regulation of adaptive thermogenesis. Their levels are controlled not only by their synthesis, but also their degradation. Since pharmacological inhibitors of cGMP-specific phosphodiesterase 9 (PDE9) can increase cGMP-dependent protein kinase signaling and uncoupling protein 1 expression in adipocytes, we sought to elucidate the role of PDE9 on energy balance and glucose homeostasis in vivo. Mice with targeted disruption of the PDE9 gene, Pde9a, were fed nutrient-matched high-fat (HFD) or low-fat diets. Pde9a -/- mice were resistant to HFD-induced obesity, exhibiting a global increase in energy expenditure, while brown adipose tissue (AT) had increased respiratory capacity and elevated expression of Ucp1 and other thermogenic genes. Reduced adiposity of HFD-fed Pde9a -/- mice was associated with improvements in glucose handling and hepatic steatosis. Cold exposure or treatment with β-adrenergic receptor agonists markedly decreased Pde9a expression in brown AT and cultured brown adipocytes, while Pde9a -/- mice exhibited a greater increase in AT browning, together suggesting that the PDE9-cGMP pathway augments classical cold-induced β-adrenergic/cAMP AT browning and energy expenditure. These findings suggest PDE9 is a previously unrecognized regulator of energy metabolism and that its inhibition may be a valuable avenue to explore for combating metabolic disease.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Dianxin Liu
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Mark K Crowder
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Sumita Mishra
- Division of Cardiology, Department of Medicine, Johns Hopkins University and School of Medicine, Baltimore, MD
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University and School of Medicine, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins University and School of Medicine, Baltimore, MD
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University and School of Medicine, Baltimore, MD
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
22
|
Bork NI, Kuret A, Cruz Santos M, Molina CE, Reiter B, Reichenspurner H, Friebe A, Skryabin BV, Rozhdestvensky TS, Kuhn M, Lukowski R, Nikolaev VO. Rise of cGMP by partial phosphodiesterase-3A degradation enhances cardioprotection during hypoxia. Redox Biol 2021; 48:102179. [PMID: 34763298 PMCID: PMC8590074 DOI: 10.1016/j.redox.2021.102179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/11/2022] Open
Abstract
3',5'-cyclic guanosine monophosphate (cGMP) is a druggable second messenger regulating cell growth and survival in a plethora of cells and disease states, many of which are associated with hypoxia. For example, in myocardial infarction and heart failure (HF), clinical use of cGMP-elevating drugs improves disease outcomes. Although they protect mice from ischemia/reperfusion (I/R) injury, the exact mechanism how cardiac cGMP signaling is regulated in response to hypoxia is still largely unknown. By monitoring real-time cGMP dynamics in murine and human cardiomyocytes using in vitro and in vivo models of hypoxia/reoxygenation (H/R) and I/R injury combined with biochemical methods, we show that hypoxia causes rapid but partial degradation of cGMP-hydrolyzing phosphodiesterase-3A (PDE3A) protein via the autophagosomal-lysosomal pathway. While increasing cGMP in hypoxia prevents cell death, partially reduced PDE3A does not change the pro-apoptotic second messenger 3',5'-cyclic adenosine monophosphate (cAMP). However, it leads to significantly enhanced protective effects of clinically relevant activators of nitric oxide-sensitive guanylyl cyclase (NO-GC). Collectively, our mouse and human data unravel a new mechanism by which cardiac cGMP improves hypoxia-associated disease conditions.
Collapse
Affiliation(s)
- Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Beate Reiter
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Andreas Friebe
- Physiologisches Institut, University of Würzburg, Würzburg, Germany
| | - Boris V Skryabin
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Münster, Germany
| | - Timofey S Rozhdestvensky
- Core Facility Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Münster, Germany
| | - Michaela Kuhn
- Physiologisches Institut, University of Würzburg, Würzburg, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
23
|
Kobalava ZD, Lazarev PV. Nitric oxide — soluble guanylate cyclase — cyclic guanosine monophosphate signaling pathway in the pathogenesis of heart failure and search for novel therapeutic targets. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2021. [DOI: 10.15829/1728-8800-2021-3035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Heart failure is a severe disease with an unfavorable prognosis, which requires intensification of therapy and the search for novel approaches to treatment. In this review, the physiological significance of soluble guanylate cyclase-related signaling pathway, reasons for decrease in its activity in heart failure and possible consequences are discussed. Pharmacological methods of stimulating the production of cyclic guanosine monophosphate using drugs with different mechanisms of action are considered. Data from clinical studies regarding their effectiveness and safety are presented. A promising approach is stimulation of soluble guanylate cyclase, which showed beneficial effects in preclinical studies, as well as in the recently completed phase III VICTORIA study.
Collapse
|
24
|
Yang L, Yang Y, Liu X, Chen Y, Chen Y, Lin Y, Sun Y, Shen B. CHDGKB: a knowledgebase for systematic understanding of genetic variations associated with non-syndromic congenital heart disease. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2021; 2020:5865522. [PMID: 32608479 PMCID: PMC7327432 DOI: 10.1093/database/baaa048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023]
Abstract
Congenital heart disease (CHD) is one of the most common birth defects, with complex genetic and environmental etiologies. The reports of genetic variation associated with CHD have increased dramatically in recent years due to the revolutionary development of molecular technology. However, CHD is a heterogeneous disease, and its genetic origins remain inconclusive in most patients. Here we present a database of genetic variations for non-syndromic CHD (NS-CHD). By manually literature extraction and analyses, 5345 NS-CHD-associated genetic variations were collected, curated and stored in the public online database. The objective of our database is to provide the most comprehensive updates on NS-CHD genetic research and to aid systematic analyses of pathogenesis of NS-CHD in molecular level and the correlation between NS-CHD genotypes and phenotypes. Database URL: http://www.sysbio.org.cn/CHDGKB/.
Collapse
Affiliation(s)
- Lan Yang
- Center for Systems Biology, Soochow University, Suzhou 215006, China.,Center of Prenatal Diagnosis, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Yang Yang
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Xingyun Liu
- Center for Systems Biology, Soochow University, Suzhou 215006, China.,Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongquan Chen
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Yalan Chen
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Yuxin Lin
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Yan Sun
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bairong Shen
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Wei J, Ni X, Dai Y, Chen X, Ding S, Bao J, Xing L. Identification of genes associated with sudden cardiac death: a network- and pathway-based approach. J Thorac Dis 2021; 13:3610-3627. [PMID: 34277054 PMCID: PMC8264674 DOI: 10.21037/jtd-21-361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/14/2021] [Indexed: 12/03/2022]
Abstract
Background Sudden cardiac death (SCD) accounts for a large proportion of the total deaths across different age groups. Although numerous candidate genes related to SCD have been identified by genetic association studies and genome wide association studies (GWAS), the molecular mechanisms underlying SCD are still unclear, and the biological functions and interactions of these genes remain obscure. To clarify this issue, we performed a comprehensive and systematic analysis of SCD-related genes by a network and pathway-based approach. Methods By screening the publications deposited in the PubMed and Gene-Cloud Biotechnology Information (GCBI) databases, we collected the genes genetically associated with SCD, which were referred to as the SCD-related gene set (SCDgset). To analyze the biological processes and biochemical pathways of the SCD-related genes, functional analysis was performed. To explore interlinks and interactions of the enriched pathways, pathway crosstalk analysis was implemented. To construct SCD-specific molecular networks, Markov cluster algorithm and Steiner minimal tree algorithm were employed. Results We collected 257 genes that were reported to be associated with SCD and summarized them in the SCDgset. Most of the biological processes and biochemical pathways were related to heart diseases, while some of the biological functions may be noncardiac causes of SCD. The enriched pathways could be roughly grouped into two modules. One module was related to calcium signaling pathway and the other was related to MAPK pathway. Moreover, two different SCD-specific molecular networks were inferred, and 23 novel genes potentially associated with SCD were also identified. Conclusions In summary, by means of a network and pathway-based methodology, we explored the pathogenetic mechanism underlying SCD. Our results provide valuable information in understanding the pathogenesis of SCD and include novel biomarkers for diagnosing potential patients with heart diseases; these may help in reducing the corresponding risks and even aid in preventing SCD.
Collapse
Affiliation(s)
- Jinhuan Wei
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China
| | - Xuejun Ni
- Department of Medical Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanfei Dai
- Radiology Department, Branch of Affiliated Hospital of Nantong University, Nantong, China
| | - Xi Chen
- Department of Medical Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Sujun Ding
- Department of Medical Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Jingyin Bao
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
26
|
Lukowski R, Cruz Santos M, Kuret A, Ruth P. cGMP and mitochondrial K + channels-Compartmentalized but closely connected in cardioprotection. Br J Pharmacol 2021; 179:2344-2360. [PMID: 33991427 DOI: 10.1111/bph.15536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 01/01/2023] Open
Abstract
The 3',5'-cGMP pathway triggers cytoprotective responses and improves cardiomyocyte survival during myocardial ischaemia and reperfusion (I/R) injury. These beneficial effects were attributed to NO-sensitive GC induced cGMP production leading to activation of cGMP-dependent protein kinase I (cGKI). cGKI in turn phosphorylates many substrates, which eventually facilitate opening of mitochondrial ATP-sensitive potassium channels (mitoKATP ) and Ca2+ -activated potassium channels of the BK type (mitoBK). Accordingly, agents activating mitoKATP or mitoBK provide protection against I/R-induced damages. Here, we provide an up-to-date summary of the infarct-limiting actions exhibited by the GC/cGMP axis and discuss how mitoKATP and mitoBK, which are present at the inner mitochondrial membrane, confer mito- and cytoprotective effects on cardiomyocytes exposed to I/R injury. In view of this, we believe that the functional connection between the cGMP cascade and mitoK+ channels should be exploited further as adjunct to reperfusion therapy in myocardial infarction.
Collapse
Affiliation(s)
- Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
27
|
Feil R, Lehners M, Stehle D, Feil S. Visualising and understanding cGMP signals in the cardiovascular system. Br J Pharmacol 2021; 179:2394-2412. [PMID: 33880767 DOI: 10.1111/bph.15500] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/14/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022] Open
Abstract
cGMP is an important signalling molecule in humans. Fluorescent cGMP biosensors have emerged as powerful tools for the sensitive analysis of cGMP pathways at the single-cell level. Here, we briefly outline cGMP's multifaceted role in (patho)physiology and pharmacotherapy. Then we summarise what new insights cGMP imaging has provided into endogenous cGMP signalling and drug action, with a focus on the cardiovascular system. Indeed, the use of cGMP biosensors has led to several conceptual advances, such as the discovery of local, intercellular and mechanosensitive cGMP signals. Importantly, single-cell imaging can provide valuable information about the heterogeneity of cGMP signals within and between individual cells of an isolated cell population or tissue. We also discuss current challenges and future directions of cGMP imaging, such as the direct visualisation of cGMP microdomains, simultaneous monitoring of cGMP and other signalling molecules and, ultimately, cGMP imaging in tissues and animals under close-to-native conditions.
Collapse
Affiliation(s)
- Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Chang P, Zhang X, Chen W, Zhang J, Wang J, Wang X, Yu J, Zhu X. Vasonatrin peptide, a synthetic natriuretic peptide, attenuates myocardial injury and oxidative stress in isoprenaline-induced cardiomyocyte hypertrophy. Peptides 2021; 137:170474. [PMID: 33359394 DOI: 10.1016/j.peptides.2020.170474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/17/2022]
Abstract
Isoprenaline-induced cardiac hypertrophy can deteriorate to heart failure, which is a leading cause of mortality. Endogenous vasonatrin peptide (VNP) has been reported to be cardioprotective against myocardial ischemia/reperfusion injury in diabetic rats. However, little is known about the effect of exogenous VNP on cardiac hypertrophy. We further explored whether VNP attenuated isoprenaline-induced cardiomyocyte hypertrophy by examining the levels and activities of cGMP and PKG. In this study, we found that VNP significantly attenuated isoprenaline-induced myocardial hypertrophy and cardiac fibroblast activation in vivo. Moreover, VNP effectively halted the activation of apoptosis and oxidative stress in the isoprenaline-treated myocardium. VNP promoted superoxide dismutase (SOD) activity. Further study revealed that the protective effects of VNP might be mediated by the activity of the cGMP-PKG signaling pathway in vivo or in vitro, while the use of agonists and antagonists confirmed these results. Therefore, we demonstrated that the antiapoptosis and antioxidative stress effects of VNP depends on elevated cGMP-PKG signaling activity both in vivo and in vitro. These results suggest that VNP may be used in the treatment of myocardial hypertrophy.
Collapse
Affiliation(s)
- Pan Chang
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China
| | - Xiaomeng Zhang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiguo Chen
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China
| | - Jing Zhang
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China
| | - Jianbang Wang
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China
| | - Xihui Wang
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China
| | - Jun Yu
- Department of Cardiology, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710038, China; Clinical Experimental Center, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, China.
| | - Xiaoling Zhu
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
29
|
Zhang K, Qin X, Wen P, Wu Y, Zhuang J. Systematic analysis of molecular mechanisms of heart failure through the pathway and network-based approach. Life Sci 2020; 265:118830. [PMID: 33259868 DOI: 10.1016/j.lfs.2020.118830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
AIMS The molecular networks and pathways involved in heart failure (HF) are still largely unknown. The present study aimed to systematically investigate the genes associated with HF, comprehensively explore their interactions and functions, and identify possible regulatory networks involved in HF. MAIN METHODS The weighted gene coexpression network analysis (WGCNA), crosstalk analysis, and Pivot analysis were used to identify gene connections, interaction networks, and molecular regulatory mechanisms. Functional analysis and protein-protein interaction (PPI) were performed using DAVID and STRING databases. Gene set variation analysis (GSVA) and receiver operating characteristic (ROC) curve analysis were also performed to evaluate the relationship of the hub genes with HF. KEY FINDINGS A total of 5968 HF-related genes were obtained to construct the co-expression networks, and 18 relatively independent and closely linked modules were identified. Pivot analysis suggested that four transcription factors and five noncoding RNAs were involved in regulating the process of HF. The genes in the module with the highest positive correlation to HF was mainly enriched in cardiac remodeling and response to stress. Five upregulated hub genes (ASPN, FMOD, NT5E, LUM, and OGN) were identified and validated. Furthermore, the GSVA scores of the five hub genes for HF had a relatively high areas under the curve (AUC). SIGNIFICANCE The results of this study revealed specific molecular networks and their potential regulatory mechanisms involved in HF. These may provide new insight into understanding the mechanisms underlying HF and help to identify more effective therapeutic targets for HF.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xianyu Qin
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Pengju Wen
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yueheng Wu
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Jian Zhuang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
30
|
Mishra S, Dunkerly-Eyring BL, Keceli G, Ranek MJ. Phosphorylation Modifications Regulating Cardiac Protein Quality Control Mechanisms. Front Physiol 2020; 11:593585. [PMID: 33281625 PMCID: PMC7689282 DOI: 10.3389/fphys.2020.593585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many forms of cardiac disease, including heart failure, present with inadequate protein quality control (PQC). Pathological conditions often involve impaired removal of terminally misfolded proteins. This results in the formation of large protein aggregates, which further reduce cellular viability and cardiac function. Cardiomyocytes have an intricately collaborative PQC system to minimize cellular proteotoxicity. Increased expression of chaperones or enhanced clearance of misfolded proteins either by the proteasome or lysosome has been demonstrated to attenuate disease pathogenesis, whereas reduced PQC exacerbates pathogenesis. Recent studies have revealed that phosphorylation of key proteins has a potent regulatory role, both promoting and hindering the PQC machinery. This review highlights the recent advances in phosphorylations regulating PQC, the impact in cardiac pathology, and the therapeutic opportunities presented by harnessing these modifications.
Collapse
Affiliation(s)
- Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brittany L Dunkerly-Eyring
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
31
|
Kopra K, Sharina I, Martin E, Härmä H. Homogeneous single-label cGMP detection platform for the functional study of nitric oxide-sensitive (soluble) guanylyl cyclases and cGMP-specific phosphodiesterases. Sci Rep 2020; 10:17469. [PMID: 33060787 PMCID: PMC7562898 DOI: 10.1038/s41598-020-74611-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
Cardiovascular diseases are the number one death worldwide. Nitric oxide (NO)-NO-sensitive (soluble) guanylyl cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway regulates diverse set of important physiological functions, including maintenance of cardiovascular homeostasis. Resting and activated sGC enzyme converts guanosine triphosphate to an important second messenger cGMP. In addition to traditional NO generators, a number of sGC activators and stimulators are currently in clinical trials aiming to support or increase sGC activity in various pathological conditions. cGMP-specific phosphodiesterases (PDEs), which degrade cGMP to guanosine monophosphate, play key role in controlling the cGMP level and the strength or length of the cGMP-dependent cellular signaling. Thus, PDE inhibitors also have clear clinical applications. Here, we introduce a homogeneous quenching resonance energy transfer (QRET) for cGMP to monitor both sGC and PDE activities using high throughput screening adoptable method. We demonstrate that using cGMP-specific antibody, sGC or PDE activity and the effect of small molecules modulating their function can be studied with sub-picomole cGMP sensitivity. The results further indicate that the method is suitable for monitoring enzyme reactions also in complex biological cellular homogenates and mixture.
Collapse
Affiliation(s)
- Kari Kopra
- Department of Chemistry, Chemistry of Drug Development, University of Turku, Vatselankatu 2, 20500, Turku, Finland.
| | - Iraida Sharina
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical School At Houston, 1941 East Road, Houston, TX, 77054, USA
| | - Emil Martin
- Division of Cardiology, Department of Internal Medicine, University of Texas Medical School At Houston, 1941 East Road, Houston, TX, 77054, USA
| | - Harri Härmä
- Department of Chemistry, Chemistry of Drug Development, University of Turku, Vatselankatu 2, 20500, Turku, Finland
| |
Collapse
|
32
|
Lan T, Li Q, Chang M, Yin C, Zhu D, Wu Z, Li X, Zhang W, Yue B, Shi J, Yuan H, Su Z, Guo H. Lei-gong-gen formula granule attenuates hyperlipidemia in rats via cGMP-PKG signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:112989. [PMID: 32526339 DOI: 10.1016/j.jep.2020.112989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/14/2020] [Accepted: 05/16/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lei-gong-gen formula granule (LFG) is a folk prescription derived from Zhuang nationality, the largest ethnic minority among the 56 nationalities in China. It is composed of three herbs, namely Centella asiatica (L.) Urb., Eclipta prostrata (L.) L., Smilax glabra Roxb. It has been widely used as health protection tea for many years to prevent cardiovascular and cerebrovascular diseases such as hyperlipidemia and hypertension. AIM OF THE STUDY This study validated the lipid-lowering effect of LFG in a hyperlipidemia rat model. Then we employed network pharmacology and molecular biological approach to identify the active ingredients of LFG, corresponding targets, and its anti-hyperlipidemia mechanisms. MATERIALS AND METHODS Hyperlipidemia rat model was established by feeding male Sprague-Dawley rats with high-fat diet for two weeks. LFG (two doses of 10 and 20 g/kg) was administered orally to hyperlipidemia rat model for 4 weeks, twice per day. Serum levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) were monitored in rats pre and post-treatment. Hematoxylin-eosin staining was applied to observe the pathology and lipid accumulation of liver. We then performed network pharmacology analysis to predict the ingredients, their associated targets, and hyperlipidemia associated targets. Pathway analysis with significant genes was carried out using KEGG pathway. These genes and proteins intersectioned between compound targets and hyperlipidemia targets were further verified with samples from hyperlipidemia rats treated with LFG using Real-time RT-PCR and Western Blot. RESULTS LFG attenuated hyperlipidemia in rat model, and this was characterized with decreased serum levels of TC, LDL-C, liver wet weight, and liver index. LFG alleviated the hepatic steatosis in hyperlipidemia rats. Network pharmacology analysis identified 53 bioactive ingredients from LFG formula (three herbs), which link to 765 potential targets. 53 hyperlipidemia associated genes were retrieved from public databases. There were 10 common genes between ingredients-targets and hyperlipidemia associated genes, which linked to 20 bioactive ingredients. Among these 10 genes, 3 of them were validated to be involved in LFG's anti-hyperlipidemia effect using Real-time RT-PCR, namely ADRB2 encoding beta-2 adrenergic receptor, NOS3 encoding nitric oxide synthase 3, LDLR encoding low-density lipoprotein receptor. The cGMP-PKG signaling pathway was enriched for hyperlipidemia after pharmacology network analysis with ADRB2, NOS3, and LDLR. Interestingly, expression of cGMP-dependent protein kinase (PKG) was downregulated in hyperlipidemia rat after LFG treatment. Molecular docking study further supported that ferulic acid, histidine, p-hydroxybenzoic acid, and linalool were potential active ingredients for LFG's anti-hyperlipidemia effect. LC-MS/MS analysis confirmed that ferulic acid and p-hydroxybenzoic acid were active ingredients of LFG. CONCLUSION LFG exhibited the lipid-lowering effect, which might be attributed to downregulating ADRB2 and NOS3, and upregulating LDLR through the cGMP-PKG signaling pathway in hyperlipidemia rat. Ferulic acid and p-hydroxybenzoic acid might be the underlying active ingredients which affect the potential targets for their anti-hyperlipidemia effect.
Collapse
Affiliation(s)
- Taijin Lan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu Dong Road, Nanning, 530001, China
| | - Qiaofeng Li
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; School of Preclinical Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Ming Chang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; School of Preclinical Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Chunli Yin
- College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Dan Zhu
- College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Zheng Wu
- College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Xiaolan Li
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Weiquan Zhang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Bangwen Yue
- College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Junlin Shi
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Hebao Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 1600 Huron Parkway, MI, 48109, USA.
| | - Zhiheng Su
- College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China.
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China; College of Pharmacy, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, 530021, China.
| |
Collapse
|
33
|
cGMP via PKG activates 26S proteasomes and enhances degradation of proteins, including ones that cause neurodegenerative diseases. Proc Natl Acad Sci U S A 2020; 117:14220-14230. [PMID: 32513741 PMCID: PMC7321992 DOI: 10.1073/pnas.2003277117] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Most studies of the regulation of proteolysis by the ubiquitin proteasome system have focused on the control of ubiquitination. However, it is now clear that the activity of the 26S proteasome and rates of protein degradation in cells are also tightly regulated through proteasome phosphorylation. Here we demonstrate that agents that raise cGMP and activate cGMP-dependent protein kinase (e.g., widely used phosphodiesterase 5 inhibitors) stimulate proteasome activities and intracellular proteolysis without affecting autophagy. Furthermore, we showed that raising cGMP reduced the levels of the disease-causing mutant tau in a zebrafish model by increasing its degradation, and also decreased the associated morphological abnormalities. Thus, activating the proteasome via cGMP is a promising strategy to prevent the progression of neurodegenerative diseases. Because raising cAMP enhances 26S proteasome activity and the degradation of cell proteins, including the selective breakdown of misfolded proteins, we investigated whether agents that raise cGMP may also regulate protein degradation. Treating various cell lines with inhibitors of phosphodiesterase 5 or stimulators of soluble guanylyl cyclase rapidly enhanced multiple proteasome activities and cellular levels of ubiquitinated proteins by activating protein kinase G (PKG). PKG stimulated purified 26S proteasomes by phosphorylating a different 26S component than is modified by protein kinase A. In cells and cell extracts, raising cGMP also enhanced within minutes ubiquitin conjugation to cell proteins. Raising cGMP, like raising cAMP, stimulated the degradation of short-lived cell proteins, but unlike cAMP, also markedly increased proteasomal degradation of long-lived proteins (the bulk of cell proteins) without affecting lysosomal proteolysis. We also tested if raising cGMP, like cAMP, can promote the degradation of mutant proteins that cause neurodegenerative diseases. Treating zebrafish models of tauopathies or Huntington’s disease with a PDE5 inhibitor reduced the levels of the mutant huntingtin and tau proteins, cell death, and the resulting morphological abnormalities. Thus, PKG rapidly activates cytosolic proteasomes, protein ubiquitination, and overall protein degradation, and agents that raise cGMP may help combat the progression of neurodegenerative diseases.
Collapse
|
34
|
Wen JJ, Cummins C, Radhakrishnan RS. Sildenafil Recovers Burn-Induced Cardiomyopathy. Cells 2020. [DOI: https:/doi.org/10.3390/cells9061393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background: Severe burn injury initiates a feedback cycle of inflammation, fibrosis, oxidative stress and cardiac mitochondrial damage via the PDE5A-cGMP-PKG pathway. Aim: To test if the PDE5A-cGMP-PKG pathway may contribute to burn-induced heart dysfunction. Methods: Sprague–Dawley rats were divided four groups: sham; sham/sildenafil; 24 h post burn (60% total body surface area scald burn, harvested at 24 h post burn); and 24 h post burn/sildenafil. We monitored heart function and oxidative adducts, as well as cardiac inflammatory, cardiac fibrosis and cardiac remodeling responses in vivo. Results: Sildenafil inhibited the burn-induced PDE5A mRNA level and increased the cGMP level and PKG activity, leading to the normalization of PKG down-regulated genes (IRAG, PLB, RGS2, RhoA and MYTP), a decreased ROS level (H2O2), decreased oxidatively modified adducts (malonyldialdehyde [MDA], carbonyls), attenuated fibrogenesis as well as fibrosis gene expression (ANP, BNP, COL1A2, COL3A2, αSMA and αsk-Actin), and reduced inflammation and related gene expression (RELA, IL-18 and TGF-β) after the burn. Additionally, sildenafil treatment preserved left ventricular heart function (CO, EF, SV, LVvol at systolic, LVPW at diastolic and FS) and recovered the oxidant/antioxidant balance (total antioxidant, total SOD activity and Cu,ZnSOD activity). Conclusions: The PDE5A-cGMP-PKG pathway mediates burn-induced heart dysfunction. Sildenafil treatment recovers burn-induced cardiac dysfunction.
Collapse
|
35
|
Sildenafil Recovers Burn-Induced Cardiomyopathy. Cells 2020; 9:cells9061393. [PMID: 32503314 PMCID: PMC7349507 DOI: 10.3390/cells9061393] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Severe burn injury initiates a feedback cycle of inflammation, fibrosis, oxidative stress and cardiac mitochondrial damage via the PDE5A-cGMP-PKG pathway. Aim: To test if the PDE5A-cGMP-PKG pathway may contribute to burn-induced heart dysfunction. Methods: Sprague–Dawley rats were divided four groups: sham; sham/sildenafil; 24 h post burn (60% total body surface area scald burn, harvested at 24 h post burn); and 24 h post burn/sildenafil. We monitored heart function and oxidative adducts, as well as cardiac inflammatory, cardiac fibrosis and cardiac remodeling responses in vivo. Results: Sildenafil inhibited the burn-induced PDE5A mRNA level and increased the cGMP level and PKG activity, leading to the normalization of PKG down-regulated genes (IRAG, PLB, RGS2, RhoA and MYTP), a decreased ROS level (H2O2), decreased oxidatively modified adducts (malonyldialdehyde [MDA], carbonyls), attenuated fibrogenesis as well as fibrosis gene expression (ANP, BNP, COL1A2, COL3A2, αSMA and αsk-Actin), and reduced inflammation and related gene expression (RELA, IL-18 and TGF-β) after the burn. Additionally, sildenafil treatment preserved left ventricular heart function (CO, EF, SV, LVvol at systolic, LVPW at diastolic and FS) and recovered the oxidant/antioxidant balance (total antioxidant, total SOD activity and Cu,ZnSOD activity). Conclusions: The PDE5A-cGMP-PKG pathway mediates burn-induced heart dysfunction. Sildenafil treatment recovers burn-induced cardiac dysfunction.
Collapse
|
36
|
Seo K, Parikh VN, Ashley EA. Stretch-Induced Biased Signaling in Angiotensin II Type 1 and Apelin Receptors for the Mediation of Cardiac Contractility and Hypertrophy. Front Physiol 2020; 11:181. [PMID: 32231588 PMCID: PMC7082839 DOI: 10.3389/fphys.2020.00181] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
The myocardium has an intrinsic ability to sense and respond to mechanical load in order to adapt to physiological demands. Primary examples are the augmentation of myocardial contractility in response to increased ventricular filling caused by either increased venous return (Frank-Starling law) or aortic resistance to ejection (the Anrep effect). Sustained mechanical overload, however, can induce pathological hypertrophy and dysfunction, resulting in heart failure and arrhythmias. It has been proposed that angiotensin II type 1 receptor (AT1R) and apelin receptor (APJ) are primary upstream actors in this acute myocardial autoregulation as well as the chronic maladaptive signaling program. These receptors are thought to have mechanosensing capacity through activation of intracellular signaling via G proteins and/or the multifunctional transducer protein, β-arrestin. Importantly, ligand and mechanical stimuli can selectively activate different downstream signaling pathways to promote inotropic, cardioprotective or cardiotoxic signaling. Studies to understand how AT1R and APJ integrate ligand and mechanical stimuli to bias downstream signaling are an important and novel area for the discovery of new therapeutics for heart failure. In this review, we provide an up-to-date understanding of AT1R and APJ signaling pathways activated by ligand versus mechanical stimuli, and their effects on inotropy and adaptive/maladaptive hypertrophy. We also discuss the possibility of targeting these signaling pathways for the development of novel heart failure therapeutics.
Collapse
Affiliation(s)
- Kinya Seo
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Victoria N. Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Euan A. Ashley
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
- Department of Genetics, Stanford University, Stanford, CA, United States
| |
Collapse
|
37
|
Wan X, Belanger K, Widen SG, Kuyumcu-Martinez MN, Garg NJ. Genes of the cGMP-PKG-Ca 2+ signaling pathway are alternatively spliced in cardiomyopathy: Role of RBFOX2. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165620. [PMID: 31778749 PMCID: PMC6954967 DOI: 10.1016/j.bbadis.2019.165620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022]
Abstract
Aberrations in the cGMP-PKG-Ca2+ pathway are implicated in cardiovascular complications of diverse etiologies, though involved molecular mechanisms are not understood. We performed RNA-Seq analysis to profile global changes in gene expression and exon splicing in Chagas disease (ChD) murine myocardium. Ingenuity-Pathway-Analysis of transcriptome dataset identified 26 differentially expressed genes associated with increased mobilization and cellular levels of Ca2+ in ChD hearts. Mixture-of-isoforms and Enrichr KEGG pathway analyses of the RNA-Seq datasets from ChD (this study) and diabetic (previous study) murine hearts identified alternative splicing (AS) in eleven genes (Arhgef10, Atp2b1, Atp2a3, Cacna1c, Itpr1, Mef2a, Mef2d, Pde2a, Plcb1, Plcb4, and Ppp1r12a) of the cGMP-PKG-Ca2+ pathway in diseased hearts. AS of these genes was validated by an exon exclusion-inclusion assay. Further, Arhgef10, Atp2b1, Mef2a, Mef2d, Plcb1, and Ppp1r12a genes consisted RBFOX2 (RNA-binding protein) binding-site clusters, determined by analyzing the RBFOX2 CLIP-Seq dataset. H9c2 rat heart cells transfected with Rbfox2 (vs. scrambled) siRNA confirmed that expression of Rbfox2 is essential for proper exon splicing of genes of the cGMP-PKG-Ca2+ pathway. We conclude that changes in gene expression may influence the Ca2+ mobilization pathway in ChD, and AS impacts the genes involved in cGMP/PKG/Ca2+ signaling pathway in ChD and diabetes. Our findings suggest that ChD patients with diabetes may be at increased risk of cardiomyopathy and heart failure and provide novel ways to restore cGMP-PKG regulated signaling networks via correcting splicing patterns of key factors using oligonucleotide-based therapies for the treatment of cardiovascular complications.
Collapse
Affiliation(s)
- Xianxiu Wan
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, 77555-1070, TX, United States of America
| | - KarryAnne Belanger
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America
| | - Steven G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America
| | - Muge N Kuyumcu-Martinez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 77555, TX, United States of America.
| | - Nisha J Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, 77555-1070, TX, United States of America; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, 77555, TX, United States of America.
| |
Collapse
|
38
|
cGMP signalling in cardiomyocyte microdomains. Biochem Soc Trans 2020; 47:1327-1339. [PMID: 31652306 DOI: 10.1042/bst20190225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
3',5'-Cyclic guanosine monophosphate (cGMP) is one of the major second messengers critically involved in the regulation of cardiac electrophysiology, hypertrophy, and contractility. Recent molecular and cellular studies have significantly advanced our understanding of the cGMP signalling cascade, its local microdomain-specific regulation and its role in protecting the heart from pathological stress. Here, we summarise recent findings on cardiac cGMP microdomain regulation and discuss their potential clinical significance.
Collapse
|
39
|
Chang P, Zhang X, Zhang M, Li G, Hu L, Zhao H, Zhu X, Wu J, Wang X, Wang K, Zhang J, Ren M, Chen B, Zhu X, Zhu M, Yu J. Swimming exercise inhibits myocardial ER stress in the hearts of aged mice by enhancing cGMP‑PKG signaling. Mol Med Rep 2019; 21:549-556. [PMID: 31974605 PMCID: PMC6947875 DOI: 10.3892/mmr.2019.10864] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022] Open
Abstract
The purpose of the present study was to explore aging‑associated cardiac dysfunction and the possible mechanism by which swimming exercise modulates cardiac dysfunction in aged mice. Aged mice were divided into two groups: i) Aged mice; and ii) aged mice subjected to swimming exercises. Another cohort of 4‑month‑old male mice served as the control group. Cardiac structure and function in mice were analyzed using hematoxylin and eosin staining, and echocardiography. The levels of oxidative stress were determined by measuring the levels of superoxide dismutase, malondialdehyde and reactive oxygen species (ROS). Levels of the endoplasmic reticulum (ER) stress‑related protein PKR‑like ER kinase, glucose‑regulated protein 78 and C/EBP homologous protein were determined to evaluate the level of ER stress. The aged group exhibited an abnormal cardiac structure and decreased cardiac function, both of which were ameliorated by swimming exercise. The hearts of the aged mice exhibited pronounced oxidative and ER stress, which were ameliorated by exercise, and was accompanied by the reactivation of myocardial cGMP and suppression of cGMP‑specific phosphodiesterase type 5 (PDE5). The inhibition of PDE5 attenuated age‑induced cardiac dysfunction, blocked ROS production and suppressed ER stress. An ER stress inducer abolished the beneficial effects of the swimming exercise on cardiac function and increased ROS production. The present study suggested that exercise restored cardiac function in mice with age‑induced cardiac dysfunction by inhibiting oxidative stress and ER stress, and increasing cGMP‑protein kinase G signaling.
Collapse
Affiliation(s)
- Pan Chang
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaomeng Zhang
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Mingyang Zhang
- Institute of Forensic Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Guohua Li
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Lang Hu
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Huishou Zhao
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xiaoxing Zhu
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Juan Wu
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Xihui Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Kaiyan Wang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Jing Zhang
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Minggang Ren
- Department of Cardiology, The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Baoying Chen
- Central Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Xiaoling Zhu
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Miaozhang Zhu
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jun Yu
- Department of Physiology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
40
|
Soares ROS, Losada DM, Jordani MC, Évora P, Castro-E-Silva O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int J Mol Sci 2019; 20:ijms20205034. [PMID: 31614478 PMCID: PMC6834141 DOI: 10.3390/ijms20205034] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) permeates a variety of diseases and is a ubiquitous concern in every transplantation proceeding, from whole organs to modest grafts. Given its significance, efforts to evade the damaging effects of both ischemia and reperfusion are abundant in the literature and they consist of several strategies, such as applying pre-ischemic conditioning protocols, improving protection from preservation solutions, thus providing extended cold ischemia time and so on. In this review, we describe many of the latest pharmacological approaches that have been proven effective against IRI, while also revisiting well-established concepts and presenting recent pathophysiological findings in this ever-expanding field. A plethora of promising protocols has emerged in the last few years. They have been showing exciting results regarding protection against IRI by employing drugs that engage several strategies, such as modulating cell-surviving pathways, evading oxidative damage, physically protecting cell membrane integrity, and enhancing cell energetics.
Collapse
Affiliation(s)
| | - Daniele M Losada
- Department of Anatomic Pathology, Faculty of Medical Sciences, University of Campinas, 13083-970 Campinas, Brazil.
| | - Maria C Jordani
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Paulo Évora
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| | - Orlando Castro-E-Silva
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| |
Collapse
|
41
|
Mitrokhin MV, Kalsin V, Kamkina O, Babkina I, Zotov A, Troitskiy VA, Mladenov MI, Kamkin GA. Participation of PKG and PKA-related pathways in the IFN-γ induced modulation of the BK Ca channel activity in human cardiac fibroblasts. J Pharmacol Sci 2019; 141:25-31. [PMID: 31533896 DOI: 10.1016/j.jphs.2019.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/31/2019] [Accepted: 08/20/2019] [Indexed: 01/19/2023] Open
Abstract
This study was devoted to elucidating the interferon (IFN)-γ-induced signaling pathway and the interaction between protein kinase G (PKG) and protein kinase A (PKA) through large-conductance Ca(2+)-activated K(+) channels in human cardiac fibroblasts. The IK currents were recorded using a whole-cell patch clamp method. A large depolarization (+50 mV) and a high Ca2+ concentration (pCa 6.0) were used in the internal pipette solution to activate only the KCa channels. Iberiotoxin (Ibtx), which selectively inhibits BKCa channels at a concentration of 100 nmol/l, caused a significant reduction of basal IK. Adding IFN-γ in the presence of Ibtx had no effect on IK. Application of the IFN-γ caused a significant reduction in total K+ current amplitude, recorded with a 500 ms depolarizing pulse duration, to +50 mV from a holding potential of -80 mV. We tested the involvement of the sGC/cGMP/PKG signaling pathway by using specific PKG inhibitor KT 5823, potent sGC inhibitor NS 2028, and specific sGC agonist BAY 41-8543. The obtained data confirmed that only sGC participated in the IFN-γ-mediated BKCa channel modulation, which was mediated further by PKA. This study represents first evidence about the participation of the IFN-γ in the mechanisms responsible for BKCa modulation in HCFs. We also believe that this process occurs via negative crosstalk between the PKG- and PKA-associated pathways.
Collapse
Affiliation(s)
- M V Mitrokhin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Ostrovitjanova 1, Moscow 117997, Russia
| | - V Kalsin
- Federal Scientific Clinical Center for Specialized Types of Medical Assistance and Medical Technologies for the Federal Medical and Biological Agency, Orekhoviy Boulevard 28, Moscow 115682, Russia
| | - O Kamkina
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Ostrovitjanova 1, Moscow 117997, Russia
| | - I Babkina
- Federal Scientific Clinical Center for Specialized Types of Medical Assistance and Medical Technologies for the Federal Medical and Biological Agency, Orekhoviy Boulevard 28, Moscow 115682, Russia
| | - A Zotov
- Federal Scientific Clinical Center for Specialized Types of Medical Assistance and Medical Technologies for the Federal Medical and Biological Agency, Orekhoviy Boulevard 28, Moscow 115682, Russia
| | - V A Troitskiy
- Federal Scientific Clinical Center for Specialized Types of Medical Assistance and Medical Technologies for the Federal Medical and Biological Agency, Orekhoviy Boulevard 28, Moscow 115682, Russia
| | - M I Mladenov
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Ostrovitjanova 1, Moscow 117997, Russia; Faculty of Natural Sciences and Mathematics, Institute of Biology, "Ss. Cyril and Methodius" University, P.O. Box 162, 1000 Skopje, Macedonia.
| | - G A Kamkin
- Department of Fundamental and Applied Physiology, Russian National Research Medical University, Ostrovitjanova 1, Moscow 117997, Russia
| |
Collapse
|
42
|
Lyle MA, Brozovich FV. HFpEF, a Disease of the Vasculature: A Closer Look at the Other Half. Mayo Clin Proc 2018; 93:1305-1314. [PMID: 30064827 DOI: 10.1016/j.mayocp.2018.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 04/12/2018] [Accepted: 05/04/2018] [Indexed: 12/31/2022]
Abstract
Patients with heart failure are commonly divided into those with reduced ejection fraction (EF<40%) and those with preserved ejection fraction (HFpEF; EF>50%). For heart failure with reduced EF, a number of therapies have been found to improve patient morbidity and mortality, and treatment is guideline based. However for patients with HFpEF, no treatment has been found to have clinical benefit. To objectively assess treatments for HFpEF, a comprehensive PubMed literature search was performed using the terms HFpEF, heart failure, smooth muscle, myosin, myosin phosphatase, and PKG (up to December 31, 2017), with an unbiased focus on pathophysiology, cell signaling, and therapy. This review provides evidence that could explain the lack of clinical benefit in treating patients with HFpEF with sildenafil and long-acting nitrates. Furthermore, the review highlights the vascular abnormalities present in patients with HFpEF, and these abnormalities of the vasculature could potentially contribute to the pathophysiology of HFpEF. Thus, focusing on HFpEF as a vascular disease could result in the development of novel and effective treatment paradigms.
Collapse
Affiliation(s)
- Melissa A Lyle
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN
| | - Frank V Brozovich
- Department of Cadiovascular Diseases, Mayo Clinic College of Medicine and Science, Rochester, MN.
| |
Collapse
|
43
|
Marques FZ, Chu PY, Ziemann M, Kaspi A, Kiriazis H, Du XJ, El-Osta A, Kaye DM. Age-Related Differential Structural and Transcriptomic Responses in the Hypertensive Heart. Front Physiol 2018; 9:817. [PMID: 30038575 PMCID: PMC6046461 DOI: 10.3389/fphys.2018.00817] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/11/2018] [Indexed: 01/20/2023] Open
Abstract
While aging is a critical risk factor for heart failure, it remains uncertain whether the aging heart responds differentially to a hypertensive stimuli. Here we investigated phenotypic and transcriptomic differences between the young and aging heart using a mineralocorticoid-excess model of hypertension. Ten-week (“young”) and 36-week (“aging”) mice underwent a unilateral uninephrectomy with deoxycorticosterone acetate (DOCA) pellet implantation (n = 6–8/group) and were followed for 6 weeks. Cardiac structure and function, blood pressure (BP) and the cardiac transcriptome were subsequently examined. Young and aging DOCA mice had high BP, increased cardiac mass, cardiac hypertrophy, and fibrosis. Left ventricular end-diastolic pressure increased in aging DOCA-treated mice in contrast to young DOCA mice. Interstitial and perivascular fibrosis occurred in response to DOCA, but perivascular fibrosis was greater in aging mice. Transcriptomic analysis showed that young mice had features of higher oxidative stress, likely due to activation of the respiratory electron transport chain. In contrast, aging mice showed up-regulation of collagen formation in association with activation of innate immunity together with markers of inflammation including cytokine and platelet signaling. In comparison to younger mice, aging mice demonstrated different phenotypic and molecular responses to hypertensive stress. These findings have potential implications for the pathogenesis of age-related forms of cardiovascular disease, particularly heart failure.
Collapse
Affiliation(s)
- Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Po-Yin Chu
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Antony Kaspi
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia.,Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Heart Centre, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
44
|
Chen L, Cao Y, Zhang H, Lv D, Zhao Y, Liu Y, Ye G, Chai Y. Network pharmacology-based strategy for predicting active ingredients and potential targets of Yangxinshi tablet for treating heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:359-368. [PMID: 29366769 DOI: 10.1016/j.jep.2017.12.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/14/2017] [Accepted: 12/11/2017] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yangxinshi tablet (YXST) is an effective treatment for heart failure and myocardial infarction; it consists of 13 herbal medicines formulated according to traditional Chinese Medicine (TCM) practices. It has been used for the treatment of cardiovascular disease for many years in China. MATERIALS AND METHODS In this study, a network pharmacology-based strategy was used to elucidate the mechanism of action of YXST for the treatment of heart failure. Cardiovascular disease-related protein target and compound databases were constructed for YXST. A molecular docking platform was used to predict the protein targets of YXST. The affinity between proteins and ingredients was determined using surface plasmon resonance (SPR) assays. The action modes between targets and representative ingredients were calculated using Glide docking, and the related pathways were predicted using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. RESULTS A protein target database containing 924 proteins was constructed; 179 compounds in YXST were identified, and 48 compounds with high relevance to the proteins were defined as representative ingredients. Thirty-four protein targets of the 48 representative ingredients were analyzed and classified into two categories: immune and cardiovascular systems. The SPR assay and molecular docking partly validated the interplay between protein targets and representative ingredients. Moreover, 28 pathways related to heart failure were identified, which provided directions for further research on YXST. CONCLUSIONS This study demonstrated that the cardiovascular protective effect of YXST mainly involved the immune and cardiovascular systems. Through the research strategy based on network pharmacology, we analysis the complex system of YXST and found 48 representative compounds, 34 proteins and 28 related pathways of YXST, which could help us understand the underlying mechanism of YSXT's anti-heart failure effect. The network-based investigation could help researchers simplify the complex system of YXSY. It may also offer a feasible approach to decipher the chemical and pharmacological bases of other TCM formulas.
Collapse
Affiliation(s)
- Langdong Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yan Cao
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, China
| | - Diya Lv
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Yahong Zhao
- Central Research Institute, Shanghai Pharmaceuticals Holding Co. Ltd., Shanghai 201203, China
| | - Yanjun Liu
- Central Research Institute, Shanghai Pharmaceuticals Holding Co. Ltd., Shanghai 201203, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co. Ltd., Shanghai 201203, China.
| | - Yifeng Chai
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
45
|
Bork NI, Nikolaev VO. cGMP Signaling in the Cardiovascular System-The Role of Compartmentation and Its Live Cell Imaging. Int J Mol Sci 2018. [PMID: 29534460 PMCID: PMC5877662 DOI: 10.3390/ijms19030801] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The ubiquitous second messenger 3′,5′-cyclic guanosine monophosphate (cGMP) regulates multiple physiologic processes in the cardiovascular system. Its intracellular effects are mediated by stringently controlled subcellular microdomains. In this review, we will illustrate the current techniques available for real-time cGMP measurements with a specific focus on live cell imaging methods. We will also discuss currently accepted and emerging mechanisms of cGMP compartmentation in the cardiovascular system.
Collapse
Affiliation(s)
- Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg 20246, Germany.
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Hamburg 20246, Germany.
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg 20246, Germany.
- German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Hamburg 20246, Germany.
| |
Collapse
|
46
|
Abstract
Novel cardioprotective agents are needed in both heart failure (HF) and myocardial infarction. Increasing evidence from cellular studies and animal models indicate protective effects of phosphodiesterase-5 (PDE5) inhibitors, drugs usually reserved as treatments of erectile dysfunction and pulmonary arterial hypertension. PDE5 inhibitors have been shown to improve contractile function in systolic HF, regress left ventricular hypertrophy, reduce myocardial infarct size and suppress ischaemia-induced ventricular arrhythmias. Underpinning these actions are complex but increasingly understood cellular mechanisms involving the cyclic GMP activation of protein kinase-G in both cardiac myocytes and the vasculature. In clinical trials, PDE5 inhibitors improve symptoms and ventricular function in systolic HF, and accumulating epidemiological data indicate a reduction in cardiovascular events and mortality in PDE5 inhibitor users at high cardiovascular risk. Here, we focus on the translation of underpinning basic science to clinical studies and report that PDE5 inhibitors act through a number of cardioprotective mechanisms, including a direct myocardial action independent of the vasculature. We conclude that future clinical trials should be designed with these mechanisms in mind to identify patient subsets that derive greatest treatment benefit from these novel cardioprotective agents.
Collapse
Affiliation(s)
- David Charles Hutchings
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Simon George Anderson
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jessica L Caldwell
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
47
|
Pavlaki N, Nikolaev VO. Imaging of PDE2- and PDE3-Mediated cGMP-to-cAMP Cross-Talk in Cardiomyocytes. J Cardiovasc Dev Dis 2018; 5:jcdd5010004. [PMID: 29367582 PMCID: PMC5872352 DOI: 10.3390/jcdd5010004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022] Open
Abstract
Cyclic nucleotides 3′,5′-cyclic adenosine monophosphate (cAMP) and 3′,5′-cyclic guanosine monophosphate (cGMP) are important second messengers that regulate cardiovascular function and disease by acting in discrete subcellular microdomains. Signaling compartmentation at these locations is often regulated by phosphodiesterases (PDEs). Some PDEs are also involved in the cross-talk between the two second messengers. The purpose of this review is to summarize and highlight recent findings about the role of PDE2 and PDE3 in cardiomyocyte cyclic nucleotide compartmentation and visualization of this process using live cell imaging techniques.
Collapse
Affiliation(s)
- Nikoleta Pavlaki
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany.
| |
Collapse
|
48
|
Gorter TM, van Veldhuisen DJ, Bauersachs J, Borlaug BA, Celutkiene J, Coats AJS, Crespo-Leiro MG, Guazzi M, Harjola VP, Heymans S, Hill L, Lainscak M, Lam CSP, Lund LH, Lyon AR, Mebazaa A, Mueller C, Paulus WJ, Pieske B, Piepoli MF, Ruschitzka F, Rutten FH, Seferovic PM, Solomon SD, Shah SJ, Triposkiadis F, Wachter R, Tschöpe C, de Boer RA. Right heart dysfunction and failure in heart failure with preserved ejection fraction: mechanisms and management. Position statement on behalf of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2017; 20:16-37. [PMID: 29044932 DOI: 10.1002/ejhf.1029] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 08/16/2017] [Accepted: 09/05/2017] [Indexed: 12/28/2022] Open
Abstract
There is an unmet need for effective treatment strategies to reduce morbidity and mortality in patients with heart failure with preserved ejection fraction (HFpEF). Until recently, attention in patients with HFpEF was almost exclusively focused on the left side. However, it is now increasingly recognized that right heart dysfunction is common and contributes importantly to poor prognosis in HFpEF. More insights into the development of right heart dysfunction in HFpEF may aid to our knowledge about this complex disease and may eventually lead to better treatments to improve outcomes in these patients. In this position paper from the Heart Failure Association of the European Society of Cardiology, the Committee on Heart Failure with Preserved Ejection Fraction reviews the prevalence, diagnosis, and pathophysiology of right heart dysfunction and failure in patients with HFpEF. Finally, potential treatment strategies, important knowledge gaps and future directions regarding the right side in HFpEF are discussed.
Collapse
Affiliation(s)
- Thomas M Gorter
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Medical School Hannover, Hannover, Germany
| | - Barry A Borlaug
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jelena Celutkiene
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Andrew J S Coats
- Monash University, Clayton Campus, Melbourne, Victoria 3800, Australia.,University of Warwick, Kirby Corner Road, Coventry CV4 8UW, UK
| | - Marisa G Crespo-Leiro
- Advanced Heart Failure and Heart Transplant Unit, Servicio de Cardiologia-CIBERCV, Complejo Hospitalario Universitario A Coruña (CHUAC), Instituto Investigación Biomedica A Coruña (INIBIC), Universidad da Coruña (UDC), La Coruña, Spain
| | - Marco Guazzi
- Heart Failure Unit, University of Milan, IRCCS Policlinico San Donato, Milan, Italy
| | - Veli-Pekka Harjola
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | - Stephane Heymans
- Department of Cardiology, CARIM, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | - Mitja Lainscak
- Department of Internal Medicine, General Hospital Murska Sobota, Murska Sobota, Slovenia
| | - Carolyn S P Lam
- Department of Cardiology, National Heart Center Singapore, Singapore Duke-NUS Graduate Medical School, Singapore
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet and Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Alexander R Lyon
- National Heart and Lung Institute, Imperial College London and Royal Brompton Hospital, London, UK
| | - Alexandre Mebazaa
- Department of Anesthesiology and Critical Care, APHP - Saint Louis Lariboisière University Hospitals, University Paris Diderot, Paris, France
| | - Christian Mueller
- Department of Cardiology, Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Walter J Paulus
- Department of Cardiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Berlin, Germany, and Department of Internal Medicine Cardiology, German Heart Center Berlin, DZHK (German Center for Cardiovascular Research) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Massimo F Piepoli
- Heart Failure Unit, Cardiac Department, G. da Saliceto Hospital, Piacenza, Italy
| | - Frank Ruschitzka
- Clinic for Cardiology, University Hospital Zurich, Zürich, Switzerland
| | - Frans H Rutten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Petar M Seferovic
- Cardiology Department, Clinical Centre Serbia, Medical School, Belgrade, Serbia
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Sanjiv J Shah
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Rolf Wachter
- Clinic and Policlinic for Cardiology, University Hospital Leipzig, Leipzig, Germany and German Cardiovascular Research Center, partner site Göttingen
| | - Carsten Tschöpe
- Department of Internal Medicine and Cardiology, Charité University Medicine, Berlin, Germany, and Department of Internal Medicine Cardiology, German Heart Center Berlin, DZHK (German Center for Cardiovascular Research) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
49
|
Liu Z, Lin Z, Chen S, Wang L, Xian S. Rapid Screening of Potential Phosphodiesterase Inhibitors from the Roots of Ilex pubescens Hook. et Arn. Using a Combination of Ultrafiltration and LC-MS. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2017; 2017:2749643. [PMID: 28424739 PMCID: PMC5382325 DOI: 10.1155/2017/2749643] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/15/2017] [Accepted: 03/12/2017] [Indexed: 11/30/2022]
Abstract
The cyclic nucleotide phosphodiesterase (PDE) plays an important role in regulating the levels of second messenger molecules cAMP and cGMP. Various PDE inhibitors have been successfully developed into drugs for targeted diseases. In addition, PDE inhibitors can also be found in different foods and natural medicines. In this study, ultrafiltration liquid chromatography-diode-array detector-electrospray ionization-ion-trap-time-of-flight-mass spectrometry (ultrafiltration LC-DAD-ESI-IT-TOF-MS) was applied to screen PDE inhibitors from the roots of Ilex pubescens Hook. et Arn. As a result, 11 major compounds were identified in I. pubescens roots, with nine compounds as potential PDE inhibitors, among which five were further confirmed to be active against PDEI and PDE5A dose-dependently in vitro, with ilexsaponin A1 and ilexsaponin B2 being the strongest. HPLC quantification of these bioactive compounds suggested that they are major components in the plant. The results demonstrate that ultrafiltration LC-DAD-ESI-IT-TOF-MS is an efficient method for rapid screening of PDE inhibitors from natural medicines.
Collapse
Affiliation(s)
- Zichen Liu
- Guangzhou Key Laboratory of Chinese Medicine Prevention and Treatment of Chronic Heart Failure, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| | - Zongtao Lin
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 28163, USA
| | - Shizhong Chen
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lingjun Wang
- Guangzhou Key Laboratory of Chinese Medicine Prevention and Treatment of Chronic Heart Failure, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| | - Shaoxiang Xian
- Guangzhou Key Laboratory of Chinese Medicine Prevention and Treatment of Chronic Heart Failure, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, China
| |
Collapse
|
50
|
Brescia M, Zaccolo M. Modulation of Compartmentalised Cyclic Nucleotide Signalling via Local Inhibition of Phosphodiesterase Activity. Int J Mol Sci 2016; 17:E1672. [PMID: 27706091 PMCID: PMC5085705 DOI: 10.3390/ijms17101672] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are the only enzymes that degrade the cyclic nucleotides cAMP and cGMP, and play a key role in modulating the amplitude and duration of the signal delivered by these two key intracellular second messengers. Defects in cyclic nucleotide signalling are known to be involved in several pathologies. As a consequence, PDEs have long been recognized as potential drug targets, and they have been the focus of intense research for the development of therapeutic agents. A number of PDE inhibitors are currently available for the treatment of disease, including obstructive pulmonary disease, erectile dysfunction, and heart failure. However, the performance of these drugs is not always satisfactory, due to a lack of PDE-isoform specificity and their consequent adverse side effects. Recent advances in our understanding of compartmentalised cyclic nucleotide signalling and the role of PDEs in local regulation of cAMP and cGMP signals offers the opportunity for the development of novel strategies for therapeutic intervention that may overcome the current limitation of conventional PDE inhibitors.
Collapse
Affiliation(s)
- Marcella Brescia
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3TP, UK.
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3TP, UK.
| |
Collapse
|