1
|
Zaccolo M, Kovanich D. Nanodomain cAMP signalling in cardiac pathophysiology: potential for developing targeted therapeutic interventions. Physiol Rev 2024:10.1152/physrev.00013.2024. [PMID: 39115424 PMCID: PMC7617275 DOI: 10.1152/physrev.00013.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
3', 5'-cyclic adenosine monophosphate (cAMP) mediates the effects of sympathetic stimulation on the rate and strength of cardiac contraction. Beyond this pivotal role, in cardiac myocytes cAMP also orchestrates a diverse array of reactions to various stimuli. To ensure specificity of response, the cAMP signaling pathway is intricately organized into multiple, spatially confined, subcellular domains, each governing a distinct cellular function. In this review, we describe the molecular components of the cAMP signalling pathway, how they organized are inside the intracellular space and how they achieve exquisite regulation of signalling within nanometer-size domains. We delineate the key experimental findings that lead to the current model of compartmentalised cAMP signaling and we offer an overview of our present understanding of how cAMP nanodomains are structured and regulated within cardiac myocytes. Furthermore, we discuss how compartmentalized cAMP signaling is affected in cardiac disease and consider the potential therapeutic opportunities arising from understanding such organization. By exploiting the nuances of compartmentalized cAMP signaling, novel and more effective therapeutic strategies for managing cardiac conditions may emerge. Finally, we highlight the unresolved questions and hurdles that must be addressed to translate these insights into interventions that may benefit patients.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Duangnapa Kovanich
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
2
|
Barthou A, Kamel R, Leroy J, Vandecasteele G, Fischmeister R. [Cyclic nucleotide phosphodiesterases: therapeutic targets in cardiac hypertrophy and failure]. Med Sci (Paris) 2024; 40:534-543. [PMID: 38986098 DOI: 10.1051/medsci/2024083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple isoforms of PDEs with different enzymatic properties and subcellular locally regulate cyclic nucleotide levels and associated cellular functions. This organisation is severely disrupted during hypertrophy and heart failure (HF), which may contribute to disease progression. Clinically, PDE inhibition has been seen as a promising approach to compensate for the catecholamine desensitisation that accompanies heart failure. Although PDE3 inhibitors such as milrinone or enoximone can be used clinically to improve systolic function and relieve the symptoms of acute CHF, their chronic use has proved detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as potential new targets for the treatment of HF, each with a unique role in local cyclic nucleotide signalling pathways. In this review, we describe cAMP and cGMP signalling in cardiomyocytes and present the different families of PDEs expressed in the heart and their modifications in pathological cardiac hypertrophy and HF. We also review results from preclinical models and clinical data indicating the use of specific PDE inhibitors or activators that may have therapeutic potential in CI.
Collapse
Affiliation(s)
| | - Rima Kamel
- Université Paris-Saclay, Inserm UMR-S 1180, Orsay, France
| | - Jérôme Leroy
- Université Paris-Saclay, Inserm UMR-S 1180, Orsay, France
| | | | | |
Collapse
|
3
|
Xie L, Huang B, Zhao X, Zhu N. Exploring the mechanisms underlying effects of bisphenol a on cardiovascular disease by network toxicology and molecular docking. Heliyon 2024; 10:e31473. [PMID: 38813174 PMCID: PMC11133888 DOI: 10.1016/j.heliyon.2024.e31473] [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: 01/22/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Background Globally, cardiovascular disease (CVD) has emerged as a leading cause of mortality. Bisphenol A (BPA), recognized as one of the most prevalent and widely distributed endocrine-disrupting chemicals (EDCs), has been consistently linked to the progression of CVD. This research centers on unraveling the molecular mechanisms responsible for the toxic effects of BPA exposure on CVD. Key targets and pathways involved in action of BPA on CVD were investigated by network toxicology. Binding abilities of BPA to core targets were evaluated by molecular docking. Methods and results Based on information retrieved from ChEMBL, DrugBank, and OMIM databases, a total of 27 potential targets were found to be associated with the influence of BPA on CVD. Furthermore, the STRING and Cytoscape software were employed to identify three central genes-ESR1, PPARG, and PTGS2-and to construct both the protein-protein interaction network and an interaction diagram of potential targets. Gene ontology (GO) and KEGG (Kyoto Encyclopedia of Genes and Genomes, KEGG) pathway enrichment analyses via WebGestalt revealed key biological processes (BP), cellular components (CC), molecular functions (MF), and pathways, such as the calcium signaling pathway, inflammatory mediator regulation of TRP channels, gap junction, adrenergic signaling in cardiomyocytes, cGMP-PKG signaling pathway, and cAMP signaling pathway, predominantly involved in BPA-induced CVD toxicity. By using molecular docking investigations, it proved that BPA binds to ESR1, PPARG, and PTGS2 steadily and strongly. Conclusion This study not only establishes a theoretical framework for understanding the molecular toxicity mechanism of BPA in cardiovascular disease (CVD) but also introduces an innovative network toxicology approach to methodically investigate the influence of environmental contaminants on CVD. This methodology sets the stage for drug discovery efforts targeting CVD linked to exposure to endocrine-disrupting chemicals (EDCs).
Collapse
Affiliation(s)
- Lina Xie
- Department of Neurosurgery, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China
| | - Xuyong Zhao
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Ning Zhu
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhang Y, Chen S, Luo L, Greenly S, Shi H, Xu JJ, Yan C. Role of cAMP in Cardiomyocyte Viability: Beneficial or Detrimental? Circ Res 2023; 133:902-923. [PMID: 37850368 PMCID: PMC10807647 DOI: 10.1161/circresaha.123.322652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND 3', 5'-cyclic AMP (cAMP) regulates numerous cardiac functions. Various hormones and neurotransmitters elevate intracellular cAMP (i[cAMP]) in cardiomyocytes through activating GsPCRs (stimulatory-G-protein-coupled-receptors) and membrane-bound ACs (adenylyl cyclases). Increasing evidence has indicated that stimulating different GsPCRs and ACs exhibits distinct, even opposite effects, on cardiomyocyte viability. However, the underlying mechanisms are not fully understood. METHODS We used molecular and pharmacological approaches to investigate how different GsPCR/cAMP signaling differentially regulate cardiomyocyte viability with in vitro, ex vivo, and in vivo models. RESULTS For prodeath GsPCRs, we explored β1AR (beta1-adrenergic receptor) and H2R (histamine-H2-receptor). We found that their prodeath effects were similarly dependent on AC5 activation, ATP release to the extracellular space via PANX1 (pannexin-1) channel, and extracellular ATP (e[ATP])-mediated signaling involving in P2X7R (P2X purinoceptor 7) and CaMKII (Ca2+/calmodulin-dependent protein kinase II). PANX1 phosphorylation at Serine 206 by cAMP-dependent-PKA (protein-kinase-A) promoted PANX1 activation, which was critical in β1AR- or H2R-induced cardiomyocyte death in vitro and in vivo. β1AR or H2R was localized proximately to PANX1, which permits ATP release. For prosurvival GsPCRs, we explored adenosine-A2-receptor (A2R), CGRPR (calcitonin-gene-related-peptide-receptor), and RXFP1 (relaxin-family peptide-receptor 1). Their prosurvival effects were dependent on AC6 activation, cAMP efflux via MRP4 (multidrug resistance protein 4), extracellular cAMP metabolism to adenosine (e[cAMP]-to-e[ADO]), and e[ADO]-mediated signaling. A2R, CGRPR, or RXFP1 was localized proximately to MRP4, which enables cAMP efflux. Interestingly, exogenously increasing e[cAMP] levels by membrane-impermeable cAMP protected against cardiomyocyte death in vitro and in ex vivo and in vivo mouse hearts with ischemia-reperfusion injuries. CONCLUSIONS Our findings indicate that the functional diversity of different GsPCRs in cardiomyocyte viability could be achieved by their ability to form unique signaling complexes (signalosomes) that determine the fate of cAMP: either stimulate ATP release by activating PKA or directly efflux to be e[cAMP].
Collapse
Affiliation(s)
- Yishuai Zhang
- Aab Cardiovascular Research Institute, Department of Medicine
| | - Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine
| | - Lingfeng Luo
- Aab Cardiovascular Research Institute, Department of Medicine
- Department of Biochemistry and Biophysics
| | - Sarah Greenly
- Aab Cardiovascular Research Institute, Department of Medicine
| | - Hangchuan Shi
- Department of Clinical and Translational Research
- Department of Public Health Sciences; University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | | | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine
| |
Collapse
|
7
|
Chen S, Chen J, Du W, Mickelsen DM, Shi H, Yu H, Kumar S, Yan C. PDE10A Inactivation Prevents Doxorubicin-Induced Cardiotoxicity and Tumor Growth. Circ Res 2023; 133:138-157. [PMID: 37232184 PMCID: PMC10428174 DOI: 10.1161/circresaha.122.322264] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Cyclic nucleotides play critical roles in cardiovascular biology and disease. PDE10A (phosphodiesterase 10A) is able to hydrolyze both cAMP and cGMP. PDE10A expression is induced in various human tumor cell lines, and PDE10A inhibition suppresses tumor cell growth. Chemotherapy drug such as doxorubicin (DOX) is widely used in chemotherapy. However, cardiotoxicity of DOX remains to be a serious clinical complication. In the current study, we aim to determine the role of PDE10A and the effect of PDE10A inhibition on cancer growth and cardiotoxicity induced by DOX. METHODS We used global PDE10A knockout (KO) mice and PDE10A inhibitor TP-10 to block PDE10A function. DOX-induced cardiotoxicity was evaluated in C57Bl/6J mice and nude mice with implanted ovarian cancer xenografts. Isolated adult mouse cardiomyocytes and a human ovarian cancer cell line were used for in vitro functional and mechanistic studies. RESULTS We found that PDE10A deficiency or inhibition alleviated DOX-induced myocardial atrophy, apoptosis, and dysfunction in C57Bl/6J mice. RNA sequencing study revealed a number of PDE10A-regulated signaling pathways involved in DOX-induced cardiotoxicity. PDE10A inhibition increased the death, decreased the proliferation, and potentiated the effect of DOX on various human cancer cells. Importantly, in nude mice with implanted ovarian cancer xenografts, PDE10A inhibition attenuated tumor growth while protecting DOX-induced cardiotoxicity. In isolated cardiomyocytes, PDE10A contributed to DOX-induced cardiomyocyte death via increasing Top2β (topoisomerase 2β) expression, mitochondrial dysfunction, and DNA damage by antagonizing cGMP/PKG (protein kinase G) signaling. PDE10A contributed to cardiomyocyte atrophy via potentiating FoxO3 (forkhead box O3) signaling via both cAMP/PKA (protein kinase A)- and cGMP/PKG-dependent signaling. CONCLUSIONS Taken together, our study elucidates a novel role for PDE10A in cardiotoxicity induced by DOX and cancer growth. Given that PDE10A has been already proven to be a safe drug target, PDE10A inhibition may represent a novel therapeutic strategy in cancer therapy, with effects preventing DOX-induced cardiotoxicity and simultaneously antagonizing cancer growth.
Collapse
Affiliation(s)
- Si Chen
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jiawei Chen
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Current position: Department of Cardiology, Ruijin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, P.R.China
- Current position: Institute of Cardiovascular Diseases, Shanghai Jiao-Tong University School of Medicine, Shanghai, P.R.China
| | - Wenting Du
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Current Position: Department of Geriatrics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Deanne M. Mickelsen
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Hangchuan Shi
- Department of Clinical and Translational Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Han Yu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sparsh Kumar
- Multidisciplinary Studies Center, University of Rochester, Rochester, NY, USA
| | - Chen Yan
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
8
|
Xu LX, Wang RX, Jiang JF, Yi GC, Chang JJ, He RL, Jiao HX, Zheng B, Gui LX, Lin JJ, Huang ZH, Lin MJ, Wu ZJ. TRPC6 promotes daunorubicin-induced mitochondrial fission and cell death in rat cardiomyocytes with the involvement of ERK1/2-DRP1 activation. Toxicol Appl Pharmacol 2023; 470:116547. [PMID: 37178933 DOI: 10.1016/j.taap.2023.116547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/02/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Daunorubicin (DNR-) induced cardiotoxicity seriously restricts its clinical application. Transient receptor potential cation channel subfamily C member 6 (TRPC6) is involved in multiple cardiovascular physiological and pathophysiological processes. However, the role of TRPC6 anthracycline-induced cardiotoxicity (AIC) remains unclear. Mitochondrial fragmentation greatly promotes AIC. TRPC6-mediated ERK1/2 activation has been shown to favor mitochondrial fission in dentate granule cells. The aim of the present study was to elucidate the effects of TRPC6 on daunorubicin- induced cardiotoxicity and identify the mechanisms associated with mitochondrial dynamics. The sparkling results showed that TRPC6 was upregulated in models in vitro and in vivo. TRPC6 knockdown protected cardiomyocytes from DNR-induced cell apoptosis and death. DNR largely facilitated mitochondrial fission, boosted mitochondrial membrane potential collapse and damaged debilitated mitochondrial respiratory function in H9c2 cells,these effects were accompanied by TRPC6 upregulation. siTRPC6 effectively inhibited these mitochondrial adverse aspects showing a positive unexposed effect on mitochondrial morphology and function. Concomitantly, ERK1/2-DRP1 which is related to mitochondrial fission was significantly activated with amplified phosphorylated forms in DNR-treated H9c2 cells. siTRPC6 effectively suppressedERK1/2-DPR1 over activation, hinting at a potential correlation between TRPC6 and ERK1/2-DRP1 by which mitochondrial dynamics are possibly modulated in AIC. TRPC6 knockdown also raised the Bcl-2/Bax ratio, which may help to block mitochondrial fragmentation-related functional impairment and apoptotic signaling. These findings suggested an essential role of TRPC6 in AIC by intensifying mitochondrial fission and cell death via ERK1/2-DPR1, which could be a potential therapeutic target for AIC.
Collapse
Affiliation(s)
- Li-Xia Xu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Rui-Xing Wang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China; Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Jian-Feng Jiang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Gao-Cheng Yi
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Jin-Jin Chang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Rui-Lan He
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China; Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Hai-Xia Jiao
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China; Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Bin Zheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, People's Republic of China; The School of Pharmacy, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Long-Xin Gui
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Jun-Jin Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Zhi-Hong Huang
- Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China
| | - Mo-Jun Lin
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China; Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China.
| | - Zhi-Juan Wu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China; Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, People's Republic of China.
| |
Collapse
|
9
|
Regulation of cardiac function by cAMP nanodomains. Biosci Rep 2023; 43:232544. [PMID: 36749130 PMCID: PMC9970827 DOI: 10.1042/bsr20220953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/08/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a diffusible intracellular second messenger that plays a key role in the regulation of cardiac function. In response to the release of catecholamines from sympathetic terminals, cAMP modulates heart rate and the strength of contraction and ease of relaxation of each heartbeat. At the same time, cAMP is involved in the response to a multitude of other hormones and neurotransmitters. A sophisticated network of regulatory mechanisms controls the temporal and spatial propagation of cAMP, resulting in the generation of signaling nanodomains that enable the second messenger to match each extracellular stimulus with the appropriate cellular response. Multiple proteins contribute to this spatiotemporal regulation, including the cAMP-hydrolyzing phosphodiesterases (PDEs). By breaking down cAMP to a different extent at different locations, these enzymes generate subcellular cAMP gradients. As a result, only a subset of the downstream effectors is activated and a specific response is executed. Dysregulation of cAMP compartmentalization has been observed in cardiovascular diseases, highlighting the importance of appropriate control of local cAMP signaling. Current research is unveiling the molecular organization underpinning cAMP compartmentalization, providing original insight into the physiology of cardiac myocytes and the alteration associated with disease, with the potential to uncover novel therapeutic targets. Here, we present an overview of the mechanisms that are currently understood to be involved in generating cAMP nanodomains and we highlight the questions that remain to be answered.
Collapse
|
10
|
Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiac hypertrophy and heart failure. Nat Rev Cardiol 2023; 20:90-108. [PMID: 36050457 DOI: 10.1038/s41569-022-00756-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) modulate the neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple PDE isozymes with different enzymatic properties and subcellular localization regulate local pools of cyclic nucleotides and specific functions. This organization is heavily perturbed during cardiac hypertrophy and heart failure (HF), which can contribute to disease progression. Clinically, PDE inhibition has been considered a promising approach to compensate for the catecholamine desensitization that accompanies HF. Although PDE3 inhibitors, such as milrinone or enoximone, have been used clinically to improve systolic function and alleviate the symptoms of acute HF, their chronic use has proved to be detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as new potential targets to treat HF, each having a unique role in local cyclic nucleotide signalling pathways. In this Review, we describe cAMP and cGMP signalling in cardiomyocytes and present the various PDE families expressed in the heart as well as their modifications in pathological cardiac hypertrophy and HF. We also appraise the evidence from preclinical models as well as clinical data pointing to the use of inhibitors or activators of specific PDEs that could have therapeutic potential in HF.
Collapse
|
11
|
Du X, Jiang Y, Li H, Zhang Q, Zhu X, Zhou L, Wang W, Zhang Y, Liu C, Niu Y, Chu C, Cai J, Chen R, Kan H. Traffic-related air pollution and genome-wide DNA methylation: A randomized, crossover trial. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 850:157968. [PMID: 35963411 DOI: 10.1016/j.scitotenv.2022.157968] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Traffic-related air pollution (TRAP) has been associated with changes in gene-specific DNA methylation. However, few studies have investigated impact of TRAP exposure on genome-wide DNA methylation in circulating blood of human. OBJECTIVE To explore the association between TRAP exposure and genome-wide DNA methylation. METHODS We conducted a randomized, crossover exposure trial among 35 healthy adults in Shanghai, China. All subjects were randomly allocated to a traffic-free park or a main road for consecutive 4 h, respectively. Blood genome-wide DNA methylation after each exposure session was measured by the Infinium Methylation EPIC BeadChip (850K). The differentially methylated CpGs loci associated with TRAP exposure were identified using linear mixed-effect model. RESULTS The average concentrations of traffic-related air pollutants including black carbon, ultrafine particles, carbon dioxide, and nitrogen dioxide were 2-3 times higher in the road compared to those in the park. Methylation levels of 68 CpG loci were significantly changed (false discovery rate < 0.05) following TRAP exposure, among which 49 were hypermethylated and 19 were hypomethylated. The annotated genes based on the differential CpGs loci were related to pathways in cardiovascular signaling, cytokine signaling, immune response, nervous system signaling, and metabolism. CONCLUSIONS We found that TRAP exposure was associated with DNA methylation in dozens of genes concerning cardiometabolic health. This trial for the first-time profiled genome-wide methylation changes induced by TRAP exposure using the 850K assay, providing epigenetic insights in understanding the cardiometabolic effects of TRAP exposure.
Collapse
Affiliation(s)
- Xihao Du
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Yixuan Jiang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Huichu Li
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Qingli Zhang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Xinlei Zhu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Lu Zhou
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Weidong Wang
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA, USA
| | - Yang Zhang
- Department of Systems Biology for Medicine and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cong Liu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Yue Niu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Chen Chu
- Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
| | - Jing Cai
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China.
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China; Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China
| |
Collapse
|
12
|
Wang C, Taskinen JH, Segersvärd H, Immonen K, Kosonen R, Tolva JM, Mäyränpää MI, Kovanen PT, Olkkonen VM, Sinisalo J, Laine M, Tikkanen I, Lakkisto P. Alterations of Cardiac Protein Kinases in Cyclic Nucleotide-Dependent Signaling Pathways in Human Ischemic Heart Failure. Front Cardiovasc Med 2022; 9:919355. [PMID: 35783854 PMCID: PMC9247256 DOI: 10.3389/fcvm.2022.919355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives Impaired protein kinase signaling is a hallmark of ischemic heart disease (IHD). Inadequate understanding of the pathological mechanisms limits the development of therapeutic approaches. We aimed to identify the key cardiac kinases and signaling pathways in patients with IHD with an effort to discover potential therapeutic strategies. Methods Cardiac kinase activity in IHD left ventricle (LV) and the related signaling pathways were investigated by kinomics, transcriptomics, proteomics, and integrated multi-omics approach. Results Protein kinase A (PKA) and protein kinase G (PKG) ranked on top in the activity shift among the cardiac kinases. In the IHD LVs, PKA activity decreased markedly compared with that of controls (62% reduction, p = 0.0034), whereas PKG activity remained stable, although the amount of PKG protein increased remarkably (65%, p = 0.003). mRNA levels of adenylate cyclases (ADCY 1, 3, 5, 9) and cAMP-hydrolysing phosphodiesterases (PDE4A, PDE4D) decreased significantly, although no statistically significant alterations were observed in that of PKGs (PRKG1 and PRKG2) and guanylate cyclases (GUCYs). The gene expression of natriuretic peptide CNP decreased remarkably, whereas those of BNP, ANP, and neprilysin increased significantly in the IHD LVs. Proteomics analysis revealed a significant reduction in protein levels of “Energy metabolism” and “Muscle contraction” in the patients. Multi-omics integration highlighted intracellular signaling by second messengers as the top enriched Reactome pathway. Conclusion The deficiency in cAMP/PKA signaling pathway is strongly implicated in the pathogenesis of IHD. Natriuretic peptide CNP could be a potential therapeutic target for the modulation of cGMP/PKG signaling.
Collapse
Affiliation(s)
- Chunguang Wang
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- *Correspondence: Chunguang Wang
| | - Juuso H. Taskinen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Heli Segersvärd
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Katariina Immonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Riikka Kosonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Johanna M. Tolva
- Transplantation Laboratory, Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Laine
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Tikkanen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Päivi Lakkisto
| |
Collapse
|
13
|
Numata G, Takimoto E. Cyclic GMP and PKG Signaling in Heart Failure. Front Pharmacol 2022; 13:792798. [PMID: 35479330 PMCID: PMC9036358 DOI: 10.3389/fphar.2022.792798] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP), produced by guanylate cyclase (GC), activates protein kinase G (PKG) and regulates cardiac remodeling. cGMP/PKG signal is activated by two intrinsic pathways: nitric oxide (NO)-soluble GC and natriuretic peptide (NP)-particulate GC (pGC) pathways. Activation of these pathways has emerged as a potent therapeutic strategy to treat patients with heart failure, given cGMP-PKG signaling is impaired in heart failure with reduced ejection fraction (HFrEF) and preserved ejection fraction (HFpEF). Large scale clinical trials in patients with HFrEF have shown positive results with agents that activate cGMP-PKG pathways. In patients with HFpEF, however, benefits were observed only in a subgroup of patients. Further investigation for cGMP-PKG pathway is needed to develop better targeting strategies for HFpEF. This review outlines cGMP-PKG pathway and its modulation in heart failure.
Collapse
Affiliation(s)
- Genri Numata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo Hospital, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
14
|
Phosphodiesterase-1 in the cardiovascular system. Cell Signal 2022; 92:110251. [DOI: 10.1016/j.cellsig.2022.110251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 11/18/2022]
|
15
|
Chen DS, Yan J, Yang PZ. Cardiomyocyte Atrophy, an Underestimated Contributor in Doxorubicin-Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:812578. [PMID: 35282350 PMCID: PMC8913904 DOI: 10.3389/fcvm.2022.812578] [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: 11/10/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022] Open
Abstract
Left ventricular (LV) mass loss is prevalent in doxorubicin (DOX)-induced cardiotoxicity and is responsible for the progressive decline of cardiac function. Comparing with the well-studied role of cell death, the part of cardiomyocyte atrophy (CMA) playing in the LV mass loss is underestimated and the knowledge of the underlying mechanism is still limited. In this review, we summarized the recent advances in the DOX-induced CMA. We found that the CMA caused by DOX is associated with the upregulation of FOXOs and “atrogenes,” the activation of transient receptor potential canonical 3-NADPH oxidase 2 (TRPC3-Nox2) axis, and the suppression of IGF-1-PI3K signaling pathway. The imbalance of anabolic and catabolic process may be the common final pathway of these mechanisms. At last, we provided some strategies that have been demonstrated to alleviate the DOX-induced CMA in animal models.
Collapse
Affiliation(s)
- De-Shu Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- Jing Yan
| | - Ping-Zhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- *Correspondence: Ping-Zhen Yang
| |
Collapse
|
16
|
Muller GK, Song J, Jani V, Wu Y, Liu T, Jeffreys WPD, O’Rourke B, Anderson ME, Kass DA. PDE1 Inhibition Modulates Ca v1.2 Channel to Stimulate Cardiomyocyte Contraction. Circ Res 2021; 129:872-886. [PMID: 34521216 PMCID: PMC8553000 DOI: 10.1161/circresaha.121.319828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Grace K Muller
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Joy Song
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vivek Jani
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yuejin Wu
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ting Liu
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - William PD Jeffreys
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Brian O’Rourke
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Graduate Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Departments of Pharmacology and Molecular Sciences and Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mark E Anderson
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Graduate Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - David A Kass
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Graduate Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Departments of Pharmacology and Molecular Sciences and Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
17
|
Che Y, Tian Y, Chen R, Xia L, Liu F, Su Z. IL-22 ameliorated cardiomyocyte apoptosis in cardiac ischemia/reperfusion injury by blocking mitochondrial membrane potential decrease, inhibiting ROS and cytochrome C. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166171. [PMID: 34015450 DOI: 10.1016/j.bbadis.2021.166171] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 01/29/2023]
Abstract
Irreversible cardiomyocyte death is one of the main reasons of heart failure following cardiac injury. Therefore, controlling cardiomyocyte death is an effective method to delay the progression of cardiac disease after injury. IL-22 plays critical roles in tissue homeostasis and repair, and has become an important bridge between the immune system and specific tissues or organs. However, whether IL-22 can prevent of cardiomyocyte apoptosis from cardiac injury remains unclear. Therefore, the present work would address the above question. Our results showed that, in vitro, IL-22 prevented cardiomyocyte apoptosis induced by Angiotensin II via enhancing the activity of SOD, blocking the decrease of mitochondrial membrane potential, inhibiting ROS production and release of cytochrome C. The similar results were also found in vivo and patients. Our results shed a light on the therapy of cardiac injury.
Collapse
Affiliation(s)
- Yang Che
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Yu Tian
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Rong Chen
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Lin Xia
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Fang Liu
- International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
18
|
Gilotra NA, DeVore AD, Povsic TJ, Hays AG, Hahn VS, Agunbiade TA, DeLong A, Satlin A, Chen R, Davis R, Kass DA. Acute Hemodynamic Effects and Tolerability of Phosphodiesterase-1 Inhibition With ITI-214 in Human Systolic Heart Failure. Circ Heart Fail 2021; 14:e008236. [PMID: 34461742 DOI: 10.1161/circheartfailure.120.008236] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND PDE1 (phosphodiesterase type 1) hydrolyzes cyclic adenosine and guanosine monophosphate. ITI-214 is a highly selective PDE1 inhibitor that induces arterial vasodilation and positive inotropy in larger mammals. Here, we assessed pharmacokinetics, hemodynamics, and tolerability of single-dose ITI-214 in humans with stable heart failure with reduced ejection fraction. METHODS Patients with heart failure with reduced ejection fraction were randomized 3:1 to 10, 30, or 90 mg ITI-214 single oral dose or placebo (n=9/group). Vital signs and electrocardiography were monitored predose to 5 hours postdose and transthoracic echoDoppler cardiography predose and 2-hours postdose. RESULTS Patient age averaged 54 years; 42% female, and 60% Black. Mean systolic blood pressure decreased 3 to 8 mm Hg (P<0.001) and heart rate increased 5 to 9 bpm (P≤0.001 for 10, 30 mg doses, RM-ANCOVA). After 4 hours, neither blood pressure or heart rate significantly differed among cohorts (supine or standing). ITI-214 increased mean left ventricular power index, a relatively load-insensitive inotropic index, by 0.143 Watts/mL2·104 (P=0.03, a +41% rise; 5-71 CI) and cardiac output by 0.83 L/min (P=0.002, +31%, 13-49 CI) both at the 30 mg dose. Systemic vascular resistance declined with 30 mg (-564 dynes·s/cm-5, P<0.001) and 90 mg (-370, P=0.016). Diastolic changes were minimal, and no parameters were significantly altered with placebo. ITI-214 was well-tolerated. Five patients had mild-moderate hypotension or orthostatic hypotension recorded adverse events. There were no significant changes in arrhythmia outcome and no serious adverse events. CONCLUSIONS Single-dose ITI-214 is well-tolerated and confers inodilator effects in humans with heart failure with reduced ejection fraction. Further investigations of its therapeutic utility are warranted. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03387215.
Collapse
Affiliation(s)
- Nisha A Gilotra
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (N.A.G., A.G.H., V.S.H., T.A.A., D.A.K.)
| | - Adam D DeVore
- Duke University School of Medicine, Durham, NC (A.D.D.)
| | | | - Allison G Hays
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (N.A.G., A.G.H., V.S.H., T.A.A., D.A.K.)
| | - Virginia S Hahn
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (N.A.G., A.G.H., V.S.H., T.A.A., D.A.K.)
| | - Tolu A Agunbiade
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (N.A.G., A.G.H., V.S.H., T.A.A., D.A.K.)
| | - Allison DeLong
- Duke Clinical Research Institute, Durham, NC (T.J.P., A.D.)
| | - Andrew Satlin
- Intra-Cellular Therapies, Inc, New York, NY (A.S., R.C., R.D.)
| | - Richard Chen
- Intra-Cellular Therapies, Inc, New York, NY (A.S., R.C., R.D.)
| | - Robert Davis
- Intra-Cellular Therapies, Inc, New York, NY (A.S., R.C., R.D.)
| | - David A Kass
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (N.A.G., A.G.H., V.S.H., T.A.A., D.A.K.)
| |
Collapse
|
19
|
Samidurai A, Xi L, Das A, Iness AN, Vigneshwar NG, Li PL, Singla DK, Muniyan S, Batra SK, Kukreja RC. Role of phosphodiesterase 1 in the pathophysiology of diseases and potential therapeutic opportunities. Pharmacol Ther 2021; 226:107858. [PMID: 33895190 DOI: 10.1016/j.pharmthera.2021.107858] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/17/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are superfamily of enzymes that regulate the spatial and temporal relationship of second messenger signaling in the cellular system. Among the 11 different families of PDEs, phosphodiesterase 1 (PDE1) sub-family of enzymes hydrolyze both 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) in a mutually competitive manner. The catalytic activity of PDE1 is stimulated by their binding to Ca2+/calmodulin (CaM), resulting in the integration of Ca2+ and cyclic nucleotide-mediated signaling in various diseases. The PDE1 family includes three subtypes, PDE1A, PDE1B and PDE1C, which differ for their relative affinities for cAMP and cGMP. These isoforms are differentially expressed throughout the body, including the cardiovascular, central nervous system and other organs. Thus, PDE1 enzymes play a critical role in the pathophysiology of diseases through the fundamental regulation of cAMP and cGMP signaling. This comprehensive review provides the current research on PDE1 and its potential utility as a therapeutic target in diseases including the cardiovascular, pulmonary, metabolic, neurocognitive, renal, cancers and possibly others.
Collapse
Affiliation(s)
- Arun Samidurai
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Lei Xi
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Anindita Das
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Audra N Iness
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Navin G Vigneshwar
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298-0204, USA.
| |
Collapse
|
20
|
Brzezinska P, Simpson NJ, Hubert F, Jacobs AN, Umana MB, MacKeil JL, Burke-Kleinman J, Payne DM, Ferguson AV, Maurice DH. Phosphodiesterase 1C integrates store-operated calcium entry and cAMP signaling in leading-edge protrusions of migrating human arterial myocytes. J Biol Chem 2021; 296:100606. [PMID: 33789162 PMCID: PMC8095186 DOI: 10.1016/j.jbc.2021.100606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 01/17/2023] Open
Abstract
In addition to maintaining cellular ER Ca2+ stores, store-operated Ca2+ entry (SOCE) regulates several Ca2+-sensitive cellular enzymes, including certain adenylyl cyclases (ADCYs), enzymes that synthesize the secondary messenger cyclic AMP (cAMP). Ca2+, acting with calmodulin, can also increase the activity of PDE1-family phosphodiesterases (PDEs), which cleave the phosphodiester bond of cAMP. Surprisingly, SOCE-regulated cAMP signaling has not been studied in cells expressing both Ca2+-sensitive enzymes. Here, we report that depletion of ER Ca2+ activates PDE1C in human arterial smooth muscle cells (HASMCs). Inhibiting the activation of PDE1C reduced the magnitude of both SOCE and subsequent Ca2+/calmodulin–mediated activation of ADCY8 in these cells. Because inhibiting or silencing Ca2+-insensitive PDEs had no such effects, these data identify PDE1C-mediated hydrolysis of cAMP as a novel and important link between SOCE and its activation of ADCY8. Functionally, we showed that PDE1C regulated the formation of leading-edge protrusions in HASMCs, a critical early event in cell migration. Indeed, we found that PDE1C populated the tips of newly forming leading-edge protrusions in polarized HASMCs, and co-localized with ADCY8, the Ca2+ release activated Ca2+ channel subunit, Orai1, the cAMP-effector, protein kinase A, and an A-kinase anchoring protein, AKAP79. Because this polarization could allow PDE1C to control cAMP signaling in a hyper-localized manner, we suggest that PDE1C-selective therapeutic agents could offer increased spatial specificity in HASMCs over agents that regulate cAMP globally in cells. Similarly, such agents could also prove useful in regulating crosstalk between Ca2+/cAMP signaling in other cells in which dysregulated migration contributes to human pathology, including certain cancers.
Collapse
Affiliation(s)
- Paulina Brzezinska
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nicholas J Simpson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Fabien Hubert
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ariana N Jacobs
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - M Bibiana Umana
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jodi L MacKeil
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jonah Burke-Kleinman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Darrin M Payne
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | - Alastair V Ferguson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Donald H Maurice
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
21
|
Canonical transient receptor potential channels and their modulators: biology, pharmacology and therapeutic potentials. Arch Pharm Res 2021; 44:354-377. [PMID: 33763843 PMCID: PMC7989688 DOI: 10.1007/s12272-021-01319-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
Canonical transient receptor potential channels (TRPCs) are nonselective, high calcium permeability cationic channels. The TRPCs family includes TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7. These channels are widely expressed in the cardiovascular and nervous systems and exist in many other human tissues and cell types, playing several crucial roles in the human physiological and pathological processes. Hence, the emergence of TRPCs modulators can help investigate these channels’ applications in health and disease. It is worth noting that the TRPCs subfamilies have structural and functional similarities, which presents a significant difficulty in screening and discovering of TRPCs modulators. In the past few years, only a limited number of selective modulators of TRPCs were detected; thus, additional research on more potent and more selective TRPCs modulators is needed. The present review focuses on the striking desired therapeutic effects of TRPCs modulators, which provides intel on the structural modification of TRPCs modulators and further pharmacological research. Importantly, TRPCs modulators can significantly facilitate future studies of TRPCs and TRPCs related diseases.
Collapse
|
22
|
Zhu JX, Ling W, Xue C, Zhou Z, Zhang YS, Yan C, Wu MP. Higenamine attenuates cardiac fibroblast abstract and fibrosis via inhibition of TGF-β1/Smad signaling. Eur J Pharmacol 2021; 900:174013. [PMID: 33766620 DOI: 10.1016/j.ejphar.2021.174013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/21/2021] [Accepted: 02/28/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE Higenamine (HG), is one of the main active components in many widely used Chinese herbs, and a common ingredient of health products in Europe and North America. Several groups, including our own, have previously shown the beneficial effects of HG against cardiomyocyte death during acute ischemic damage. However, the effect of HG on chronic cardiac remodeling, such as cardiac fibrosis, remains unknown. OBJECTIVE Herein, we aim to investigate the role of HG in cardiac fibrosis in vivo as well as its cellular and molecular mechanisms. METHODS AND RESULTS Chronic pressure overload with transverse aortic constriction (TAC) significantly increased cardiac hypertrophy, fibrosis, and cardiac dysfunction in mice, which were significantly attenuated by HG. Consistently, cardiac fibrosis induced by the chronic infusion of isoproterenol (ISO), was also significantly reduced by HG. Interestingly, our results showed that HG had no effect on adult mouse CM hypertrophy in vitro. However, HG suppressed the activation of cardiac fibroblasts (CFs) in vitro. Furthermore, TGF-β1-induced expression of ACTA2, a marker of fibroblast activation, was significantly suppressed by HG. Concomitantly, HG inhibited TGF-β1-induced phosphorylation of Smad2/3 in CFs. HG also reduced the expression of extracellular matrix molecules such as collagen I and collagen III. To our surprise, the inhibitory effect of HG on CFs activation was independent of the activation of the beta2 adrenergic receptor (β2-AR) that is known to mediate the effect of HG on antagonizing CMs apoptosis. CONCLUSION Our findings suggest that HG ameliorates pathological cardiac fibrosis and dysfunction at least partially by suppressing TGF-β1/Smad signaling and CFs activation.
Collapse
Affiliation(s)
- Jin-Xing Zhu
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang RD, Shanghai, 200071, China; Department of Emergency Intensive Care Unit, Traditional Chinese Hospital of LuAn, Anhui University of Traditional Chinese Medicine, 76 Renmin RD, LuAn Anhui, 237001, China
| | - Wang Ling
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang RD, Shanghai, 200071, China
| | - Chao Xue
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave Box CVRI, Rochester, NY, 14642, United States
| | - Zhen Zhou
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang RD, Shanghai, 200071, China
| | - Yi-Shuai Zhang
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave Box CVRI, Rochester, NY, 14642, United States
| | - Chen Yan
- Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave Box CVRI, Rochester, NY, 14642, United States
| | - Mei-Ping Wu
- Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang RD, Shanghai, 200071, China; Aab Cardiovascular Research Institute, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave Box CVRI, Rochester, NY, 14642, United States.
| |
Collapse
|
23
|
Chen S, Yan C. An update of cyclic nucleotide phosphodiesterase as a target for cardiac diseases. Expert Opin Drug Discov 2021; 16:183-196. [PMID: 32957823 PMCID: PMC7854486 DOI: 10.1080/17460441.2020.1821643] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cyclic nucleotides, cAMP, and cGMP, are important second messengers of intracellular signaling and play crucial roles in cardiovascular biology and diseases. Cyclic nucleotide phosphodiesterases (PDEs) control the duration, magnitude, and compartmentalization of cyclic nucleotide signaling by catalyzing the hydrolysis of cyclic nucleotides. Individual PDEs modulate distinct signaling pathways and biological functions in the cell, making it a potential therapeutic target for the treatment of different cardiovascular disorders. The clinical success of several PDE inhibitors has ignited continued interest in PDE inhibitors and in PDE-target therapeutic strategies. AREAS COVERED This review concentrates on recent research advances of different PDE isoforms with regard to their expression patterns and biological functions in the heart. The limitations of current research and future directions are then discussed. The current and future development of PDE inhibitors is also covered. EXPERT OPINION Despite the therapeutic success of several marketed PDE inhibitors, the use of PDE inhibitors can be limited by their side effects, lack of efficacy, and lack of isoform selectivity. Advances in our understanding of the mechanisms by which cellular functions are changed through PDEs may enable the development of new approaches to achieve effective and specific PDE inhibition for various cardiac therapies.
Collapse
Affiliation(s)
- Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
24
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
25
|
Abstract
Cyclic nucleotide phosphodiesterases comprise an 11-member superfamily yielding near 100 isoform variants that hydrolyze cAMP or cGMP to their respective 5'-monophosphate form. Each plays a role in compartmentalized cyclic nucleotide signaling, with varying selectivity for each substrate, and conveying cell and intracellular-specific localized control. This review focuses on the 5 phosphodiesterases (PDEs) expressed in the cardiac myocyte capable of hydrolyzing cGMP and that have been shown to play a role in cardiac physiological and pathological processes. PDE1, PDE2, and PDE3 catabolize cAMP as well, whereas PDE5 and PDE9 are cGMP selective. PDE3 and PDE5 are already in clinical use, the former for heart failure, and PDE1, PDE9, and PDE5 are all being actively studied for this indication in patients. Research in just the past few years has revealed many novel cardiac influences of each isoform, expanding the therapeutic potential from their selective pharmacological blockade or in some instances, activation. PDE1C inhibition was found to confer cell survival protection and enhance cardiac contractility, whereas PDE2 inhibition or activation induces beneficial effects in hypertrophied or failing hearts, respectively. PDE3 inhibition is already clinically used to treat acute decompensated heart failure, although toxicity has precluded its long-term use. However, newer approaches including isoform-specific allosteric modulation may change this. Finally, inhibition of PDE5A and PDE9A counter pathological remodeling of the heart and are both being pursued in clinical trials. Here, we discuss recent research advances in each of these PDEs, their impact on the myocardium, and cardiac therapeutic potential.
Collapse
|
26
|
Nadur NF, de Azevedo LL, Caruso L, Graebin CS, Lacerda RB, Kümmerle AE. The long and winding road of designing phosphodiesterase inhibitors for the treatment of heart failure. Eur J Med Chem 2020; 212:113123. [PMID: 33412421 DOI: 10.1016/j.ejmech.2020.113123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/14/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a superfamily of enzymes known to play a critical role in the indirect regulation of several intracellular metabolism pathways through the selective hydrolysis of the phosphodiester bonds of specific second messenger substrates such as cAMP (3',5'-cyclic adenosine monophosphate) and cGMP (3',5'-cyclic guanosine monophosphate), influencing the hypertrophy, contractility, apoptosis and fibroses in the cardiovascular system. The expression and/or activity of multiple PDEs is altered during heart failure (HF), which leads to changes in levels of cyclic nucleotides and function of cardiac muscle. Within the cardiovascular system, PDEs 1-5, 8 and 9 are expressed and are interesting targets for the HF treatment. In this comprehensive review we will present a briefly description of the biochemical importance of each cardiovascular related PDE to the HF, and cover almost all the "long and winding road" of designing and discovering ligands, hits, lead compounds, clinical candidates and drugs as PDE inhibitors in the last decade.
Collapse
Affiliation(s)
- Nathalia Fonseca Nadur
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Luciana Luiz de Azevedo
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Lucas Caruso
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Cedric Stephan Graebin
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Renata Barbosa Lacerda
- Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Arthur Eugen Kümmerle
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil.
| |
Collapse
|
27
|
Abstract
Heart failure (HF) is a common consequence of several cardiovascular diseases and is understood as a vicious cycle of cardiac and hemodynamic decline. The current inventory of treatments either alleviates the pathophysiological features (eg, cardiac dysfunction, neurohumoral activation, and ventricular remodeling) and/or targets any underlying pathologies (eg, hypertension and myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine-3',5'-monophosphate (cGMP), synthesized by nitric oxide (NO)- and natriuretic peptide (NP)-responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signaling, which can occur after GC deactivation and the upregulation of cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs), promotes cardiac dysfunction. In this study, we review the role that NO/cGMP and NP/cGMP signaling plays in HF. After considering disease etiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from preclinical models and patients that compromised cGMP signaling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined.
Collapse
|
28
|
Abstract
The cyclic nucleotides cyclic adenosine-3′,5′-monophosphate (cAMP) and cyclic guanosine-3′,5′-monophosphate (cGMP) maintain physiological cardiac contractility and integrity. Cyclic nucleotide–hydrolysing phosphodiesterases (PDEs) are the prime regulators of cAMP and cGMP signalling in the heart. During heart failure (HF), the expression and activity of multiple PDEs are altered, which disrupt cyclic nucleotide levels and promote cardiac dysfunction. Given that the morbidity and mortality associated with HF are extremely high, novel therapies are urgently needed. Herein, the role of PDEs in HF pathophysiology and their therapeutic potential is reviewed. Attention is given to PDEs 1–5, and other PDEs are briefly considered. After assessing the role of each PDE in cardiac physiology, the evidence from pre-clinical models and patients that altered PDE signalling contributes to the HF phenotype is examined. The potential of pharmacologically harnessing PDEs for therapeutic gain is considered.
Collapse
|
29
|
TRPC Channels in Cardiac Plasticity. Cells 2020; 9:cells9020454. [PMID: 32079284 PMCID: PMC7072762 DOI: 10.3390/cells9020454] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 01/21/2023] Open
Abstract
The heart flexibly changes its structure in response to changing environments and oxygen/nutrition demands of the body. Increased and decreased mechanical loading induces hypertrophy and atrophy of cardiomyocytes, respectively. In physiological conditions, these structural changes of the heart are reversible. However, chronic stresses such as hypertension or cancer cachexia cause irreversible remodeling of the heart, leading to heart failure. Accumulating evidence indicates that calcium dyshomeostasis and aberrant reactive oxygen species production cause pathological heart remodeling. Canonical transient receptor potential (TRPC) is a nonselective cation channel subfamily whose multimodal activation or modulation of channel activity play important roles in a plethora of cellular physiology. Roles of TRPC channels in cardiac physiology have been reported in pathological cardiac remodeling. In this review, we summarize recent findings regarding the importance of TRPC channels in flexible cardiac remodeling (i.e., cardiac plasticity) in response to environmental stresses and discuss questions that should be addressed in the near future.
Collapse
|
30
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
31
|
He Z, Xu X, Chen C, Li H, Wang DW. Adenosine 2A Receptor Activation Contributes to Ang II–Induced Aortic Remodeling by Promoting Macrophage Retention. Hypertension 2020; 75:119-130. [DOI: 10.1161/hypertensionaha.119.13709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The A
2A
R (adenosine 2A receptor) plays a crucial role in the pathophysiological process of cardiovascular diseases, yet its effect on aortic remodeling remains unclear. We observed elevated adenosine and A
2A
R levels following infusion of mice with Ang II (angiotensin II), suggesting a potential role for the adenosine-A
2A
R system in macrophage accumulation and subsequent aortic remodeling. The effects and mechanisms of A
2A
R on macrophage dynamics during aortic remodeling were further investigated using mice with macrophage knockout of A
2A
R and by transplantation of A
2A
R
−/−
bone marrow. We demonstrated that macrophage knockout of A
2A
R inhibited macrophage accumulation and subsequent aortic remodeling by inhibiting macrophage retention. This was shown to occur via promotion of macrophage emigration to the draining lymph node. These effects correlated with restoration of the expression and surface content of CCR7 (CC chemokine receptor 7). Consistently, A
2A
R
−/−
bone marrow transplantation relieved Ang II–induced aortic remodeling, macrophage retention, and CCR7 downregulation and internalization, all of which were rescued by A
2A
R
+
/
+
bone marrow transplantation. In addition, CCR7 antibody treatment blocked all the protective effects observed in A
2A
R-cKO mice, including attenuation of aortic remodeling and decreased macrophage retention. In in vitro studies, A
2A
R activation induced by Ang II suppressed macrophage migration to CCL19 (CC-chemokine ligand) 19 through downregulation and internalization of CCR7. In summary, A
2A
R activation contributes to Ang II–induced macrophage retention and subsequent aortic remodeling by inhibiting migration of macrophages to the draining lymph node through regulating CCR7 expression and internalization.
Collapse
Affiliation(s)
- Zuowen He
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Xizhen Xu
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Chen Chen
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Huaping Li
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| | - Dao Wen Wang
- From the Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
- Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiological Disorders, Wuhan, China (Z.H., X.X., C.C., H.L., D.W.W.)
| |
Collapse
|
32
|
Chen S, Zhang Y, Lighthouse JK, Mickelsen DM, Wu J, Yao P, Small EM, Yan C. A Novel Role of Cyclic Nucleotide Phosphodiesterase 10A in Pathological Cardiac Remodeling and Dysfunction. Circulation 2019; 141:217-233. [PMID: 31801360 DOI: 10.1161/circulationaha.119.042178] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Heart failure is a leading cause of death worldwide. Cyclic nucleotide phosphodiesterases (PDEs), through degradation of cyclic nucleotides, play critical roles in cardiovascular biology and disease. Our preliminary screening studies have revealed PDE10A upregulation in the diseased heart. However, the roles of PDE10A in cardiovascular biology and disease are largely uncharacterized. The current study is aimed to investigate the regulation and function of PDE10A in cardiac cells and in the progression of cardiac remodeling and dysfunction. METHODS We used isolated adult mouse cardiac myocytes and fibroblasts, as well as preclinical mouse models of hypertrophy and heart failure. The PDE10A selective inhibitor TP-10, and global PDE10A knock out mice were used. RESULTS We found that PDE10A expression remains relatively low in normal and exercised heart tissues. However, PDE10A is significantly upregulated in mouse and human failing hearts. In vitro, PDE10A deficiency or inhibiting PDE10A with selective inhibitor TP-10, attenuated cardiac myocyte pathological hypertrophy induced by Angiotensin II, phenylephrine, and isoproterenol, but did not affect cardiac myocyte physiological hypertrophy induced by IGF-1 (insulin-like growth factor 1). TP-10 also reduced TGF-β (transforming growth factor-β)-stimulated cardiac fibroblast activation, proliferation, migration and extracellular matrix synthesis. TP-10 treatment elevated both cAMP and cGMP levels in cardiac myocytes and cardiac fibroblasts, consistent with PDE10A as a cAMP/cGMP dual-specific PDE. In vivo, global PDE10A deficiency significantly attenuated myocardial hypertrophy, cardiac fibrosis, and dysfunction induced by chronic pressure overload via transverse aorta constriction or chronic neurohormonal stimulation via Angiotensin II infusion. Importantly, we demonstrated that the pharmacological effect of TP-10 is specifically through PDE10A inhibition. In addition, TP-10 is able to reverse pre-established cardiac hypertrophy and dysfunction. RNA-Sequencing and bioinformatics analysis further identified a PDE10A-regualted transcriptome involved in cardiac hypertrophy, fibrosis, and cardiomyopathy. CONCLUSIONS Taken together, our study elucidates a novel role for PDE10A in the regulation of pathological cardiac remodeling and development of heart failure. Given that PDE10A has been proven to be a safe drug target, PDE10A inhibition may represent a novel therapeutic strategy for preventing and treating cardiac diseases associated with cardiac remodeling.
Collapse
Affiliation(s)
- Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Pharmacology and Physiology (S.C.), University of Rochester School of Medicine and Dentistry, NY
| | - Yishuai Zhang
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Deanne M Mickelsen
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Jiangbin Wu
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Peng Yao
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Biochemistry and Biophysics (P.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Eric M Small
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine (S.C., Y.Z., J.K.L., D.M.M., J.W., P.Y., E.M.S., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
33
|
Bandleon S, Strunz PP, Pickel S, Tiapko O, Cellini A, Miranda-Laferte E, Eder-Negrin P. FKBP52 regulates TRPC3-dependent Ca 2+ signals and the hypertrophic growth of cardiomyocyte cultures. J Cell Sci 2019; 132:jcs.231506. [PMID: 31540954 DOI: 10.1242/jcs.231506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
The transient receptor potential (TRP; C-classical, TRPC) channel TRPC3 allows a cation (Na+/Ca2+) influx that is favored by the stimulation of Gq protein-coupled receptors (GPCRs). An enhanced TRPC3 activity is related to adverse effects, including pathological hypertrophy in chronic cardiac disease states. In the present study, we identified FK506-binding protein 52 (FKBP52, also known as FKBP4) as a novel interaction partner of TRPC3 in the heart. FKBP52 was recovered from a cardiac cDNA library by a C-terminal TRPC3 fragment (amino acids 742-848) in a yeast two-hybrid screen. Downregulation of FKBP52 promoted a TRPC3-dependent hypertrophic response in neonatal rat cardiomyocytes (NRCs). A similar effect was achieved by overexpressing peptidyl-prolyl isomerase (PPIase)-deficient FKBP52 mutants. Mechanistically, expression of the FKBP52 truncation mutants elevated TRPC3-mediated currents and Ca2+ fluxes, and the activation of calcineurin and the nuclear factor of activated T-cells in NRCs. Our data demonstrate that FKBP52 associates with TRPC3 via an as-yet-undescribed binding site in the C-terminus of TRPC3 and modulates TRPC3-dependent Ca2+ signals in a PPIase-dependent manner. This functional interaction might be crucial for limiting TRPC3-dependent signaling during chronic hypertrophic stimulation.
Collapse
Affiliation(s)
- Sandra Bandleon
- Comprehensive Heart Failure Center Wuerzburg, The Department of Internal Medicine I, University Hospital Wuerzburg, Am Schwarzenberg 15, 97078 Wuerzburg, Germany
| | - Patrick P Strunz
- Comprehensive Heart Failure Center Wuerzburg, The Department of Internal Medicine I, University Hospital Wuerzburg, Am Schwarzenberg 15, 97078 Wuerzburg, Germany
| | - Simone Pickel
- Institute of Physiology, University of Wuerzburg, Röntgenring 9, 97070 Wuerzburg, Germany
| | - Oleksandra Tiapko
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, 8010 Graz, Austria
| | - Antonella Cellini
- Comprehensive Heart Failure Center Wuerzburg, The Department of Internal Medicine I, University Hospital Wuerzburg, Am Schwarzenberg 15, 97078 Wuerzburg, Germany
| | - Erick Miranda-Laferte
- Institute of Physiology, University of Wuerzburg, Röntgenring 9, 97070 Wuerzburg, Germany
| | - Petra Eder-Negrin
- Comprehensive Heart Failure Center Wuerzburg, The Department of Internal Medicine I, University Hospital Wuerzburg, Am Schwarzenberg 15, 97078 Wuerzburg, Germany
| |
Collapse
|
34
|
Golshiri K, Ataei Ataabadi E, Portilla Fernandez EC, Jan Danser AH, Roks AJM. The importance of the nitric oxide-cGMP pathway in age-related cardiovascular disease: Focus on phosphodiesterase-1 and soluble guanylate cyclase. Basic Clin Pharmacol Toxicol 2019; 127:67-80. [PMID: 31495057 DOI: 10.1111/bcpt.13319] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022]
Abstract
Among ageing-related illnesses, cardiovascular disease (CVD) remains the leading cause of morbidity and mortality causing one-third of all deaths worldwide. Ageing evokes a number of functional, pharmacological and morphological changes in the vasculature, accompanied by a progressive failure of protective and homeostatic mechanisms, resulting in target organ damage. Impaired vasomotor, proliferation, migration, antithrombotic and anti-inflammatory function in both the endothelial and vascular smooth muscle cells are parts of the vascular ageing phenotype. The endothelium regulates these functions by the release of a wide variety of active molecules including endothelium-derived relaxing factors such as nitric oxide, prostacyclin (PGI2 ) and endothelium-derived hyperpolarization (EDH). During ageing, a functional decay of the nitric oxide pathway takes place. Nitric oxide signals to VSMC and other important cell types for vascular homeostasis through the second messenger cyclic guanosine monophosphate (cGMP). Maintenance of proper cGMP levels is an important goal in sustainment of proper vascular function during ageing. For this purpose, different components can be targeted in this signalling system, and among them, phosphodiesterase-1 (PDE1) and soluble guanylate cyclase (sGC) are crucial. This review focuses on the role of PDE1 and sGC in conditions that are relevant for vascular ageing.
Collapse
Affiliation(s)
- Keivan Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ehsan Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eliana C Portilla Fernandez
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Hashimoto T, Kim GE, Tunin RS, Adesiyun T, Hsu S, Nakagawa R, Zhu G, O'Brien JJ, Hendrick JP, Davis RE, Yao W, Beard D, Hoxie HR, Wennogle LP, Lee DI, Kass DA. Acute Enhancement of Cardiac Function by Phosphodiesterase Type 1 Inhibition. Circulation 2019; 138:1974-1987. [PMID: 30030415 DOI: 10.1161/circulationaha.117.030490] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Phosphodiesterase type-1 (PDE1) hydrolyzes cAMP and cGMP and is constitutively expressed in the heart, although cardiac effects from its acute inhibition in vivo are largely unknown. Existing data are limited to rodents expressing mostly the cGMP-favoring PDE1A isoform. Human heart predominantly expresses PDE1C with balanced selectivity for cAMP and cGMP. Here, we determined the acute effects of PDE1 inhibition in PDE1C-expressing mammals, dogs, and rabbits, in normal and failing hearts, and explored its regulatory pathways. METHODS Conscious dogs chronically instrumented for pressure-volume relations were studied before and after tachypacing-induced heart failure (HF). A selective PDE1 inhibitor (ITI-214) was administered orally or intravenously±dobutamine. Pressure-volume analysis in anesthetized rabbits tested the role of β-adrenergic and adenosine receptor signaling on ITI-214 effects. Sarcomere and calcium dynamics were studied in rabbit left ventricular myocytes. RESULTS In normal and HF dogs, ITI-214 increased load-independent contractility, improved relaxation, and reduced systemic arterial resistance, raising cardiac output without altering systolic blood pressure. Heart rate increased, but less so in HF dogs. ITI-214 effects were additive to β-adrenergic receptor agonism (dobutamine). Dobutamine but not ITI-214 increased plasma cAMP. ITI-214 induced similar cardiovascular effects in rabbits, whereas mice displayed only mild vasodilation and no contractility effects. In rabbits, β-adrenergic receptor blockade (esmolol) prevented ITI-214-mediated chronotropy, but inotropy and vasodilation remained unchanged. By contrast, adenosine A2B-receptor blockade (MRS-1754) suppressed ITI-214 cardiovascular effects. Adding fixed-rate atrial pacing did not alter the findings. ITI-214 alone did not affect sarcomere or whole-cell calcium dynamics, whereas β-adrenergic receptor agonism (isoproterenol) or PDE3 inhibition (cilostamide) increased both. Unlike cilostamide, which further enhanced shortening and peak calcium when combined with isoproterenol, ITI-214 had no impact on these responses. Both PDE1 and PDE3 inhibitors increased shortening and accelerated calcium decay when combined with forskolin, yet only cilostamide increased calcium transients. CONCLUSIONS PDE1 inhibition by ITI-214 in vivo confers acute inotropic, lusitropic, and arterial vasodilatory effects in PDE1C-expressing mammals with and without HF. The effects appear related to cAMP signaling that is different from that provided via β-adrenergic receptors or PDE3 modulation. ITI-214, which has completed phase I trials, may provide a novel therapy for HF.
Collapse
Affiliation(s)
- Toru Hashimoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Grace E Kim
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Richard S Tunin
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Tolulope Adesiyun
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.).,Dr Adesiyun's current affiliation is Department of Cardiovascular Medicine, Kyushu University Hospital3 Chome-1-1 Maidashi, Higashi Ward, Fukuoka, Japan
| | - Steven Hsu
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Ryo Nakagawa
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - Jennifer J O'Brien
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Joseph P Hendrick
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Robert E Davis
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Wei Yao
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - David Beard
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Helen R Hoxie
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Lawrence P Wennogle
- Intra-Cellular Therapies, Inc, New York, NY (J.J.O'B., J.P.H., R.E.D., W.Y., D.B., H.R.H., L.P.W.)
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (T.H., G.E.K., R.S.T., T.A., S.H., R.N., G.Z., D.I.L., D.A.K.)
| |
Collapse
|
36
|
Qiao N, Pan J, Kang Z, Liu G, Tang Z, Li Y. Effect of a background Ca 2+ entry pathway mediated by TRPC1 on myocardial damage of broilers with induced ascites syndrome. Avian Pathol 2019; 48:429-436. [PMID: 31084377 DOI: 10.1080/03079457.2019.1617834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Ascites syndrome (AS) in chickens is associated with profound vascular remodelling and increased pulmonary artery pressure as well as right ventricular hypertrophy. Classical transient receptor potential cation channels (TRPCs) are key regulators of cardiac hypertrophy that act via regulation of calcium influx in mammals. We investigated whether classical transient receptor potential channels in chickens with right ventricular hypertrophy still possess this mechanism for regulating Ca2+ flux. Intravenous injection of cellulose particles was successfully used to induce AS in chickens, and tissues were examined 22 days after treatment. The chickens in the test group showed cardiac hypertrophy with oedema of the cardiac muscle and disruption of myofilaments. The right-to-total ventricle weight ratio (RV/TV), the levels of serum aspartate aminotransferase (AST) and creatine kinase (CK) of the test group were significantly higher than in the control group. Intracellular calcium levels were significantly increased in cardiomyocytes from chickens in the test group. Gene expression of TRPC3, TRPC4, TRPC5, TRPC6 and TRPC7 in heart tissues from the test group showed no significant differences compared with controls. However, TRPC1 protein levels, as well as mRNA levels, were down-regulated in the heart muscle of AS chickens (P < 0.05). Although we observed an increase in calcium concentration, the expression of TRPC1 decreased in cardiac cells. We hypothesized that an increase in intracellular free calcium concentration could inversely regulate calcium channel expression. RESEARCH HIGHLIGHTS Intracellular Ca2+ levels were increased in the myocardium of AS broilers. Expression of TRPC1, which mediates calcium influx, was decreased in the myocardium of AS broilers. The relationship between intracellular Ca2+ levels and expression of TRPC1 requires further study.
Collapse
Affiliation(s)
- Na Qiao
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| | - Zhenlong Kang
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| | - Gaoyang Liu
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University , Guangzhou , P. R. People's Republic of China
| |
Collapse
|
37
|
Reddy YNV, Borlaug BA, O’Connor CM, Gersh BJ. Novel approaches to the management of chronic systolic heart failure: future directions and unanswered questions. Eur Heart J 2019; 41:1764-1774. [DOI: 10.1093/eurheartj/ehz364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/25/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract
Despite improvements in outcomes in the last few decades for heart failure (HF) with reduced ejection fraction (HFrEF), there still remains a need for novel therapies as many patients incompletely recover with existing therapies and progress to advanced HF. In this review, we will discuss recent advances in the management of HFrEF with a focus on upcoming therapies that hold the greatest promise for clinical use. We will discuss novel pharmacological therapies and areas of uncertainty with existing therapies. We will also discuss the potential utility and controversy surrounding novel interventions for HF such as percutaneous mitral valve repair, atrial fibrillation ablation, and other emerging interventions with positive signals for benefit in HFrEF. Finally, we will summarize the current state of stem cell and gene therapy for HFrEF and future directions.
Collapse
Affiliation(s)
- Yogesh N V Reddy
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| | - Barry A Borlaug
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| | | | - Bernard J Gersh
- The Department of Cardiovascular Medicine, Mayo Clinic Rochester, 200 First Street SW, MN 55906, USA
| |
Collapse
|
38
|
Abstract
Advances in the treatment of heart failure with reduced ejection fraction due to systolic dysfunction are engaging an ever-expanding compendium of molecular signaling targets. Well established approaches modifying hemodynamics and cell biology by neurohumoral receptor blockade are evolving, exploring the role and impact of modulating intracellular signaling pathways with more direct myocardial effects. Even well-tread avenues are being reconsidered with new insights into the signaling engaged and thus opportunity to treat underlying myocardial disease. This review explores therapies that have proven successful, those that have not, those that are moving into the clinic but whose utility remains to be confirmed, and those that remain in the experimental realm. The emphasis is on signaling pathways that are tractable for therapeutic manipulation. Of the approaches yet to be tested in humans, we chose those with a well-established experimental history, where clinical translation may be around the corner. The breadth of opportunities bodes well for the next generation of heart failure therapeutics.
Collapse
Affiliation(s)
| | | | - David A. Kass
- Division of Cardiology, Department of Medicine
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore Maryland, 21205
| |
Collapse
|
39
|
Zhang H, Pan B, Wu P, Parajuli N, Rekhter MD, Goldberg AL, Wang X. PDE1 inhibition facilitates proteasomal degradation of misfolded proteins and protects against cardiac proteinopathy. SCIENCE ADVANCES 2019; 5:eaaw5870. [PMID: 31131329 PMCID: PMC6531002 DOI: 10.1126/sciadv.aaw5870] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/11/2019] [Indexed: 06/09/2023]
Abstract
No current treatment targets cardiac proteotoxicity or can reduce mortality of heart failure (HF) with preserved ejection fraction (HFpEF). Selective degradation of misfolded proteins by the ubiquitin-proteasome system (UPS) is vital to the cell. Proteasome impairment contributes to HF. Activation of cAMP-dependent protein kinase (PKA) or cGMP-dependent protein kinase (PKG) facilitates proteasome functioning. Phosphodiesterase 1 (PDE1) hydrolyzes both cyclic nucleotides and accounts for most PDE activities in human myocardium. We report that PDE1 inhibition (IC86430) increases myocardial 26S proteasome activities and UPS proteolytic function in mice. Mice with CryABR120G-based proteinopathy develop HFpEF and show increased myocardial PDE1A expression. PDE1 inhibition markedly attenuates HFpEF, improves mouse survival, increases PKA-mediated proteasome phosphorylation, and reduces myocardial misfolded CryAB. Therefore, PDE1 inhibition induces PKA- and PKG-mediated promotion of proteasomal degradation of misfolded proteins and treats HFpEF caused by CryABR120G, representing a potentially new therapeutic strategy for HFpEF and heart disease with increased proteotoxic stress.
Collapse
Affiliation(s)
- Hanming Zhang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Bo Pan
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Penglong Wu
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
- Department of Pathophysiology, Guangzhou Medical University College of Basic Medical Sciences, Guangzhou, Guangdong 511436, China
| | - Nirmal Parajuli
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Mark D. Rekhter
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Alfred L. Goldberg
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| |
Collapse
|
40
|
Shete V, Liu N, Jia Y, Viswakarma N, Reddy JK, Thimmapaya B. Mouse Cardiac Pde1C Is a Direct Transcriptional Target of Pparα. Int J Mol Sci 2018; 19:ijms19123704. [PMID: 30469494 PMCID: PMC6321386 DOI: 10.3390/ijms19123704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 1C (PDE1C) is expressed in mammalian heart and regulates cardiac functions by controlling levels of second messenger cyclic AMP and cyclic GMP (cAMP and cGMP, respectively). However, molecular mechanisms of cardiac Pde1c regulation are currently unknown. In this study, we demonstrate that treatment of wild type mice and H9c2 myoblasts with Wy-14,643, a potent ligand of nuclear receptor peroxisome-proliferator activated receptor alpha (PPARα), leads to elevated cardiac Pde1C mRNA and cardiac PDE1C protein, which correlate with reduced levels of cAMP. Furthermore, using mice lacking either Pparα or cardiomyocyte-specific Med1, the major subunit of Mediator complex, we show that Wy-14,643-mediated Pde1C induction fails to occur in the absence of Pparα and Med1 in the heart. Finally, using chromatin immunoprecipitation assays we demonstrate that PPARα binds to the upstream Pde1C promoter sequence on two sites, one of which is a palindrome sequence (agcTAGGttatcttaacctagc) that shows a robust binding. Based on these observations, we conclude that cardiac Pde1C is a direct transcriptional target of PPARα and that Med1 may be required for the PPARα mediated transcriptional activation of cardiac Pde1C.
Collapse
Affiliation(s)
- Varsha Shete
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Janardan K Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|