1
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
2
|
Moss WJ, Brusini L, Kuehnel R, Brochet M, Brown KM. Apicomplexan phosphodiesterases in cyclic nucleotide turnover: conservation, function, and therapeutic potential. mBio 2024; 15:e0305623. [PMID: 38132724 PMCID: PMC10865986 DOI: 10.1128/mbio.03056-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Apicomplexa encompasses a large number of intracellular parasites infecting a wide range of animals. Cyclic nucleotide signaling is crucial for a variety of apicomplexan life stages and cellular processes. The cyclases and kinases that synthesize and respond to cyclic nucleotides (i.e., 3',5'-cyclic guanosine monophosphate and 3',5'-cyclic adenosine monophosphate) are highly conserved and essential throughout the parasite phylum. Growing evidence indicates that phosphodiesterases (PDEs) are also critical for regulating cyclic nucleotide signaling via cyclic nucleotide hydrolysis. Here, we discuss recent advances in apicomplexan PDE biology and opportunities for therapeutic interventions, with special emphasis on the major human apicomplexan parasite genera Plasmodium, Toxoplasma, Cryptosporidium, and Babesia. In particular, we show a highly flexible repertoire of apicomplexan PDEs associated with a wide range of cellular requirements across parasites and lifecycle stages. Despite this phylogenetic diversity, cellular requirements of apicomplexan PDEs for motility, host cell egress, or invasion are conserved. However, the molecular wiring of associated PDEs is extremely malleable suggesting that PDE diversity and redundancy are key for the optimization of cyclic nucleotide turnover to respond to the various environments encountered by each parasite and life stage. Understanding how apicomplexan PDEs are regulated and integrating multiple signaling systems into a unified response represent an untapped avenue for future exploration.
Collapse
Affiliation(s)
- William J. Moss
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lorenzo Brusini
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ronja Kuehnel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kevin M. Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
3
|
Mahmood S, Lozano Gonzalez M, Tummalapalli S, Eberhard J, Ly J, Hoffman CS, Kelly MP, Gordon J, Colussi D, Childers W, Rotella DP. First Optimization of Novel, Potent, Selective PDE11A4 Inhibitors for Age-Related Cognitive Decline. J Med Chem 2023; 66:14597-14608. [PMID: 37862143 PMCID: PMC10641827 DOI: 10.1021/acs.jmedchem.3c01088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Indexed: 10/22/2023]
Abstract
Phosphodiesterase 11A4 (PDE11A4) is a dual-acting cyclic nucleotide hydrolase expressed in neurons in the CA1, subiculum, amygdalostriatal transition area and amygdalohippocampal area of the extended hippocampal formation. PDE11A4 is the only PDE enzyme to emanate solely from hippocampal formation, a key brain region for the formation of long-term memory. PDE11A4 expression increases in the hippocampal formation of both humans and rodents as they age. Interestingly, PDE11A knockout mice do not show age-related deficits in associative memory and show no gross histopathology. This suggests that inhibition of PDE11A4 might serve as a therapeutic option for age-related cognitive decline. A novel, yeast-based high throughput screen previously identified moderately potent, selective PDE11A4 inhibitors, and this work describes initial efforts that improved potency more than 10-fold and improved some pharmaceutical properties of one of these scaffolds, leading to selective, cell-penetrant PDE11A4 inhibitors, one of which is 10-fold more potent compared to tadalafil in cell-based activity.
Collapse
Affiliation(s)
- Shams
ul Mahmood
- Department
of Chemistry & Biochemistry, Montclair
State University, Montclair, New Jersey 07043, United States
- Sokol
Institute of Pharmaceutical Life Sciences, Montclair State University, Montclair, New Jersey 07043, United States
| | - Mariana Lozano Gonzalez
- Department
of Chemistry & Biochemistry, Montclair
State University, Montclair, New Jersey 07043, United States
- Sokol
Institute of Pharmaceutical Life Sciences, Montclair State University, Montclair, New Jersey 07043, United States
| | - Sreedhar Tummalapalli
- Department
of Chemistry & Biochemistry, Montclair
State University, Montclair, New Jersey 07043, United States
- Sokol
Institute of Pharmaceutical Life Sciences, Montclair State University, Montclair, New Jersey 07043, United States
| | - Jeremy Eberhard
- Biology
Department, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Judy Ly
- Biology
Department, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Charles S. Hoffman
- Biology
Department, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Michy P. Kelly
- Department
of Anatomy & Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - John Gordon
- Moulder
Center for Drug Discovery Research, Temple
University, Philadelphia, Pennsylvania 19140, United States
| | - Dennis Colussi
- Moulder
Center for Drug Discovery Research, Temple
University, Philadelphia, Pennsylvania 19140, United States
| | - Wayne Childers
- Moulder
Center for Drug Discovery Research, Temple
University, Philadelphia, Pennsylvania 19140, United States
| | - David P. Rotella
- Department
of Chemistry & Biochemistry, Montclair
State University, Montclair, New Jersey 07043, United States
- Sokol
Institute of Pharmaceutical Life Sciences, Montclair State University, Montclair, New Jersey 07043, United States
| |
Collapse
|
4
|
Satori NA, Pacini ESA, Godinho RO. Impact of the cAMP efflux and extracellular cAMP-adenosine pathway on airway smooth muscle relaxation induced by formoterol and phosphodiesterase inhibitors. Chem Biol Interact 2023; 382:110630. [PMID: 37442289 DOI: 10.1016/j.cbi.2023.110630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
β2-adrenoceptors agonists and phosphodiesterase (PDE) inhibitors are effective bronchodilators, due to their ability to increase intracellular cyclic AMP (cAMP) levels and induce airway smooth muscle (ASM) relaxation. We have shown that increment of intracellular cAMP induced by β2-adrenoceptors agonist fenoterol is followed by efflux of cAMP, which is converted by ecto-PDE and ecto-5'-nucleotidases (ecto-5'NT) to adenosine, leading to ASM contraction. Here we evaluate whether other classical bronchodilators used to treat asthma and chronic obstructive pulmonary disease (COPD) could induce cAMP efflux and, as consequence, influence the ASM contractility. Our results showed that β2-adrenoceptor agonists formoterol and PDE inhibitors IBMX, aminophylline and roflumilast induced cAMP efflux and a concentration-dependent relaxation of rat trachea precontracted with carbachol. Pretreatment of tracheas with MK-571 (MRP transporter inhibitor), AMP-CP (ecto-5'NT inhibitor) or CGS-15943 (nonselective adenosine receptor antagonist) potentiated the relaxation induced by β2-adrenoceptor agonists but did not change the relaxation induced by PDE inhibitors. These data showed that all bronchodilators tested were able to induce cAMP efflux. However, only β2-adrenoceptor-induced relaxation of tracheal smooth muscle was affected by cAMP efflux and extracellular cAMP-adenosine pathway.
Collapse
Affiliation(s)
- Naiara Ayako Satori
- Division of Cellular Pharmacology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Enio Setsuo Arakaki Pacini
- Division of Cellular Pharmacology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil
| | - Rosely Oliveira Godinho
- Division of Cellular Pharmacology, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM/UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Campolo F, Assenza MR, Venneri MA, Barbagallo F. Once upon a Testis: The Tale of Cyclic Nucleotide Phosphodiesterase in Testicular Cancers. Int J Mol Sci 2023; 24:ijms24087617. [PMID: 37108780 PMCID: PMC10146088 DOI: 10.3390/ijms24087617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Phosphodiesterases are key regulators that fine tune the intracellular levels of cyclic nucleotides, given their ability to hydrolyze cAMP and cGMP. They are critical regulators of cAMP/cGMP-mediated signaling pathways, modulating their downstream biological effects such as gene expression, cell proliferation, cell-cycle regulation but also inflammation and metabolic function. Recently, mutations in PDE genes have been identified and linked to human genetic diseases and PDEs have been demonstrated to play a potential role in predisposition to several tumors, especially in cAMP-sensitive tissues. This review summarizes the current knowledge and most relevant findings regarding the expression and regulation of PDE families in the testis focusing on PDEs role in testicular cancer development.
Collapse
Affiliation(s)
- Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Rita Assenza
- Faculty of Medicine and Surgery, "Kore" University of Enna, 94100 Enna, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Barbagallo
- Faculty of Medicine and Surgery, "Kore" University of Enna, 94100 Enna, Italy
| |
Collapse
|
6
|
Sheng J, Zhang S, Wu L, Kumar G, Liao Y, GK P, Fan H. Inhibition of phosphodiesterase: A novel therapeutic target for the treatment of mild cognitive impairment and Alzheimer's disease. Front Aging Neurosci 2022; 14:1019187. [PMID: 36268188 PMCID: PMC9577554 DOI: 10.3389/fnagi.2022.1019187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is ranked as the 6th leading cause of death in the US. The prevalence of AD and dementia is steadily increasing and expected cases in USA is 14.8 million by 2050. Neuroinflammation and gradual neurodegeneration occurs in Alzheimer's disease. However, existing medications has limitation to completely abolish, delay, or prevent disease progression. Phosphodiesterases (PDEs) are large family of enzymes to hydrolyze the 3'-phosphodiester links in cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) in signal-transduction pathways for generation of 5'-cyclic nucleotides. It plays vital role to orchestrate several pharmacological activities for proper cell functioning and regulating the levels of cAMP and cGMP. Several evidence has suggested that abnormal cAMP signaling is linked to cognitive problems in neurodegenerative disorders like AD. Therefore, the PDE family has become a widely accepted and multipotential therapeutic target for neurodegenerative diseases. Notably, modulation of cAMP/cGMP by phytonutrients has a huge potential for the management of AD. Natural compounds have been known to inhibit phosphodiesterase by targeting key enzymes of cGMP synthesis pathway, however, the mechanism of action and their therapeutic efficacy has not been explored extensively. Currently, few PDE inhibitors such as Vinpocetine and Nicergoline have been used for treatment of central nervous system (CNS) disorders. Considering the role of flavonoids to inhibit PDE, this review discussed the therapeutic potential of natural compounds with PDE inhibitory activity for the treatment of AD and related dementia.
Collapse
Affiliation(s)
- Jianwen Sheng
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Shanjin Zhang
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Lule Wu
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Yuanhang Liao
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Pratap GK
- Department of Biochemistry, Davangere University, Davangere, India
| | - Huizhen Fan
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| |
Collapse
|
7
|
Kong G, Lee H, Vo TTT, Juang U, Kwon SH, Park J, Park J, Kim SH. Functional characteristics and research trends of PDE11A in human diseases (Review). Mol Med Rep 2022; 26:298. [PMID: 35929507 PMCID: PMC9434997 DOI: 10.3892/mmr.2022.12814] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/15/2022] [Indexed: 11/06/2022] Open
Abstract
cAMP and cGMP are important secondary messengers involved in cell regulation and metabolism driven by the G protein-coupled receptor. cAMP is converted via adenylyl cyclase (AC) and activates protein kinase A to phosphorylate intracellular proteins that mediate specific responses. cAMP signaling serves a role at multiple steps in tumorigenesis. The level of cAMP is increased in association with cancer cell formation through activation of AC-stimulatory G protein by mutation. Phosphodiesterases (PDEs) hydrolyze cAMP and cGMP to AMP and GMP. PDEs are composed of 11 families, and each can hydrolyze cAMP and cGMP or both cAMP and cGMP. PDEs perform various roles depending on their location and expression site, and are involved in several diseases, including male erectile dysfunction, pulmonary hypertension, Alzheimer's disease and schizophrenia. PDE11A is the 11th member of the PDE family and is characterized by four splice variants with varying tissue expression and N-terminal regulatory regions. Among tissues, the expression of PDE11A was highest in the prostate, and it was also expressed in hepatic skeletal muscle, pituitary, pancreas and kidney. PDE11A is the first PDE associated with an adrenocortical tumor associated genetic condition. In several studies, three PDE11A mutations have been reported in patients with Cushing syndrome with primary pigmented nodular adrenocortical disease or isolated micronodular adrenocortical disease without other genetic defects. It has been reported that an increase in PDE11A expression affects the proliferation of glioblastoma and worsens patient prognosis. The present mini-review summarizes the location of PDE11A expression, the impact of structural differences and disease relevance.
Collapse
Affiliation(s)
- Gyeyeong Kong
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyunji Lee
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Thuy-Trang T Vo
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Uijin Juang
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Jisoo Park
- Mitos Research Institute, Mitos Therapeutics Inc., Daejeon 34134, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
8
|
African Swine Fever Virus EP364R and C129R Target Cyclic GMP-AMP To Inhibit the cGAS-STING Signaling Pathway. J Virol 2022; 96:e0102222. [PMID: 35861515 PMCID: PMC9364804 DOI: 10.1128/jvi.01022-22] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
African swine fever virus (ASFV) is a highly pathogenic swine DNA virus with high mortality that causes African swine fever (ASF) in domestic pigs and wild boars. For efficient viral infection, ASFV has developed complex strategies to evade key components of antiviral innate immune responses. However, the immune escape mechanism of ASFV remains unclear. Upon ASFV infection, cyclic GMP-AMP (2′,3′-cGAMP) synthase (cGAS), a cytosolic DNA sensor, recognizes ASFV DNA and synthesizes the second messenger 2′,3′-cGAMP, which triggers interferon (IFN) production to interfere with viral replication. In this study, we demonstrated a novel immune evasion mechanism of ASFV EP364R and C129R, which blocks cellular cyclic 2′,3′-cGAMP-mediated antiviral responses. ASFV EP364R and C129R with nuclease homology inhibit IFN-mediated responses by specifically interacting with 2′,3′-cGAMP and exerting their phosphodiesterase (PDE) activity to cleave 2′,3′-cGAMP. Particularly notable is that ASFV EP364R had a region of homology with the stimulator of interferon genes (STING) protein containing a 2′,3′-cGAMP-binding motif and point mutations in the Y76S and N78A amino acids of EP364R that impaired interaction with 2′,3′-cGAMP and restored subsequent antiviral responses. These results highlight a critical role for ASFV EP364R and C129R in the inhibition of IFN responses and could be used to develop ASFV live attenuated vaccines. IMPORTANCE African swine fever (ASF) is a highly contagious hemorrhagic disease in domestic pigs and wild boars caused by African swine fever virus (ASFV). ASF is a deadly epidemic disease in the global pig industry, but no drugs or vaccines are available. Understanding the pathogenesis of ASFV is essential to developing an effective live attenuated ASFV vaccine, and investigating the immune evasion mechanisms of ASFV is crucial to improve the understanding of its pathogenesis. In this study, for the first time, we identified the EP364R and C129R, uncharacterized proteins that inhibit type I interferon signaling. ASFV EP364R and C129R specifically interacted with 2′,3′-cGAMP, the mammalian second messenger, and exerted phosphodiesterase activity to cleave 2′,3′-cGAMP. In this study, we discovered a novel mechanism by which ASFV inhibits IFN-mediated antiviral responses, and our findings can guide the understanding of ASFV pathogenesis and the development of live attenuated ASFV vaccines.
Collapse
|
9
|
Bolger GB. The cAMP-signaling cancers: Clinically-divergent disorders with a common central pathway. Front Endocrinol (Lausanne) 2022; 13:1024423. [PMID: 36313756 PMCID: PMC9612118 DOI: 10.3389/fendo.2022.1024423] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/27/2022] [Indexed: 12/01/2022] Open
Abstract
The cAMP-signaling cancers, which are defined by functionally-significant somatic mutations in one or more elements of the cAMP signaling pathway, have an unexpectedly wide range of cell origins, clinical manifestations, and potential therapeutic options. Mutations in at least 9 cAMP signaling pathway genes (TSHR, GPR101, GNAS, PDE8B, PDE11A, PRKARA1, PRKACA, PRKACB, and CREB) have been identified as driver mutations in human cancer. Although all cAMP-signaling pathway cancers are driven by mutation(s) that impinge on a single signaling pathway, the ultimate tumor phenotype reflects interactions between five critical variables: (1) the precise gene(s) that undergo mutation in each specific tumor type; (2) the effects of specific allele(s) in any given gene; (3) mutations in modifier genes (mutational "context"); (4) the tissue-specific expression of various cAMP signaling pathway elements in the tumor stem cell; and (5) and the precise biochemical regulation of the pathway components in tumor cells. These varying oncogenic mechanisms reveal novel and important targets for drug discovery. There is considerable diversity in the "druggability" of cAMP-signaling components, with some elements (GPCRs, cAMP-specific phosphodiesterases and kinases) appearing to be prime drug candidates, while other elements (transcription factors, protein-protein interactions) are currently refractory to robust drug-development efforts. Further refinement of the precise driver mutations in individual tumors will be essential for directing priorities in drug discovery efforts that target these mutations.
Collapse
|
10
|
Poitras EL, Gust SL, Kerr PM, Plane F. Repurposing of the PDE5 Inhibitor Sildenafil for the Treatment of Persistent Pulmonary Hypertension in Neonates. Curr Med Chem 2021; 28:2418-2437. [PMID: 32964819 DOI: 10.2174/0929867327666200923151924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/21/2020] [Accepted: 08/21/2020] [Indexed: 11/22/2022]
Abstract
Nitric oxide (NO), an important endogenous signaling molecule released from vascular endothelial cells and nerves, activates the enzyme soluble guanylate cyclase to catalyze the production of cyclic guanosine monophosphate (cGMP) from guanosine triphosphate. cGMP, in turn, activates protein kinase G to phosphorylate a range of effector proteins in smooth muscle cells that reduce intracellular Ca2+ levels to inhibit both contractility and proliferation. The enzyme phosphodiesterase type 5 (PDE5) curtails the actions of cGMP by hydrolyzing it into inactive 5'-GMP. Small molecule PDE5 inhibitors (PDE5is), such as sildenafil, prolong the availability of cGMP and therefore, enhance NO-mediated signaling. PDE5is are the first-line treatment for erectile dysfunction but are also now approved for the treatment of pulmonary arterial hypertension (PAH) in adults. Persistent pulmonary hypertension in neonates (PPHN) is currently treated with inhaled NO, but this is an expensive option and around 1/3 of newborns are unresponsive, resulting in the need for alternative approaches. Here the development, chemistry and pharmacology of PDE5is, the use of sildenafil for erectile dysfunction and PAH, are summarized and then current evidence for the utility of further repurposing of sildenafil, as a treatment for PPHN, is critically reviewed.
Collapse
Affiliation(s)
- Erika L Poitras
- Department of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Stephen L Gust
- Department of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Paul M Kerr
- Faculty of Nursing, Robbins Health Learning Centre, MacEwan University, Edmonton, Alberta T5J 4S2, Canada
| | - Frances Plane
- Department of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| |
Collapse
|
11
|
Sun J, Xiao Z, Haider A, Gebhard C, Xu H, Luo HB, Zhang HT, Josephson L, Wang L, Liang SH. Advances in Cyclic Nucleotide Phosphodiesterase-Targeted PET Imaging and Drug Discovery. J Med Chem 2021; 64:7083-7109. [PMID: 34042442 DOI: 10.1021/acs.jmedchem.1c00115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) control the intracellular concentrations of cAMP and cGMP in virtually all mammalian cells. Accordingly, the PDE family regulates a myriad of physiological functions, including cell proliferation, differentiation and apoptosis, gene expression, central nervous system function, and muscle contraction. Along this line, dysfunction of PDEs has been implicated in neurodegenerative disorders, coronary artery diseases, chronic obstructive pulmonary disease, and cancer development. To date, 11 PDE families have been identified; however, their distinct roles in the various pathologies are largely unexplored and subject to contemporary research efforts. Indeed, there is growing interest for the development of isoform-selective PDE inhibitors as potential therapeutic agents. Similarly, the evolving knowledge on the various PDE isoforms has channeled the identification of new PET probes, allowing isoform-selective imaging. This review highlights recent advances in PDE-targeted PET tracer development, thereby focusing on efforts to assess disease-related PDE pathophysiology and to support isoform-selective drug discovery.
Collapse
Affiliation(s)
- Jiyun Sun
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Zhiwei Xiao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Ahmed Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, Zurich 8006, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Schlieren 8952, Switzerland
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Han-Ting Zhang
- Departments of Neuroscience, Behavioral Medicine & Psychiatry, and Physiology & Pharmacology, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia 26506, United States
| | - Lee Josephson
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Lu Wang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States.,Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
12
|
Epstein PM, Basole C, Brocke S. The Role of PDE8 in T Cell Recruitment and Function in Inflammation. Front Cell Dev Biol 2021; 9:636778. [PMID: 33937235 PMCID: PMC8085600 DOI: 10.3389/fcell.2021.636778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice to treat inflammatory disorders, with three PDE4 inhibitors currently in clinical use as therapeutics for psoriasis, psoriatic arthritis, atopic dermatitis and chronic obstructive pulmonary disease. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. It was shown that PDE8A plays a major role in controlling T cell and breast cancer cell motility, including adhesion to endothelial cells under physiological shear stress and chemotaxis. This is a unique function of PDE8 not shared by PDE4, another cAMP specific PDE, employed, as noted, as an anti-inflammatory therapeutic. Additionally, a regulatory role was shown for the PDE8A-rapidly accelerated fibrosarcoma (Raf)-1 kinase signaling complex in myelin antigen reactive CD4+ effector T cell adhesion and locomotion by a mechanism differing from that of PDE4. The PDE8A-Raf-1 kinase signaling complex affects T cell motility, at least in part, via regulating the LFA-1 integrin mediated adhesion to ICAM-1. The findings that PDE8A and its isoforms are expressed at higher levels in naive and myelin oligodendrocyte glycoprotein (MOG)35–55 activated effector T (Teff) cells compared to regulatory T (Treg) cells and that PDE8 inhibition specifically affects MOG35–55 activated Teff cell adhesion, indicates that PDE8A could represent a new beneficial target expressed in pathogenic Teff cells in CNS inflammation. The implications of this work for targeting PDE8 in inflammation will be discussed in this review.
Collapse
Affiliation(s)
- Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, CT, United States
| | - Chaitali Basole
- Department of Immunology, UConn Health, Farmington, CT, United States
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
13
|
Qin W, Zhou A, Zuo X, Jia L, Li F, Wang Q, Li Y, Wei Y, Jin H, Cruchaga C, Benitez BA, Jia J. Exome sequencing revealed PDE11A as a novel candidate gene for early-onset Alzheimer's disease. Hum Mol Genet 2021; 30:811-822. [PMID: 33835157 PMCID: PMC8161517 DOI: 10.1093/hmg/ddab090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 11/14/2022] Open
Abstract
To identify novel risk genes and better understand the molecular pathway underlying Alzheimer's disease (AD), whole-exome sequencing was performed in 215 early-onset AD (EOAD) patients and 255 unrelated healthy controls of Han Chinese ethnicity. Subsequent validation, computational annotation and in vitro functional studies were performed to evaluate the role of candidate variants in EOAD. We identified two rare missense variants in the phosphodiesterase 11A (PDE11A) gene in individuals with EOAD. Both variants are located in evolutionarily highly conserved amino acids, are predicted to alter the protein conformation and are classified as pathogenic. Furthermore, we found significantly decreased protein levels of PDE11A in brain samples of AD patients. Expression of PDE11A variants and knockdown experiments with specific short hairpin RNA (shRNA) for PDE11A both resulted in an increase of AD-associated Tau hyperphosphorylation at multiple epitopes in vitro. PDE11A variants or PDE11A shRNA also caused increased cyclic adenosine monophosphate (cAMP) levels, protein kinase A (PKA) activation and cAMP response element-binding protein phosphorylation. In addition, pretreatment with a PKA inhibitor (H89) suppressed PDE11A variant-induced Tau phosphorylation formation. This study offers insight into the involvement of Tau phosphorylation via the cAMP/PKA pathway in EOAD pathogenesis and provides a potential new target for intervention.
Collapse
Affiliation(s)
- Wei Qin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Aihong Zhou
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Xiumei Zuo
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Ying Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Hongmei Jin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO 63110, USA
- Department of Genetics, Washington University, St. Louis, MO 63110, USA
| | - Bruno A Benitez
- Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO 63110, USA
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Capital Medical University, Beijing 10053, China
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 10053, China
- Center of Alzheimer’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 10053, China
- To whom correspondence should be addressed at: Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing 100053, P.R. China. Tel: 0086 10 83199449; Fax: 0086 10 83128678; ,
| |
Collapse
|
14
|
The PDE-Opathies: Diverse Phenotypes Produced by a Functionally Related Multigene Family. Trends Genet 2021; 37:669-681. [PMID: 33832760 DOI: 10.1016/j.tig.2021.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/30/2022]
Abstract
The phosphodiesterase (PDE)-opathies, an expanding set of disorders caused by germline mutations in cyclic nucleotide PDEs, present an intriguing paradox. The enzymes encoded by the PDE family all hydrolyze cAMP and/or cGMP, but mutations in different family members produce very divergent phenotypes. Three interacting factors have been shown recently to contribute to this phenotypic diversity: (i) the 21 genes encode over 80 different isoforms, using alternative mRNA splicing and related mechanisms; (ii) the various isoforms have different regulatory mechanisms, mediated by their unique amino-terminal regulatory domains; (iii) the isoforms differ widely in their pattern of tissue expression. These mechanisms explain why many PDE-opathies are gain-of-function mutations and how they exemplify uniqueness and redundancy within a multigene family.
Collapse
|
15
|
Regan JT, Mirczuk SM, Scudder CJ, Stacey E, Khan S, Worwood M, Powles T, Dennis-Beron JS, Ginley-Hidinger M, McGonnell IM, Volk HA, Strickland R, Tivers MS, Lawson C, Lipscomb VJ, Fowkes RC. Sensitivity of the Natriuretic Peptide/cGMP System to Hyperammonaemia in Rat C6 Glioma Cells and GPNT Brain Endothelial Cells. Cells 2021; 10:cells10020398. [PMID: 33672024 PMCID: PMC7919485 DOI: 10.3390/cells10020398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/05/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
C-type natriuretic peptide (CNP) is the major natriuretic peptide of the central nervous system and acts via its selective guanylyl cyclase-B (GC-B) receptor to regulate cGMP production in neurons, astrocytes and endothelial cells. CNP is implicated in the regulation of neurogenesis, axonal bifurcation, as well as learning and memory. Several neurological disorders result in toxic concentrations of ammonia (hyperammonaemia), which can adversely affect astrocyte function. However, the relationship between CNP and hyperammonaemia is poorly understood. Here, we examine the molecular and pharmacological control of CNP in rat C6 glioma cells and rat GPNT brain endothelial cells, under conditions of hyperammonaemia. Concentration-dependent inhibition of C6 glioma cell proliferation by hyperammonaemia was unaffected by CNP co-treatment. Furthermore, hyperammonaemia pre-treatment (for 1 h and 24 h) caused a significant inhibition in subsequent CNP-stimulated cGMP accumulation in both C6 and GPNT cells, whereas nitric-oxide-dependent cGMP accumulation was not affected. CNP-stimulated cGMP efflux from C6 glioma cells was significantly reduced under conditions of hyperammonaemia, potentially via a mechanism involving changed in phosphodiesterase expression. Hyperammonaemia-stimulated ROS production was unaffected by CNP but enhanced by a nitric oxide donor in C6 cells. Extracellular vesicle production from C6 cells was enhanced by hyperammonaemia, and these vesicles caused impaired CNP-stimulated cGMP signalling in GPNT cells. Collectively, these data demonstrate functional interaction between CNP signalling and hyperammonaemia in C6 glioma and GPNT cells, but the exact mechanisms remain to be established.
Collapse
Affiliation(s)
- Jacob T. Regan
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Samantha M. Mirczuk
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (I.M.M.); (C.L.)
| | - Christopher J. Scudder
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (I.M.M.); (C.L.)
| | - Emily Stacey
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Sabah Khan
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Michael Worwood
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Torinn Powles
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - J. Sebastian Dennis-Beron
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Matthew Ginley-Hidinger
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
| | - Imelda M. McGonnell
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (I.M.M.); (C.L.)
| | - Holger A. Volk
- Stiftung Tierärztliche Hochschule Hannover, Klinik für Kleintiere, Bünteweg, 930559 Hannover, Germany;
| | - Rhiannon Strickland
- Clinical Sciences & Services, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (R.S.); (V.J.L.)
| | - Michael S. Tivers
- Paragon Veterinary Referrals, Paragon Business Village Paragon Way, Red Hall Cres, Wakefield WF1 2DF, UK;
| | - Charlotte Lawson
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (I.M.M.); (C.L.)
| | - Victoria J. Lipscomb
- Clinical Sciences & Services, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (R.S.); (V.J.L.)
| | - Robert C. Fowkes
- Endocrine Signalling Group, Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (J.T.R.); (S.M.M.); (C.J.S.); (E.S.); (S.K.); (M.W.); (T.P.); (J.S.D.-B.); (M.G.-H.)
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK; (I.M.M.); (C.L.)
- Correspondence: ; Tel.: +44-207-468-1215
| |
Collapse
|
16
|
Levy I, Szarek E, Maria AG, Starrost M, De La Luz Sierra M, Faucz FR, Stratakis CA. A phosphodiesterase 11 (Pde11a) knockout mouse expressed functional but reduced Pde11a: Phenotype and impact on adrenocortical function. Mol Cell Endocrinol 2021; 520:111071. [PMID: 33127481 PMCID: PMC7771190 DOI: 10.1016/j.mce.2020.111071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 01/23/2023]
Abstract
Phosphodiesterases catalyze the hydrolysis of cyclic nucleotides and maintain physiologic levels of intracellular concentrations of cyclic adenosine and guanosine mono-phosphate (cAMP and cGMP, respectively). Increased cAMP signaling has been associated with adrenocortical tumors and Cushing syndrome. Genetic defects in phosphodiesterase 11A (PDE11A) may lead to increased cAMP signaling and have been found to predispose to the development of adrenocortical, prostate, and testicular tumors. A previously reported Pde11a knockout (Pde11a-/-) mouse line was studied and found to express PDE11A mRNA and protein still, albeit at reduced levels; functional studies in various tissues showed increased cAMP levels and reduced PDE11A activity. Since patients with PDE11A defects and Cushing syndrome have PDE11A haploinsufficiency, it was particularly pertinent to study this hypomorphic mouse line. Indeed, Pde11a-/- mice failed to suppress corticosterone secretion in response to low dose dexamethasone, and in addition exhibited adrenal subcapsular hyperplasia with predominant fetal-like features in the inner adrenal cortex, mimicking other mouse models of increased cAMP signaling in the adrenal cortex. We conclude that a previously reported Pde11a-/- mouse showed continuing expression and function of PDE11A in most tissues. Nevertheless, Pde11a partial inactivation in mice led to an adrenocortical phenotype that was consistent with what we see in patients with PDE11A haploinsufficiency.
Collapse
Affiliation(s)
- Isaac Levy
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA; Endocrine and Diabetes Unit. Edmond and Lily Safra Children's Hospital, Tel-Hashomer. Ramat Gan. Sackler School of Medicine, Ramat-aviv, Israel
| | - Eva Szarek
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Andrea Gutierrez Maria
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Matthew Starrost
- Division of Veterinary Resources, National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Maria De La Luz Sierra
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Fabio R Faucz
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.
| | - Constantine A Stratakis
- Section on Endocrinology & Genetics (SEGEN), Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
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
|
18
|
Shaaban MA, Elshaier YA, Hammad AH, Farag NA, Hassan Haredy H, AbdEl-Ghany AA, Mohamed KO. Design and synthesis of pyrazolo[3,4-d]pyrimidinone derivatives: Discovery of selective phosphodiesterase-5 inhibitors. Bioorg Med Chem Lett 2020; 30:127337. [DOI: 10.1016/j.bmcl.2020.127337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/01/2020] [Accepted: 06/06/2020] [Indexed: 11/25/2022]
|
19
|
Shekouhy M, Karimian S, Moaddeli A, Faghih Z, Delshad Y, Khalafi-Nezhad A. The synthesis and biological evaluation of nucleobases/tetrazole hybrid compounds: A new class of phosphodiesterase type 3 (PDE3) inhibitors. Bioorg Med Chem 2020; 28:115540. [PMID: 32503691 DOI: 10.1016/j.bmc.2020.115540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/22/2020] [Accepted: 04/29/2020] [Indexed: 01/12/2023]
Abstract
Spired by the chemical structure of Cilostazol, a selective phosphodiesterase 3A (PDE3A) inhibitor, several novel hybrid compounds of nucleobases (uracil, 6-azauracil, 2-thiuracil, adenine, guanine, theophylline and theobromine) and tetrazole were designed and successfully synthesized and their inhibitory effects on PDE3A as well as their cytotoxicity on HeLa and MCF-7 cancerous cell lines were studied. Obtained results show the linear correlation between the inhibitory effect of synthesized compounds and their cytotoxicity. In some cases, the PDE3A inhibitory effects of synthesized compounds are higher than the Cilostazol. Besides, compared to a standard anticancer drug methotrexate, some of the synthesized compounds showed the higher cytotoxicity against the HeLa and MCF-7 cancerous cell lines.
Collapse
Affiliation(s)
- Mohsen Shekouhy
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran.
| | - Somaye Karimian
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Moaddeli
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran; Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Zeinab Faghih
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yousef Delshad
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran
| | - Ali Khalafi-Nezhad
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 71454, Iran.
| |
Collapse
|
20
|
Wang X, Liu H, Pang M, Fu B, Yu X, He S, Tong J. Construction of a high-density genetic linkage map and mapping of quantitative trait loci for growth-related traits in silver carp (Hypophthalmichthys molitrix). Sci Rep 2019; 9:17506. [PMID: 31767872 PMCID: PMC6877629 DOI: 10.1038/s41598-019-53469-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/29/2019] [Indexed: 01/26/2023] Open
Abstract
High-density genetic map and quantitative trait loci (QTL) mapping are powerful tools for identifying genomic regions that may be responsible for such polygenic trait as growth. A high-density genetic linkage map was constructed by sequencing 198 individuals in a F1 family of silver carp (Hypophthalmichthys molitrix) in this study. This genetic map spans a length of 2,721.07 cM with 3,134 SNPs distributed on 24 linkage groups (LGs). Comparative genomic mapping presented a high level of syntenic relationship between silver carp and zebrafish. We detected one major and nineteen suggestive QTL for 4 growth-related traits (body length, body height, head length and body weight) at 6, 12 and 18 months post hatch (mph), explaining 10.2~19.5% of phenotypic variation. All six QTL for growth traits of 12 mph generally overlapped with QTL for 6 mph, while the majority of QTL for 18 mph were identified on two additional LGs, which may reveal a different genetic modulation during early and late muscle growth stages. Four potential candidate genes were identified from the QTL regions by homology searching of marker sequences against zebrafish genome. Hepcidin, a potential candidate gene identified from a QTL interval on LG16, was significantly associated with growth traits in the analyses of both phenotype-SNP association and mRNA expression between small-size and large-size groups of silver carp. These results provide a basis for elucidating the genetic mechanisms for growth and body formation in silver carp, a world aquaculture fish.
Collapse
Affiliation(s)
- Xinhua Wang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, China.,State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Haiyang Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Meixia Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Beide Fu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xiaomu Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Shunping He
- Key Laboratory of Aquatic Biodiversity and Conservation of the CAS, Institute of Hydrobiology, the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jingou Tong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
21
|
Nabavi SM, Talarek S, Listos J, Nabavi SF, Devi KP, Roberto de Oliveira M, Tewari D, Argüelles S, Mehrzadi S, Hosseinzadeh A, D'onofrio G, Orhan IE, Sureda A, Xu S, Momtaz S, Farzaei MH. Phosphodiesterase inhibitors say NO to Alzheimer's disease. Food Chem Toxicol 2019; 134:110822. [PMID: 31536753 DOI: 10.1016/j.fct.2019.110822] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/18/2022]
Abstract
Phosphodiesterases (PDEs) consisted of 11 subtypes (PDE1 to PDE11) and over 40 isoforms that regulate levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP), the second messengers in cell functions. PDE inhibitors (PDEIs) have been attractive therapeutic targets due to their involvement in diverse medical conditions, e.g. cardiovascular diseases, autoimmune diseases, Alzheimer's disease (AD), etc. Among them; AD with a complex pathology is a progressive neurodegenerative disorder which affect mostly senile people in the world and only symptomatic treatment particularly using cholinesterase inhibitors in clinic is available at the moment for AD. Consequently, novel treatment strategies towards AD are still searched extensively. Since PDEs are broadly expressed in the brain, PDEIs are considered to modulate neurodegenerative conditions through regulating cAMP and cGMP in the brain. In this sense, several synthetic or natural molecules inhibiting various PDE subtypes such as rolipram and roflumilast (PDE4 inhibitors), vinpocetine (PDE1 inhibitor), cilostazol and milrinone (PDE3 inhibitors), sildenafil and tadalafil (PDE5 inhibitors), etc have been reported showing encouraging results for the treatment of AD. In this review, PDE superfamily will be scrutinized from the view point of structural features, isoforms, functions and pharmacology particularly attributed to PDEs as target for AD therapy.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, 630003, Tamil Nadu, India.
| | - Marcos Roberto de Oliveira
- Departamento de Química (DQ), Instituto de Ciências Exatas e da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Cuiabá, Brazil.
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Grazia D'onofrio
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS "Casa Sollievo della Sofferenza", Viale Cappuccini 1, 71013, San Giovanni Rotondo, FG, Italy.
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands, CIBEROBN (Physiopathology of Obesity and Nutrition), E-07122, Palma de Mallorca, Balearic Islands, Spain.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA.
| | - Saeedeh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
22
|
Gul M, Serefoglu EC. An update on the drug safety of treating erectile dysfunction. Expert Opin Drug Saf 2019; 18:965-975. [DOI: 10.1080/14740338.2019.1659244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Murat Gul
- Department of Urology, Aksaray University School of Medicine, Aksaray, Turkey
- Laboratory of Reproductive Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ege Can Serefoglu
- Department of Urology, Bahceci Health Group, Istanbul, Turkey
- Department of Embriology and Histology, Medipol University, Istanbul, Turkey
| |
Collapse
|
23
|
Murkamilov IT, Aitbaev KA, Fomin VV, Murkamilova ZA, Bayzhigitova AA. Pentoxifylline and nephroprotection: effects on renal dysfunction and cardiovascular risks. TERAPEVT ARKH 2019; 91:95-100. [PMID: 31090379 DOI: 10.26442/00403660.2019.01.000037] [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: 11/22/2022]
Abstract
Generalized data on nephroprotective efficacy of pentoxifylline in chronic kidney disease (CKD) are presented. The potential of this drug in treating people suffering from CKD and cardiovascular diseases (CVD) with a high risk of developing the terminal stage of renal dysfunction is considered. Antiproteinuric, antifibrotic and anti-inflammatory effects of pentoxifylline significantly reduce the risk of progression of CKD and joining of CVD in the future. Efficacy in preventing the onset of the uremic stage of CKD, safety andapplicability at all stages of renal dysfunction development make pentoxifylline a very appealing drug not only for nephrologists but also for physicians. Keywords: chronic kidney disease, progression, pentoxifylline, nephroprotection, cardiovascular diseases.
Collapse
Affiliation(s)
- I T Murkamilov
- Kyrgyz State Medical Academy named after I.K. Akhunbaev, Bishkek, Kyrgyzstan.,Kyrgyz Russian Slavic University named after the First President of Russia B.N. Yeltsin, Bishkek, Kyrgyzstan
| | - K A Aitbaev
- Scientific Research Institute of Molecular Biology and Medicine, Bishkek, Kyrgyzstan
| | - V V Fomin
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian
| | | | - A A Bayzhigitova
- National Hospital under the Ministry of Health of the Kyrgyz Republic, Bishkek, Kyrgyzstan
| |
Collapse
|
24
|
Zaprinast diminished pain and enhanced opioid analgesia in a rat neuropathic pain model. Eur J Pharmacol 2018; 839:21-32. [PMID: 30213497 DOI: 10.1016/j.ejphar.2018.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/25/2018] [Accepted: 09/04/2018] [Indexed: 01/11/2023]
Abstract
The mechanism of neuropathic pain is complex and unclear. Based on our results, we postulate that an intensification of the kynurenine pathway occurs as a consequence of nerve injury. The G protein-coupled receptor 35 (GPR35) is important for kynurenine pathway activation. Cyclic GMP-specific phosphodiesterase inhibitors have also been shown to have beneficial effects on neuropathic pain. Therefore, the aims of our research were to elucidate how a substance that acts as both an agonist of GPR35 and an inhibitor of phosphodiesterase influences neuropathic pain in a rat model. Here, we demonstrated that preemptive and repeated intrathecal (i.t.) administration (16 h and 1 h before injury and then after nerve ligation daily for 7 days) of zaprinast (1 μg/5 μl) significantly attenuated mechanical (von Frey test) and thermal (cold plate test) hypersensitivity measured on day 7 after chronic constriction injury, and the effect of even a single injection lasted up to 24 h. Our data indicate that zaprinast diminished the number of IBA1-positive cells and consequently attenuated the levels of IL-1beta, IL-6, IL-18, and NOS2 in the lumbar spinal cord and/or dorsal root ganglia. Our results also demonstrated that zaprinast potentiated the analgesic properties of morphine and buprenorphine. In summary, in a neuropathic pain model, zaprinast significantly reduced pain symptoms and enhanced the effectiveness of opioids. Our data provide new evidence that modulation of both GPR35 and phosphodiesterase could be an important strategy for innovative pharmacological treatments designed to decrease hypersensitivity evoked by nerve injury.
Collapse
|
25
|
Peng T, Gong J, Jin Y, Zhou Y, Tong R, Wei X, Bai L, Shi J. Inhibitors of phosphodiesterase as cancer therapeutics. Eur J Med Chem 2018; 150:742-756. [PMID: 29574203 DOI: 10.1016/j.ejmech.2018.03.046] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/26/2018] [Accepted: 03/16/2018] [Indexed: 01/05/2023]
Abstract
Phosphodiesterases (PDEs) are a class of enzymes that hydrolyze cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) which is involved in many physiological processes including visual transduction, cell proliferation and differentiation, cell-cycle regulation, gene expression, inflammation, apoptosis, and metabolic function. PDEs are composed of 11 different families and each family contains different subtypes. The distribution, expression, regulation mode and sensitivity to inhibitors of each subtype are different, and they are involved in cancer, inflammation, asthma, depression, erectile dysfunction and other pathological processes of development. A large number of studies have shown that PDEs play an important role in the development of tumors by affecting the intracellular level of cAMP and/or cGMP and PDEs could become diagnostic markers or therapeutic targets. This review will give a brief overview of the expression and regulation of PDE families in the process of tumorigenesis and their anti-tumor inhibitors, which may guide the design of novel therapeutic drugs targeting PDEs for anticancer agent.
Collapse
Affiliation(s)
- Ting Peng
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun Gong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yongzhe Jin
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanping Zhou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xin Wei
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
26
|
Tajima T, Shinoda T, Urakawa N, Shimizu K, Kaneda T. Phosphodiesterase 9 (PDE9) regulates bovine tracheal smooth muscle relaxation. J Vet Med Sci 2018; 80:499-502. [PMID: 29386424 PMCID: PMC5880833 DOI: 10.1292/jvms.18-0011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The present study was designed to clarify phosphodiesterase 9 (PDE9) expression in bovine tracheal smooth muscle tissue, and to elucidate that PDE9 may contribute to the regulation of airway relaxation. PDE9 mRNA expression was detected in bovine tracheal smooth muscle. Sodium nitroprusside (an NO donor) and BAY 73-6691 (a selective PDE9 inhibitor) reduced high K+- and carbachol-induced contraction. BAY 73-6691 relaxed tracheal tissue on the same level with vardenafil (a selective PDE5 inhibitor). These results support our hypothesis that PDE9 plays functional role in the tracheal smooth muscle relaxation. PDE9 inhibitors are expected to be a novel target of the add-on treatment of airway hyperresponsiveness.
Collapse
Affiliation(s)
- Tsuyoshi Tajima
- Laboratory of Veterinary Pharmacology, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Tamami Shinoda
- Laboratory of Veterinary Pharmacology, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Norimoto Urakawa
- Laboratory of Veterinary Pharmacology, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Kazumasa Shimizu
- Laboratory of Veterinary Pharmacology, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Takeharu Kaneda
- Laboratory of Veterinary Pharmacology, Nippon Veterinary and Life Science University, 1-7-1 Kyounan-cho, Musashino-shi, Tokyo 180-8602, Japan
| |
Collapse
|
27
|
A Role for Phosphodiesterase 11A (PDE11A) in the Formation of Social Memories and the Stabilization of Mood. ADVANCES IN NEUROBIOLOGY 2018; 17:201-230. [PMID: 28956334 DOI: 10.1007/978-3-319-58811-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The most recently discovered 3',5'-cyclic nucleotide phosphodiesterase family is the Phosphodiesterase 11 (PDE11) family, which is encoded by a single gene PDE11A. PDE11A is a dual-specific PDE, breaking down both cAMP and cGMP. There are four PDE11A splice variants (PDE11A1-4) with distinct tissue expression profiles and unique N-terminal regulatory regions, suggesting that each isoform could be individually targeted with a small molecule or biologic. PDE11A4 is the PDE11A isoform expressed in brain and is found in the hippocampal formation of humans and rodents. Studies in rodents show that PDE11A4 mRNA expression in brain is, in fact, restricted to the hippocampal formation (CA1, possibly CA2, subiculum, and the adjacently connected amygdalohippocampal area). Within the hippocampal formation of rodents, PDE11A4 protein is expressed in neurons but not astrocytes, with a distribution across nuclear, cytoplasmic, and membrane compartments. This subcellular localization of PDE11A4 is altered in response to social experience in mouse, and in vitro studies show the compartmentalization of PDE11A4 is controlled, at least in part, by homodimerization and N-terminal phosphorylation. PDE11A4 expression dramatically increases in the hippocampus with age in the rodent hippocampus, from early postnatal life to late aging, suggesting PDE11A4 function may evolve across the lifespan. Interestingly, PDE11A4 protein shows a three to tenfold enrichment in the rodent ventral hippocampal formation (VHIPP; a.k.a. anterior in primates) versus dorsal hippocampal formation (DHIPP). Consistent with this enrichment in VHIPP, studies in knockout mice show that PDE11A regulates the formation of social memories and the stabilization of mood and is a critical mechanism by which social experience feeds back to modify the brain and subsequent social behaviors. PDE11A4 likely controls behavior by regulating hippocampal glutamatergic, oxytocin, and cytokine signaling, as well as protein translation. Given its unique tissue distribution and relatively selective effects on behavior, PDE11A may represent a novel therapeutic target for neuropsychiatric, neurodevelopmental, or age-related disorders. Therapeutically targeting PDE11A4 may be a way to selectively restore aberrant cyclic nucleotide signaling in the hippocampal formation while leaving the rest of the brain and periphery untouched, thus, relieving deficits while avoiding unwanted side effects.
Collapse
|
28
|
Laqqan M, Solomayer EF, Hammadeh M. Association between alterations in DNA methylation level of spermatozoa at CpGs dinucleotide and male subfertility problems. Andrologia 2017; 50. [DOI: 10.1111/and.12832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2017] [Indexed: 01/01/2023] Open
Affiliation(s)
- M. Laqqan
- Gynecology & Assisted Reproduction Laboratory; Department of Obstetrics; Saarland University; Saarland Germany
| | - E. F. Solomayer
- Gynecology & Assisted Reproduction Laboratory; Department of Obstetrics; Saarland University; Saarland Germany
| | - M. Hammadeh
- Gynecology & Assisted Reproduction Laboratory; Department of Obstetrics; Saarland University; Saarland Germany
| |
Collapse
|
29
|
Tan P, Liu L, Wei S, Tang Z, Yang L, Wei Q. The Effect of Oral Phosphodiesterase-5 Inhibitors on Sperm Parameters: A Meta-analysis and Systematic Review. Urology 2017; 105:54-61. [PMID: 28259808 DOI: 10.1016/j.urology.2017.02.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 02/10/2017] [Accepted: 02/20/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to evaluate the effect of phosphodiesterase-5 (PDE5) inhibitors on sperm parameters. METHODS A systematic literature search was performed. Mean value and its standard deviation (mean ± standard deviation) were used to perform quantitative analysis. Statistic heterogeneity scores were assessed with the standard Cochran Q test and I2 statistic. All statistical analyses were conducted by RevMan 5.3. RESULTS Eleven studies embraced a total of 1317 participants and 19 subgroups or trials were included. Acute administration of PDE5 inhibitors had no effect on semen volume (mean value [MD] = 0.26; 95% confidence interval [CI]: 0.00-0.48) and sperm concentration (MD = 2.04; 95%CI: -2.95 to 7.04). However, the percentage of motile spermatozoa (MD = 7.05; 95%CI: 2.59-11.51), the percentage of total progressive motility (MD = 6.23; 95%CI: 2.43-10.04), and rapid progressive motility (MD = 3.11; 95%CI: 0.23-5.99) were increased after oral PDE5 inhibitors treatment. Interestingly, these significant changes were observed only in infertile men but not in normal patients (MD = 6.89, P < .001 vs MD = 0.67, P = .71; MD = 6.64, P = .001 vs MD = 2.11, P > .05; and MD = 3.89, P = .04 vs MD = 0.92, P = .59, respectively). The percentage of morphologically normal spermatozoa also increased in infertile men (MD = 12.15; 95%CI: 5.16-19.15). Limited evidence showed the linearity, abnormal forms of spermatozoa, as well as reproductive hormones (total testosterone, free testosterone, luteinizing hormone, and follicle-stimulating hormone) did not benefit from PDE5 inhibitors treatment. CONCLUSION Oral PDE5 inhibitors treatment could modestly increase the sperm motility and morphology in infertile men.
Collapse
Affiliation(s)
- Ping Tan
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyou Wei
- Department of Cardiovascular and Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhuang Tang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Yang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Tetsi L, Charles AL, Paradis S, Lejay A, Talha S, Geny B, Lugnier C. Effects of cyclic nucleotide phosphodiesterases (PDEs) on mitochondrial skeletal muscle functions. Cell Mol Life Sci 2017; 74:1883-1893. [PMID: 28039524 PMCID: PMC11107545 DOI: 10.1007/s00018-016-2446-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022]
Abstract
Mitochondria play a critical role in skeletal muscle metabolism and function, notably at the level of tissue respiration, which conduct muscle strength as well as muscle survival. Pathological conditions induce mitochondria dysfunctions notably characterized by free oxygen radical production disturbing intracellular signaling. In that way, the second messengers, cyclic AMP and cyclic GMP, control intracellular signaling at the physiological and transcription levels by governing phosphorylation cascades. Both nucleotides are specifically and selectively hydrolyzed in their respective 5'-nucleotide by cyclic nucleotide phosphodiesterases (PDEs), which constitute a multi-genic family differently tissue distributed and subcellularly compartmentalized. These PDEs are presently recognized as therapeutic targets for cardiovascular, pulmonary, and neurologic diseases. However, very few data concerning cyclic nucleotides and PDEs in skeletal muscle, specifically in mitochondria, are reported in the literature. The knowledge of PDE implication in mitochondrial signaling would be helpful for resolving critical mitochondrial dysfunctions in skeletal muscle.
Collapse
Affiliation(s)
- Liliane Tetsi
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Anne-Laure Charles
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Stéphanie Paradis
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Anne Lejay
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Samy Talha
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Bernard Geny
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Claire Lugnier
- EA 3072 "Mitochondrie, Stress Oxydant et Protection Musculaire", Fédération de Médecine Translationnelle, Faculté de Médecine, Institut de Physiologie, Université de Strasbourg, 4, Rue Kirschleger, 67085, Strasbourg Cedex, France.
| |
Collapse
|
31
|
Mackenzie AE, Milligan G. The emerging pharmacology and function of GPR35 in the nervous system. Neuropharmacology 2017; 113:661-671. [DOI: 10.1016/j.neuropharm.2015.07.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/20/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
|
32
|
Snyder GL, Vanover KE. PDE Inhibitors for the Treatment of Schizophrenia. ADVANCES IN NEUROBIOLOGY 2017; 17:385-409. [DOI: 10.1007/978-3-319-58811-7_14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Identification of lead BAY60-7550 analogues as potential inhibitors that utilize the hydrophobic groove in PDE2A: a molecular dynamics simulation study. J Mol Model 2016; 23:7. [PMID: 27966018 DOI: 10.1007/s00894-016-3171-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/16/2016] [Indexed: 12/22/2022]
Abstract
The phosphodiesterase (PDE) family of proteins are important regulators of signal transduction, which they achieve by controlling the secondary messengers cyclic AMP (cAMP) and cyclic GMP (cGMP). cAMP and cGMP are involved in many critical intracellular processes such as gene transcription, kinase activation, signal transduction in learning and memory, and channel function as secondary messengers. The involvement of PDEs in neuronal communication has made them important therapeutic targets. Considering the recent discovery that PDE2A inhibition can improve cognitive functioning, a combined molecular dynamics simulation and scoring and docking study was carried out to identify selective inhibitors of PDE2A that specifically interact with the recently discovered hydrophobic groove in PDE2A. Using the X-ray crystal structure of PDE2A (from PDB ID: 4HTX), we investigated the binding modes of a range of promising inhibitors based on the known PDE2A inhibitor BAY60-7550 to PDE2A. Graphical abstract The lead molecule showing highest MMPBSA binding energy with 2D and 3D binding pose in hydrophobic groove.
Collapse
|
34
|
Kokkonen K, Kass DA. Nanodomain Regulation of Cardiac Cyclic Nucleotide Signaling by Phosphodiesterases. Annu Rev Pharmacol Toxicol 2016; 57:455-479. [PMID: 27732797 DOI: 10.1146/annurev-pharmtox-010716-104756] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) form an 11-member superfamily comprising 100 different isoforms that regulate the second messengers cyclic adenosine or guanosine 3',5'-monophosphate (cAMP or cGMP). These PDE isoforms differ with respect to substrate selectivity and their localized control of cAMP and cGMP within nanodomains that target specific cellular pools and synthesis pathways for the cyclic nucleotides. Seven PDE family members are physiologically relevant to regulating cardiac function, disease remodeling of the heart, or both: PDE1 and PDE2, both dual-substrate (cAMP and cGMP) esterases; PDE3, PDE4, and PDE8, which principally hydrolyze cAMP; and PDE5A and PDE9A, which target cGMP. New insights regarding the different roles of PDEs in health and disease and their local signaling control are broadening the potential therapeutic utility for PDE-selective inhibitors. In this review, we discuss these PDEs, focusing on the different mechanisms by which they control cardiac function in health and disease by regulating intracellular nanodomains.
Collapse
Affiliation(s)
- Kristen Kokkonen
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; .,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
35
|
D'Andrea MR, Qiu Y, Haynes-Johnson D, Bhattacharjee S, Kraft P, Lundeen S. Expression of PDE11A in Normal and Malignant Human Tissues. J Histochem Cytochem 2016; 53:895-903. [PMID: 15995148 DOI: 10.1369/jhc.5a6625.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cyclic nucleotide phosphodiesterase 11A (PDE11A) is the newest member in the PDE family. Although the tissue distribution of PDE11A mRNA has been shown, its protein expression pattern has not been well studied. The goal of this report is to investigate the distribution of PDE11A proteins in a wide range of normal and malignant human tissues. We utilized a polyclonal antibody that recognized all four PDE11A isoforms. Its specificity was demonstrated by Western blot analysis on a recombinant human PDE11A protein and native PDE11A proteins in various human tissues. Immunohistochemistry showed that PDE11A is widely expressed. Various degrees of immunoreactivity were observed in the epithelial cells, endothelial cells, and smooth muscle cells of all tissues examined. The highest expression was in the epithelial, endothelial, and smooth muscle cells of the prostate, Leydig, and spermatogenic cells of the testis, the tubule epithelial cells in the kidney, the epithelial and endothelial cells in the adrenal, the epithelial cells and macrophages in the colon, and the epidermis in the skin. Furthermore, PDE11A expression was also detected in several human carcinomas. Our results suggest that PDE11A might be involved in multiple physiological processes in various organs via its ability to modulate intracellular cAMP and cGMP levels.
Collapse
Affiliation(s)
- Michael R D'Andrea
- Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development, LLC, 1000 Route 202, Raritan, NJ 08869, USA
| | | | | | | | | | | |
Collapse
|
36
|
Gross I, Durner J. In Search of Enzymes with a Role in 3', 5'-Cyclic Guanosine Monophosphate Metabolism in Plants. FRONTIERS IN PLANT SCIENCE 2016; 7:576. [PMID: 27200049 PMCID: PMC4858519 DOI: 10.3389/fpls.2016.00576] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/14/2016] [Indexed: 05/07/2023]
Abstract
In plants, nitric oxide (NO)-mediated 3', 5'-cyclic guanosine monophosphate (cGMP) synthesis plays an important role during pathogenic stress response, stomata closure upon osmotic stress, the development of adventitious roots and transcript regulation. The NO-cGMP dependent pathway is well characterized in mammals. The binding of NO to soluble guanylate cyclase enzymes (GCs) initiates the synthesis of cGMP from guanosine triphosphate. The produced cGMP alters various cellular responses, such as the function of protein kinase activity, cyclic nucleotide gated ion channels and cGMP-regulated phosphodiesterases. The signal generated by the second messenger is terminated by 3', 5'-cyclic nucleotide phosphodiesterase (PDEs) enzymes that hydrolyze cGMP to a non-cyclic 5'-guanosine monophosphate. To date, no homologues of mammalian cGMP-synthesizing and degrading enzymes have been found in higher plants. In the last decade, six receptor proteins from Arabidopsis thaliana have been reported to have guanylate cyclase activity in vitro. Of the six receptors, one was shown to be a NO dependent guanylate cyclase enzyme (NOGC1). However, the role of these proteins in planta remains to be elucidated. Enzymes involved in the degradation of cGMP remain elusive, albeit, PDE activity has been detected in crude protein extracts from various plants. Additionally, several research groups have partially purified and characterized PDE enzymatic activity from crude protein extracts. In this review, we focus on presenting advances toward the identification of enzymes involved in the cGMP metabolism pathway in higher plants.
Collapse
Affiliation(s)
- Inonge Gross
- Nitric Oxide Production and Signalling Group, Institute of Biochemical Plant Pathology, Helmholtz Center MunichGermany
- *Correspondence: Inonge Gross,
| | - Jörg Durner
- Nitric Oxide Production and Signalling Group, Institute of Biochemical Plant Pathology, Helmholtz Center MunichGermany
- Chair of Biochemical Plant Pathology, Technische Universität München, FreisingGermany
| |
Collapse
|
37
|
Trans effects of chromosome aneuploidies on DNA methylation patterns in human Down syndrome and mouse models. Genome Biol 2015; 16:263. [PMID: 26607552 PMCID: PMC4659173 DOI: 10.1186/s13059-015-0827-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 11/09/2015] [Indexed: 11/18/2022] Open
Abstract
Background Trisomy 21 causes Down syndrome (DS), but the mechanisms by which the extra chromosome leads to deficient intellectual and immune function are not well understood. Results Here, we profile CpG methylation in DS and control cerebral and cerebellar cortex of adults and cerebrum of fetuses. We purify neuronal and non-neuronal nuclei and T lymphocytes and find biologically relevant genes with DS-specific methylation (DS-DM) in each of these cell types. Some genes show brain-specific DS-DM, while others show stronger DS-DM in T cells. Both 5-methyl-cytosine and 5-hydroxy-methyl-cytosine contribute to the DS-DM. Thirty percent of genes with DS-DM in adult brain cells also show DS-DM in fetal brains, indicating early onset of these epigenetic changes, and we find early maturation of methylation patterns in DS brain and lymphocytes. Some, but not all, of the DS-DM genes show differential expression. DS-DM preferentially affected CpGs in or near specific transcription factor binding sites (TFBSs), implicating a mechanism involving altered TFBS occupancy. Methyl-seq of brain DNA from mouse models with sub-chromosomal duplications mimicking DS reveals partial but significant overlaps with human DS-DM and shows that multiple chromosome 21 genes contribute to the downstream epigenetic effects. Conclusions These data point to novel biological mechanisms in DS and have general implications for trans effects of chromosomal duplications and aneuploidies on epigenetic patterning. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0827-6) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Ioki S, Wu QS, Takayama O, Motohashi HH, Wakai T, Funahashi H. A phosphodiesterase type-5 inhibitor, sildenafil, induces sperm capacitation and penetration into porcine oocytes in a chemically defined medium. Theriogenology 2015; 85:428-33. [PMID: 26443234 DOI: 10.1016/j.theriogenology.2015.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/04/2015] [Accepted: 09/04/2015] [Indexed: 01/21/2023]
Abstract
The present study was undertaken to determine the effect of a phosphodiesterase (PDE) type-5 (cyclic guanosine monophosphate-specific) inhibitor, sildenafil, on capacitation and penetration of boar spermatozoa in a basic chemically defined medium (adenosine- and theophylline-free PGM-tac4). When ejaculated spermatozoa were cultured for 90 minutes in the absence or presence of sildenafil at 2.5 mM, the inhibitor significantly increased the percentage of capacitated/acrosome-reacted spermatozoa, as a result of the chlortetracycline assay. When fresh spermatozoa were co-cultured with oocytes in the presence of sildenafil at a different concentration (0, 2.5, 25, or 250 μM), higher sildenafil concentrations (25 and 250 μM) significantly resulted in higher sperm penetration rates. When oocytes matured in vitro were co-cultured with spermatozoa in the presence of 25 μM sildenafil or 25 mM caffeine benzoate for 8 hours, the incidence of penetrated oocytes did not differ between two groups, whereas the incidence of monospermic oocytes in penetrated one was significantly higher in the presence of sildenafil. Immunocytochemical analysis reported the presence of PDE type-5 on the acrosome region of boar spermatozoa. These results report that regulation of cyclic guanosine monophosphate-specific PDE type-5 by sildenafil somehow can increase the penetrability of boar spermatozoa in vitro.
Collapse
Affiliation(s)
- Sumire Ioki
- Department of Animal Science, Okayama University, Okayama, Japan
| | - Qing-Shan Wu
- Department of Animal Science, Okayama University, Okayama, Japan
| | - Osamu Takayama
- Assisted Reproductive Technology Center, Okayama University, Okayama, Japan
| | | | - Takuya Wakai
- Department of Animal Science, Okayama University, Okayama, Japan
| | - Hiroaki Funahashi
- Department of Animal Science, Okayama University, Okayama, Japan; Assisted Reproductive Technology Center, Okayama University, Okayama, Japan.
| |
Collapse
|
39
|
de Alexandre RB, Horvath AD, Szarek E, Manning AD, Leal LF, Kardauke F, Epstein JA, Carraro DM, Soares FA, Apanasovich TV, Stratakis CA, Faucz FR. Phosphodiesterase sequence variants may predispose to prostate cancer. Endocr Relat Cancer 2015; 22:519-30. [PMID: 25979379 PMCID: PMC4499475 DOI: 10.1530/erc-15-0134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 12/11/2022]
Abstract
We hypothesized that mutations that inactivate phosphodiesterase (PDE) activity and lead to increased cAMP and cyclic guanosine monophosphate levels may be associated with prostate cancer (PCa). We sequenced the entire PDE coding sequences in the DNA of 16 biopsy samples from PCa patients. Novel mutations were confirmed in the somatic or germline state by Sanger sequencing. Data were then compared to the 1000 Genome Project. PDE, CREB and pCREB protein expression was also studied in all samples, in both normal and abnormal tissue, by immunofluorescence. We identified three previously described PDE sequence variants that were significantly more frequent in PCa. Four novel sequence variations, one each in the PDE4B,PDE6C, PDE7B and PDE10A genes, respectively, were also found in the PCa samples. Interestingly, PDE10A and PDE4B novel variants that were present in 19 and 6% of the patients were found in the tumor tissue only. In patients carrying PDE defects, there was pCREB accumulation (P<0.001), and an increase of the pCREB:CREB ratio (patients 0.97±0.03; controls 0.52±0.03; P-value <0.001) by immunohistochemical analysis. We conclude that PDE sequence variants may play a role in the predisposition and/or progression to PCa at the germline and/or somatic state respectively.
Collapse
Affiliation(s)
- Rodrigo B de Alexandre
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Anelia D Horvath
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Eva Szarek
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Allison D Manning
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Leticia F Leal
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Fabio Kardauke
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Jonathan A Epstein
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Dirce M Carraro
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Fernando A Soares
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Tatiyana V Apanasovich
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Constantine A Stratakis
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| | - Fabio R Faucz
- Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA Section on Endocrinology and GeneticsProgram on Developmental Endocrinology and Genetics (PDEGEN) and Pediatric Endocrinology Inter-institute Training Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USASchool of Health and BiosciencesPontifícia Universidade Católica do Paraná (PUCPR), Curitiba, PR 80215-901, BrazilDepartment of Pharmacology and PhysiologyGeorge Washington University, Washington, DC 20037, USALaboratory of Genomics and Molecular BiologyCIPEDepartment of PathologyA.C. Camargo Cancer Center, 01509-010 São Paulo, SP, BrazilDepartment of StatisticsGeorge Washington University, Washington, DC 20037, USA
| |
Collapse
|
40
|
|
41
|
Sokolowska M, Chen LY, Liu Y, Martinez-Anton A, Qi HY, Logun C, Alsaaty S, Park YH, Kastner DL, Chae JJ, Shelhamer JH. Prostaglandin E2 Inhibits NLRP3 Inflammasome Activation through EP4 Receptor and Intracellular Cyclic AMP in Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 194:5472-5487. [PMID: 25917098 DOI: 10.4049/jimmunol.1401343] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 03/24/2015] [Indexed: 01/13/2023]
Abstract
PGE2 is a potent lipid mediator involved in maintaining homeostasis but also promotion of acute inflammation or immune suppression in chronic inflammation and cancer. Nucleotide-binding domain, leucine-rich repeat-containing protein (NLR)P3 inflammasome plays an important role in host defense. Uncontrolled activation of the NLRP3 inflammasome, owing to mutations in the NLRP3 gene, causes cryopyrin-associated periodic syndromes. In this study, we showed that NLRP3 inflammasome activation is inhibited by PGE2 in human primary monocyte-derived macrophages. This effect was mediated through PGE2 receptor subtype 4 (EP4) and an increase in intracellular cAMP, independently of protein kinase A or exchange protein directly activated by cAMP. A specific agonist of EP4 mimicked, whereas its antagonist or EP4 knockdown reversed, PGE2-mediated NLRP3 inhibition. PGE2 caused an increase in intracellular cAMP. Blockade of adenylate cyclase by its inhibitor reversed PGE2-mediated NLRP3 inhibition. Increase of intracellular cAMP by an activator of adenylate cyclase or an analog of cAMP, or a blockade of cAMP degradation by phosphodiesterase inhibitor decreased NLRP3 activation. Protein kinase A or exchange protein directly activated by cAMP agonists did not mimic, and their antagonists did not reverse, PGE2-mediated NLRP3 inhibition. Additionally, constitutive IL-1β secretion from LPS-primed PBMCs of cryopyrin-associated periodic fever syndromes patients was substantially reduced by high doses of PGE2. Moreover, blocking cytosolic phospholipase A2α by its inhibitor or small interfering RNA or inhibiting cyclooxygenase 2, resulting in inhibition of endogenous PGE2 production, caused an increase in NLRP3 inflammasome activation. Our results suggest that PGE2 might play a role in maintaining homeostasis during the resolution phase of inflammation and might serve as an autocrine and paracrine regulator.
Collapse
Affiliation(s)
- Milena Sokolowska
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Li-Yuan Chen
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Yueqin Liu
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | | | - Hai-Yan Qi
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Carolea Logun
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Sara Alsaaty
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| | - Yong Hwan Park
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Jae Jin Chae
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - James H Shelhamer
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD, USA
| |
Collapse
|
42
|
Davari AS, Abnous K, Mehri S, Ghandadi M, Hadizadeh F. Synthesis and biological evaluation of novel pyridine derivatives as potential anticancer agents and phosphodiesterase-3 inhibitors. Bioorg Chem 2014; 57:83-89. [DOI: 10.1016/j.bioorg.2014.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/28/2014] [Accepted: 09/08/2014] [Indexed: 01/04/2023]
|
43
|
Otero C, Peñaloza JP, Rodas PI, Fernández-Ramires R, Velasquez L, Jung JE. Temporal and spatial regulation of cAMP signaling in disease: role of cyclic nucleotide phosphodiesterases. Fundam Clin Pharmacol 2014; 28:593-607. [PMID: 24750474 DOI: 10.1111/fcp.12080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/28/2014] [Accepted: 04/17/2014] [Indexed: 01/19/2023]
Abstract
Since its discovery, cAMP has been proposed as one of the most versatile second messengers. The remarkable feature of cAMP to tightly control highly diverse physiological processes, including metabolism, homeostasis, secretion, muscle contraction, cell proliferation and migration, immune response, and gene transcription, is reflected by millions of different articles worldwide. Compartmentalization of cAMP in space and time, maintained by mainly phosphodiesterases, contributes to the maintenance of equilibrium inside the cell where one signal can trigger many different events. Novel cAMP sensors seem to carry out certain unexpected signaling properties of cAMP and thereby to permit delicate adaptations of biologic responses. Measuring space and time events with biosensors will increase our current knowledge on the pathophysiology of diseases, such as chronic obstructive pulmonary disease, asthma, cognitive impairment, cancer, and renal and heart failure. Further insights into the cAMP dynamics will help to optimize the pharmacological treatment for these diseases.
Collapse
Affiliation(s)
- Carolina Otero
- Center for Integrative Medicine and Innovative Science, Universidad Andres Bello, Santiago, Chile; Centro para el Desarrollo de la Nanociencia y Nanotecnologia, Santiago, Chile
| | | | | | | | | | | |
Collapse
|
44
|
Azevedo MF, Faucz FR, Bimpaki E, Horvath A, Levy I, de Alexandre RB, Ahmad F, Manganiello V, Stratakis CA. Clinical and molecular genetics of the phosphodiesterases (PDEs). Endocr Rev 2014; 35:195-233. [PMID: 24311737 PMCID: PMC3963262 DOI: 10.1210/er.2013-1053] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 11/06/2013] [Indexed: 12/31/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are enzymes that have the unique function of terminating cyclic nucleotide signaling by catalyzing the hydrolysis of cAMP and GMP. They are critical regulators of the intracellular concentrations of cAMP and cGMP as well as of their signaling pathways and downstream biological effects. PDEs have been exploited pharmacologically for more than half a century, and some of the most successful drugs worldwide today affect PDE function. Recently, mutations in PDE genes have been identified as causative of certain human genetic diseases; even more recently, functional variants of PDE genes have been suggested to play a potential role in predisposition to tumors and/or cancer, especially in cAMP-sensitive tissues. Mouse models have been developed that point to wide developmental effects of PDEs from heart function to reproduction, to tumors, and beyond. This review brings together knowledge from a variety of disciplines (biochemistry and pharmacology, oncology, endocrinology, and reproductive sciences) with emphasis on recent research on PDEs, how PDEs affect cAMP and cGMP signaling in health and disease, and what pharmacological exploitations of PDEs may be useful in modulating cyclic nucleotide signaling in a way that prevents or treats certain human diseases.
Collapse
Affiliation(s)
- Monalisa F Azevedo
- Section on Endocrinology Genetics (M.F.A., F.R.F., E.B., A.H., I.L., R.B.d.A., C.A.S.), Program on Developmental Endocrinology Genetics, Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland 20892; Section of Endocrinology (M.F.A.), University Hospital of Brasilia, Faculty of Medicine, University of Brasilia, Brasilia 70840-901, Brazil; Group for Advanced Molecular Investigation (F.R.F., R.B.d.A.), Graduate Program in Health Science, Medical School, Pontificia Universidade Catolica do Paraná, Curitiba 80215-901, Brazil; Cardiovascular Pulmonary Branch (F.A., V.M.), National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland 20892; and Pediatric Endocrinology Inter-Institute Training Program (C.A.S.), NICHD, NIH, Bethesda, Maryland 20892
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The cyclic nucleotide cAMP is a universal regulator of a variety of cell functions in response to activated G-protein coupled receptors. In particular, cAMP exerts positive or negative effects on cell proliferation in different cell types. As demonstrated by several in vitro studies, in somatotrophs and in other endocrine cells, cAMP is a mitogenic factor. In agreement with this notion, it has been found that the mutations of genes coding for proteins that contribute to increases in the cAMP signaling cascade may cause endocrine tumor development. This review will discuss the central role of cAMP signaling in the pituitary, focusing on the cAMP pathway alterations involved in pituitary tumorigenesis, as well as on poorly investigated the aspects of cAMP cascade, such as crosstalk with the ERK signaling pathway and new cAMP effectors.
Collapse
Affiliation(s)
- Erika Peverelli
- Endocrine Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Endocrine Unit, IRCCS Humanitas Clinical Institute, Rozzano, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
46
|
Ückert S, Kuczyk MA, Oelke M. Phosphodiesterase inhibitors in clinical urology. Expert Rev Clin Pharmacol 2014; 6:323-32. [DOI: 10.1586/ecp.13.16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
47
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol 2013; 170:1797-867. [PMID: 24528243 PMCID: PMC3892293 DOI: 10.1111/bph.12451] [Citation(s) in RCA: 415] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Enzymes are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, catalytic receptors and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
48
|
Cichero E, D'Ursi P, Moscatelli M, Bruno O, Orro A, Rotolo C, Milanesi L, Fossa P. Homology Modeling, Docking Studies and Molecular Dynamic Simulations Using Graphical Processing Unit Architecture to Probe the Type-11 Phosphodiesterase Catalytic Site: A Computational Approach for the Rational Design of Selective Inhibitors. Chem Biol Drug Des 2013; 82:718-31. [DOI: 10.1111/cbdd.12193] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 06/14/2013] [Accepted: 07/09/2013] [Indexed: 01/21/2023]
Affiliation(s)
- Elena Cichero
- Dipartimento di Farmacia; Sezione di Chimica del Farmaco e del Prodotto Cosmetico; Università degli Studi di Genova; Viale Benedetto XV 3 16132 Genova Italy
| | - Pasqualina D'Ursi
- Institute for Biomedical Technologies-National Research Council (ITB-CNR); Via Fratelli Cervi 93 20090 Segrate (MI) Italy
| | - Marco Moscatelli
- Institute for Biomedical Technologies-National Research Council (ITB-CNR); Via Fratelli Cervi 93 20090 Segrate (MI) Italy
- University of Milano-Bicocca; DISAT; Piazza della Scienza 1 20126 Milan Italy
| | - Olga Bruno
- Dipartimento di Farmacia; Sezione di Chimica del Farmaco e del Prodotto Cosmetico; Università degli Studi di Genova; Viale Benedetto XV 3 16132 Genova Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies-National Research Council (ITB-CNR); Via Fratelli Cervi 93 20090 Segrate (MI) Italy
| | - Chiara Rotolo
- Dipartimento di Farmacia; Sezione di Chimica del Farmaco e del Prodotto Cosmetico; Università degli Studi di Genova; Viale Benedetto XV 3 16132 Genova Italy
| | - Luciano Milanesi
- Institute for Biomedical Technologies-National Research Council (ITB-CNR); Via Fratelli Cervi 93 20090 Segrate (MI) Italy
| | - Paola Fossa
- Dipartimento di Farmacia; Sezione di Chimica del Farmaco e del Prodotto Cosmetico; Università degli Studi di Genova; Viale Benedetto XV 3 16132 Genova Italy
| |
Collapse
|
49
|
Demirbas D, Wyman AR, Shimizu-Albergine M, Cakici O, Beavo JA, Hoffman CS. A yeast-based chemical screen identifies a PDE inhibitor that elevates steroidogenesis in mouse Leydig cells via PDE8 and PDE4 inhibition. PLoS One 2013; 8:e71279. [PMID: 23967182 PMCID: PMC3743849 DOI: 10.1371/journal.pone.0071279] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/04/2013] [Indexed: 02/01/2023] Open
Abstract
A cell-based high-throughput screen (HTS) was developed to detect phosphodiesterase 8 (PDE8) and PDE4/8 combination inhibitors. By replacing the Schizosaccharomyces pombe PDE gene with the murine PDE8A1 gene in strains lacking adenylyl cyclase, we generated strains whose protein kinase A (PKA)-stimulated growth in 5-fluoro orotic acid (5FOA) medium reflects PDE8 activity. From our previously-identified PDE4 and PDE7 inhibitors, we identified a PDE4/8 inhibitor that allowed us to optimize screening conditions. Of 222,711 compounds screened, ∼0.2% displayed composite Z scores of >20. Additional yeast-based assays using the most effective 367 compounds identified 30 candidates for further characterization. Among these, compound BC8-15 displayed the lowest IC₅₀ value for both PDE4 and PDE8 inhibition in in vitro enzyme assays. This compound also displays significant activity against PDE10A and PDE11A. BC8-15 elevates steroidogenesis in mouse Leydig cells as a single pharmacological agent. Assays using BC8-15 and two structural derivatives support a model in which PDE8 is a primary regulator of testosterone production by Leydig cells, with an additional role for PDE4 in this process. BC8-15, BC8-15A, and BC8-15C, which are commercially available compounds, display distinct patterns of activity against PDE4, PDE8, PDE10A, and PDE11A, representing a chemical toolkit that could be used to examine the biological roles of these enzymes in cell culture systems.
Collapse
Affiliation(s)
- Didem Demirbas
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Arlene R. Wyman
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Masami Shimizu-Albergine
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Ozgur Cakici
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
| | - Joseph A. Beavo
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Charles S. Hoffman
- Biology Department, Boston College, Chestnut Hill, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
50
|
Shi F, Perez E, Wang T, Peitz B, Lapolt PS. Stage- and Cell-Specific Expression of Soluble Guanylyl Cyclase Alpha and Beta Subunits, cGMP-Dependent Protein Kinase I Alpha and Beta, and Cyclic Nucleotide-Gated Channel Subunit 1 in the Rat Testis. ACTA ACUST UNITED AC 2013; 26:258-63. [PMID: 15713832 DOI: 10.1002/j.1939-4640.2005.tb01093.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several studies suggest that nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) modulate testicular function. In this study, we examined the expression of cGMP-dependent protein kinase G-I (PKG-I), and cyclic nucleotide-gated channel 1 (CNG-1), 2 known mediators of cGMP action, and the expression of soluble guanylyl cyclase (sGC) subunits in the rat testis. Immunohistochemical analysis revealed that the alpha subunit of sGC was expressed in the blood vessels and Leydig cells of adult rat testes. In addition, the sGC alpha subunit was observed in the acrosomal structures of spermatids undergoing the middle and later stages of spermiogenesis, but not in mature spermatozoa. Similar localization and expression patterns were seen for the sGC beta subunit, indicating coexpression of the sGC subunits. PKG-I was expressed in blood vessels and in the acrosomal region of spermatids during the early and middle stages of spermiogenesis but was not observed in Leydig cells or in mature spermatozoa. In contrast to sGC and PKG-I, CNG-1 was expressed only in cytoplasm and the residual bodies of late-stage (17-19) spermatids, with no staining observed in blood vessels and Leydig cells. These results demonstrate that sGC, PKG-I, and CNG-1 are expressed in a stage- and cell-specific manner in the rat testis. The distinct temporal patterns of expression of these components of cGMP signaling pathways suggest different physiological roles for sGC, PKG-I, and CNG-1 in spermiogenesis and steroidogenesis.
Collapse
Affiliation(s)
- Fangxiong Shi
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing, 210095 China.
| | | | | | | | | |
Collapse
|