1
|
Zhang C, Xue ZH, Luo WH, Jiang MY, Wu Y. The therapeutic potential of phosphodiesterase 9 (PDE9) inhibitors: a patent review (2018-present). Expert Opin Ther Pat 2024; 34:759-772. [PMID: 38979973 DOI: 10.1080/13543776.2024.2376632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION Phosphodiesterase 9 (PDE9) has been demonstrated as a potential target for neurological disorders and cardiovascular diseases, such as Alzheimer's disease and heart failure. For the last few years, a series of PDE9 inhibitors with structural diversities have been developed and patented by researchers and pharmaceutical companies, providing insights into first-in-class therapies of PDE9 drug candidates. AREA COVERED This review provides an overview of PDE9 inhibitors in patents from 2018 to the present. EXPERT OPINION Only a few of the current PDE9 inhibitors are highly selective over other PDEs, which limits their application in pharmacological and clinical research. The design and development of highly selective PDE9 inhibitors remain the top priority in future research. The advantages of targeting PDE9 rather than other PDEs in treating neurodegenerative diseases need to be explained thoroughly. Besides, application of PDE9 inhibitor-based combination therapies sheds light on treating diabetes and refractory heart diseases. Finally, PDE9 inhibitors should be further explored in clinical indications beyond neurological disorders and cardiovascular diseases.
Collapse
Affiliation(s)
- Chen Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Zhao-Hang Xue
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Wei-Hao Luo
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Mei-Yan Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Yinuo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
2
|
Singh N, Singh AK. In Silico Structural Modeling and Binding Site Analysis of Cerebroside Sulfotransferase (CST): A Therapeutic Target for Developing Substrate Reduction Therapy for Metachromatic Leukodystrophy. ACS OMEGA 2024; 9:10748-10768. [PMID: 38463293 PMCID: PMC10918841 DOI: 10.1021/acsomega.3c09462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 03/12/2024]
Abstract
Cerebroside sulfotransferase (CST) is emerging as an important therapeutic target to develop substrate reduction therapy (SRT) for metachromatic leukodystrophy (MLD), a rare neurodegenerative lysosomal storage disorder. MLD develops with progressive impairment and destruction of the myelin sheath as a result of accumulation of sulfatide around the nerve cells in the absence of its recycling mechanism with deficiency of arylsulfatase A (ARSA). Sulfatide is the product of the catalytic action of cerebroside sulfotransferase (CST), which needs to be regulated under pathophysiological conditions by inhibitor development. To carry out in silico-based preliminary drug screening or for designing new drug candidates, a high-quality three-dimensional (3D) structure is needed in the absence of an experimentally derived three-dimensional crystal structure. In this study, a 3D model of the protein was developed using a primary sequence with the SWISS-MODEL server by applying the top four GMEQ score-based templates belonging to the sulfotransferase family as a reference. The 3D model of CST highlights the features of the protein responsible for its catalytic action. The CST model comprises five β-strands, which are flanked by ten α-helices from both sides as well as form the upside cover of the catalytic pocket of CST. CST has two catalytic regions: PAPS (-sulfo donor) binding and galactosylceramide (-sulfo acceptor) binding. The catalytic action of CST was proposed via molecular docking and molecular dynamic (MD) simulation with PAPS, galactosylceramide (GC), PAPS-galactosylceramide, and PAP. The stability of the model and its catalytic action were confirmed using molecular dynamic simulation-based trajectory analysis. CST response against the inhibition potential of the experimentally reported competitive inhibitor of CST was confirmed via molecular docking and molecular dynamics simulation, which suggested the suitability of the CST model for future drug discovery to strengthen substrate reduction therapy for MLD.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Dravyaguna,
Faculty of Ayurveda, Institute of Medical
Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Anil Kumar Singh
- Department of Dravyaguna,
Faculty of Ayurveda, Institute of Medical
Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
3
|
Zheng L, Zhou ZZ. An overview of phosphodiesterase 9 inhibitors: Insights from skeletal structure, pharmacophores, and therapeutic potential. Eur J Med Chem 2023; 259:115682. [PMID: 37536210 DOI: 10.1016/j.ejmech.2023.115682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 08/05/2023]
Abstract
Cyclic nucleotide phosphodiesterase 9 (PDE9), a specifically hydrolytic enzyme with the highest affinity for cyclic guanosine monophosphate (cGMP) among the phosphodiesterases family, plays a critical role in many biological processes. Consequently, the development of PDE9 inhibitors has received increasing attention in recent years, with several compounds undergoing clinical trials for the treatment of central nervous system (CNS) diseases such as Alzheimer's disease, schizophrenia, and psychotic disorders, as well as heart failure and sickle cell disease. This review analyzes the recent primary literatures and patents published from 2004 to 2023, focusing on the structure, pharmacophores, selectivity, and therapeutic potential of PDE9 inhibitors. It hoped to provide a comprehensive overview of the field's current state to inform the development of novel PDE9 inhibitors.
Collapse
Affiliation(s)
- Lei Zheng
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhong-Zhen Zhou
- Innovation Program of Drug Research on Neurological and Metabolic Diseases, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Pyrrolotriazinone as an Underexplored Scaffold in Drug Discovery. Pharmaceuticals (Basel) 2021; 14:ph14121275. [PMID: 34959675 PMCID: PMC8705011 DOI: 10.3390/ph14121275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Heterocyclic amino derivatives have been extensively synthesized and validated as potent bioactive compounds, and nowadays, numerous marketed drugs share these scaffolds, from very simple structures (monoamino, monocyclic compounds) to much more complex molecules (polycyclic derivatives with two or more nitrogen atoms within the (fused) rings). In a constant quest for new chemical entities in drug discovery, a few novel heterocycles have emerged in recent years as promising building blocks for the obtainment of bioactive modulators. In this context, pyrrolotriazinones have attracted attention, and some show promising biological activities. Here, we offer an extensive review of pyrrolo[2,1-f][1,2,4]triazin-4(1H)-one and pyrrolo[1,2-d][1,2,4]triazin-4(3H)-one, describing their biological properties en route to drug discovery.
Collapse
|
5
|
Diversifying the xanthine scaffold for potential phosphodiesterase 9A inhibitors: synthesis and validation. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02722-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
6
|
Susmi TF, Rahman A, Khan MMR, Yasmin F, Islam MS, Nasif O, Alharbi SA, Batiha GES, Hossain MU. Prognostic and clinicopathological insights of phosphodiesterase 9A gene as novel biomarker in human colorectal cancer. BMC Cancer 2021; 21:577. [PMID: 34016083 PMCID: PMC8136133 DOI: 10.1186/s12885-021-08332-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND PDE9A (Phosphodiesterase 9A) plays an important role in proliferation of cells, their differentiation and apoptosis via intracellular cGMP (cyclic guanosine monophosphate) signaling. The expression pattern of PDE9A is associated with diverse tumors and carcinomas. Therefore, PDE9A could be a prospective candidate as a therapeutic target in different types of carcinoma. The study presented here was designed to carry out the prognostic value as a biomarker of PDE9A in Colorectal cancer (CRC). The present study integrated several cancer databases with in-silico techniques to evaluate the cancer prognosis of CRC. RESULTS The analyses suggested that the expression of PDE9A was significantly down-regulated in CRC tissues than in normal tissues. Moreover, methylation in the DNA promoter region might also manipulate PDE9A gene expression. The Kaplan-Meier curves indicated that high level of expression of PDE9A gene was associated to higher survival in OS, RFS, and DSS in CRC patients. PDE9A demonstrated the highest positive correlation for rectal cancer recurrence with a marker gene CEACAM7. Furtheremore, PDE9A shared consolidated pathways with MAPK14 to induce survival autophagy in CRC cells and showed interaction with GUCY1A2 to drive CRPC. CONCLUSIONS Overall, the prognostic value of PDE9A gene could be used as a potential tumor biomarker for CRC.
Collapse
Affiliation(s)
- Tasmina Ferdous Susmi
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Atikur Rahman
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Fermentation Engineering, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Md. Moshiur Rahman Khan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Farzana Yasmin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Md. Shariful Islam
- Department of Reproductive and Developmental Biology, Graduate School of Life Science, Hokkaido University, Sapporo, 5 Chome Kita 8 Jonishi, Kita Ward, Sapporo, Hokkaido 060-0808 Japan
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506-022 USA
| | - Omaima Nasif
- Department of Physiology, College of Medicine, King Saud University [Medical City], King Khalid University Hospital, PO Box 2925, Riyadh, 11461 Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany & Microbiology, College of Science, King Saud University, P.O Box 2455, Riyadh, 11451 Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511 Egypt
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349 Bangladesh
| |
Collapse
|
7
|
Su M, Gong X, Liu F. An update on the emerging approaches for histone deacetylase (HDAC) inhibitor drug discovery and future perspectives. Expert Opin Drug Discov 2021; 16:745-761. [PMID: 33530771 DOI: 10.1080/17460441.2021.1877656] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION HDACs catalyze the removal of acetyl groups from the ε-N-acetylated lysine residues of various protein substrates including both histone and nonhistone proteins. Different HDACs have distinct biological functions and are recruited to specific regions of the genome. HDAC inhibitors have attracted much attention in recent decades; indeed, there have been more than thirty HDAC inhibitors investigated in clinic trials with five approvals being achieved. AREAS COVERED This review covers the emerging approaches for HDAC inhibitor drug discovery from the past five years and includes discussion of structure-based rational design, isoform selectivity, and dual mechanism/multi-targeting. Chemical structures in addition to the in vitro and in vivo inhibiting activity of these compounds have also been discussed. EXPERT OPINION The exact role and biological functions of HDACs is still under investigation with a variety of HDAC inhibitors having been designed and evaluated. HDAC inhibitors have shown promise in treating cancer, AD, metabolic disease, viral infection, and multiple sclerosis, but there is still a lot of room for clinical improvement. In the future, more efforts should be put into (i) HDAC isoform identification (ii) the optimization of selectivity, activity, and pharmacokinetics; and (iii) unconventional approaches for discovering different effective scaffolds and pharmacophores.
Collapse
Affiliation(s)
- Ma Su
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, PR China
| | - Xingyu Gong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, PR China
| | - Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, PR China
| |
Collapse
|
8
|
Nadur NF, de Azevedo LL, Caruso L, Graebin CS, Lacerda RB, Kümmerle AE. The long and winding road of designing phosphodiesterase inhibitors for the treatment of heart failure. Eur J Med Chem 2020; 212:113123. [PMID: 33412421 DOI: 10.1016/j.ejmech.2020.113123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/14/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a superfamily of enzymes known to play a critical role in the indirect regulation of several intracellular metabolism pathways through the selective hydrolysis of the phosphodiester bonds of specific second messenger substrates such as cAMP (3',5'-cyclic adenosine monophosphate) and cGMP (3',5'-cyclic guanosine monophosphate), influencing the hypertrophy, contractility, apoptosis and fibroses in the cardiovascular system. The expression and/or activity of multiple PDEs is altered during heart failure (HF), which leads to changes in levels of cyclic nucleotides and function of cardiac muscle. Within the cardiovascular system, PDEs 1-5, 8 and 9 are expressed and are interesting targets for the HF treatment. In this comprehensive review we will present a briefly description of the biochemical importance of each cardiovascular related PDE to the HF, and cover almost all the "long and winding road" of designing and discovering ligands, hits, lead compounds, clinical candidates and drugs as PDE inhibitors in the last decade.
Collapse
Affiliation(s)
- Nathalia Fonseca Nadur
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Luciana Luiz de Azevedo
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Lucas Caruso
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Cedric Stephan Graebin
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Renata Barbosa Lacerda
- Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil
| | - Arthur Eugen Kümmerle
- Laboratório de Diversidade Molecular e Química Medicinal (LaDMol-QM, Molecular Diversity and Medicinal Chemistry Laboratory), Chemistry Institute, Rural Federal University of Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil; Programa de Pós-Gradução em Química (PPGQ), Universidade Federal Rural do Rio de Janeiro, Seropédica, Rio de Janeiro, 23897-000, Brazil.
| |
Collapse
|
9
|
Ballas SK. The Evolving Pharmacotherapeutic Landscape for the Treatment of Sickle Cell Disease. Mediterr J Hematol Infect Dis 2020; 12:e2020010. [PMID: 31934320 PMCID: PMC6951351 DOI: 10.4084/mjhid.2020.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 01/02/2023] Open
Abstract
Sickle cell disease (SCD) is an extremely heterogeneous disease that has been associated with global morbidity and early mortality. More effective and inexpensive therapies are needed. During the last five years, the landscape of the pharmacotherapy of SCD has changed dramatically. Currently, 54 drugs have been used or under consideration to use for the treatment of SCD. These fall into 3 categories: the first category includes the four drugs (Hydroxyurea, L-Glutamine, Crizanlizumab tmca and Voxelotor) that have been approved by the United States Food and Drug Administration (FDA) based on successful clinical trials. The second category includes 22 drugs that failed, discontinued or terminated for now and the third category includes 28 drugs that are actively being considered for the treatment of SCD. Crizanlizumab and Voxelotor are included in the first and third categories because they have been used in more than one trial. New therapies targeting multiple pathways in the complex pathophysiology of SCD have been achieved or are under continued investigation. The emerging trend seems to be the use of multimodal drugs (i.e. drugs that have different mechanisms of action) to treat SCD similar to the use of multiple chemotherapeutic agents to treat cancer.
Collapse
Affiliation(s)
- Samir K Ballas
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
10
|
Rabal O, Sánchez-Arias JA, Cuadrado-Tejedor M, de Miguel I, Pérez-González M, García-Barroso C, Ugarte A, Estella-Hermoso de Mendoza A, Sáez E, Espelosin M, Ursua S, Tan H, Wu W, Xu M, Pineda-Lucena A, Garcia-Osta A, Oyarzabal J. Multitarget Approach for the Treatment of Alzheimer's Disease: Inhibition of Phosphodiesterase 9 (PDE9) and Histone Deacetylases (HDACs) Covering Diverse Selectivity Profiles. ACS Chem Neurosci 2019; 10:4076-4101. [PMID: 31441641 DOI: 10.1021/acschemneuro.9b00303] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Here, we present a series of dual-target phosphodiesterase 9 (PDE9) and histone deacetylase (HDAC) inhibitors devised as pharmacological tool compounds for assessing the implications of these two targets in Alzheimer's disease (AD). These novel inhibitors were designed taking into account the key pharmacophoric features of known selective PDE9 inhibitors as well as privileged chemical structures, bearing zinc binding groups (hydroxamic acids and ortho-amino anilides) that hit HDAC targets. These substituents were selected according to rational criteria and previous knowledge from our group to explore diverse HDAC selectivity profiles (pan-HDAC, HDAC6 selective, and class I selective) that were confirmed in biochemical screens. Their functional response in inducing acetylation of histone and tubulin and phosphorylation of cAMP response element binding (CREB) was measured as a requisite for further progression into complete in vitro absorption, distribution, metabolism and excretion (ADME) and in vivo brain penetration profiling. Compound 31b, a selective HDAC6 inhibitor with acceptable brain permeability, was chosen for assessing in vivo efficacy of these first-in-class inhibitors, as well as studying their mode of action (MoA).
Collapse
Affiliation(s)
| | | | - Mar Cuadrado-Tejedor
- Pathology, Anatomy and Physiology Department, School of Medicine, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
- Health Research Institute of Navarra (IDISNA), E-31008 Pamplona, Spain
| | | | | | | | | | | | | | - Maria Espelosin
- Health Research Institute of Navarra (IDISNA), E-31008 Pamplona, Spain
| | - Susana Ursua
- Health Research Institute of Navarra (IDISNA), E-31008 Pamplona, Spain
| | - Haizhong Tan
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | - Wei Wu
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | - Musheng Xu
- WuXi Apptec (Tianjin) Co. Ltd., TEDA, No. 111 HuangHai Road, fourth Avenue, Tianjin 300456, PR China
| | | | - Ana Garcia-Osta
- Health Research Institute of Navarra (IDISNA), E-31008 Pamplona, Spain
| | | |
Collapse
|
11
|
Cuadrado-Tejedor M, Pérez-González M, García-Muñoz C, Muruzabal D, García-Barroso C, Rabal O, Segura V, Sánchez-Arias JA, Oyarzabal J, Garcia-Osta A. Taking Advantage of the Selectivity of Histone Deacetylases and Phosphodiesterase Inhibitors to Design Better Therapeutic Strategies to Treat Alzheimer's Disease. Front Aging Neurosci 2019; 11:149. [PMID: 31281249 PMCID: PMC6597953 DOI: 10.3389/fnagi.2019.00149] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/06/2019] [Indexed: 12/19/2022] Open
Abstract
The discouraging results with therapies for Alzheimer’s disease (AD) in clinical trials, highlights the urgent need to adopt new approaches. Like other complex diseases, it is becoming clear that AD therapies should focus on the simultaneous modulation of several targets implicated in the disease. Recently, using reference compounds and the first-in class CM-414, we demonstrated that the simultaneous inhibition of histone deacetylases [class I histone deacetylases (HDACs) and HDAC6] and phosphodiesterase 5 (PDE5) has a synergistic therapeutic effect in AD models. To identify the best inhibitory balance of HDAC isoforms and PDEs that provides a safe and efficient therapy to combat AD, we tested the compound CM-695 in the Tg2576 mouse model of this disease. CM-695 selectively inhibits HDAC6 over class I HDAC isoforms, which largely overcomes the toxicity associated with HDAC class 1 inhibition. Furthermore, CM-695 inhibits PDE9, which is expressed strongly in the brain and has been proposed as a therapeutic target for AD. Chronic treatment of aged Tg2576 mice with CM-695 ameliorates memory impairment and diminishes brain Aβ, although its therapeutic effect was no longer apparent 4 weeks after the treatment was interrupted. An increase in the presence of 78-KDa glucose regulated protein (GRP78) and heat shock protein 70 (Hsp70) chaperones may underlie the therapeutic effect of CM-695. In summary, chronic treatment with CM-695 appears to reverse the AD phenotype in a safe and effective manner. Taking into account that AD is a multifactorial disorder, the multimodal action of these compounds and the different events they affect may open new avenues to combat AD.
Collapse
Affiliation(s)
- Mar Cuadrado-Tejedor
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Marta Pérez-González
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Cristina García-Muñoz
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Damián Muruzabal
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Carolina García-Barroso
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Víctor Segura
- Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Juan A Sánchez-Arias
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Ana Garcia-Osta
- Neurobiology of Alzheimer's Disease, Neurosciences Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| |
Collapse
|
12
|
Taal K, Tuvikene J, Rullinkov G, Piirsoo M, Sepp M, Neuman T, Tamme R, Timmusk T. Neuralized family member NEURL1 is a ubiquitin ligase for the cGMP-specific phosphodiesterase 9A. Sci Rep 2019; 9:7104. [PMID: 31068605 PMCID: PMC6506465 DOI: 10.1038/s41598-019-43069-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/01/2019] [Indexed: 11/15/2022] Open
Abstract
Neuralized functions as a positive regulator of the Notch pathway by promoting ubiquitination of Notch ligands via its E3 ligase activity, resulting in their efficient endocytosis and signaling. Using a yeast two-hybrid screen, we have identified a cGMP-hydrolysing phosphodiesterase, PDE9A, as a novel interactor and substrate of Neuralized E3 ubiquitin protein ligase 1 (NEURL1). We confirmed this interaction with co-immunoprecipitation experiments and show that both Neuralized Homology Repeat domains of NEURL1 can interact with PDE9A. We also demonstrate that NEURL1 can promote polyubiquitination of PDE9A that leads to its proteasome-mediated degradation mainly via lysine residue K27 of ubiquitin. Our results suggest that NEURL1 acts as a novel regulator of protein levels of PDE9A.
Collapse
Affiliation(s)
- Kati Taal
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Grete Rullinkov
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Marko Piirsoo
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.,Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Mari Sepp
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.,Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, D-69120, Heidelberg, Germany
| | | | - Richard Tamme
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
| | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
| |
Collapse
|
13
|
Abstract
IMPACT STATEMENT Sickle cell disease (SCD) is one of the most common inherited diseases and is associated with a reduced life expectancy and acute and chronic complications, including frequent painful vaso-occlusive episodes that often require hospitalization. At present, treatment of SCD is limited to hematopoietic stem cell transplant, transfusion, and limited options for pharmacotherapy, based principally on hydroxyurea therapy. This review highlights the importance of intracellular cGMP-dependent signaling pathways in SCD pathophysiology; modulation of these pathways with soluble guanylate cyclase (sGC) stimulators or phosphodiesterase (PDE) inhibitors could potentially provide vasorelaxation and anti-inflammatory effects, as well as elevate levels of anti-sickling fetal hemoglobin.
Collapse
Affiliation(s)
- Nicola Conran
- Hematology Center, University of Campinas – UNICAMP,
Cidade Universitária, Campinas-SP 13083-878-SP, Brazil
| | - Lidiane Torres
- Hematology Center, University of Campinas – UNICAMP,
Cidade Universitária, Campinas-SP 13083-878-SP, Brazil
| |
Collapse
|
14
|
Singh N, Shreshtha AK, Thakur M, Patra S. Xanthine scaffold: scope and potential in drug development. Heliyon 2018; 4:e00829. [PMID: 30302410 PMCID: PMC6174542 DOI: 10.1016/j.heliyon.2018.e00829] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023] Open
Abstract
Medicinal plants have been the basis for discovery of various important marketed drugs. Xanthine is one such lead molecule. Xanthines in various forms (caffeine, theophylline, theobromine, etc) are abode in tea, coffee, cocoa, chocolate etc. giving them popular recognition. These compounds are best known for their diverse pharmaceutical applications as cyclic nucleotide phosphodiesterase inhibition, antagonization of adenosine receptor, anti-inflammatory, anti-microbial, anti-oxidant and anti-tumor activities. These properties incentivize to use xanthine as scaffold to develop new derivatives. Chemical synthesis contributes greater diversity in xanthine based derivatisation. With highlighting the existing challenges in chemical synthesis, the present review focuses the probable solution to fill existing lacuna. The review summarizes the available knowledge of xanthine based drugs development along with exploring new xanthine led chemical synthesis path for bringing diversification in xanthine based research. The main objective of this review is to explore the immense potential of xanthine as scaffold in drug development.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | | | - M.S. Thakur
- Fermentation Technology and Bioengineering Department, Central Food Technological Research Institute, Mysore, India
| | - Sanjukta Patra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
15
|
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
|
16
|
Patel NS, Klett J, Pilarzyk K, Lee DI, Kass D, Menniti FS, Kelly MP. Identification of new PDE9A isoforms and how their expression and subcellular compartmentalization in the brain change across the life span. Neurobiol Aging 2018; 65:217-234. [PMID: 29505961 DOI: 10.1016/j.neurobiolaging.2018.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 12/18/2017] [Accepted: 01/26/2018] [Indexed: 01/21/2023]
Abstract
3',5'-Cyclic nucleotide phosphodiesterases (PDEs) degrade 3',5' cyclic adenonosine monophosphate (cAMP) and 3',5' cyclic guanosine monophosphate (cGMP), with PDE9A having the highest affinity for cGMP. We show PDE9A6 and 3 novel PDE9 isoforms (PDE9X-100, PDE9X-120, and PDE9X-175) are reliably detected in the brain and lung of mice, whereas PDE9A2 and other isoforms are found elsewhere. PDE9A localizes to the membrane in all organs except the bladder, where it is cytosolic. Brain additionally shows PDE9 in the nuclear fraction. PDE9A mRNA expression/localization dramatically changes across neurodevelopment in a manner that is strikingly consistent between mice and humans (i.e., decreased expression in the hippocampus and cortex and inverted-U in the cerebellum). Study of the 4 PDE9 isoforms in the mouse brain from postnatal day 7 through 24 months similarly identifies dramatic effects of age on expression and subcellular compartmentalization that are isoform specific and brain region specific. Finally, PDE9A mRNA is elevated in the aged human hippocampus with dementia when there is a history of traumatic brain injury. Thus, brain PDE9 is localized to preferentially regulate nuclear- and membrane-proximal pools of cGMP, and its function likely changes across the life span.
Collapse
Affiliation(s)
- Neema S Patel
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Jennifer Klett
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Katy Pilarzyk
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Dong Ik Lee
- Division of Cardiology, Department of Medicine, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - David Kass
- Division of Cardiology, Department of Medicine, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Frank S Menniti
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
17
|
Wang JB, Lu HL, St. Leger RJ. The genetic basis for variation in resistance to infection in the Drosophila melanogaster genetic reference panel. PLoS Pathog 2017; 13:e1006260. [PMID: 28257468 PMCID: PMC5352145 DOI: 10.1371/journal.ppat.1006260] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/15/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Individuals vary extensively in the way they respond to disease but the genetic basis of this variation is not fully understood. We found substantial individual variation in resistance and tolerance to the fungal pathogen Metarhizium anisopliae Ma549 using the Drosophila melanogaster Genetic Reference Panel (DGRP). In addition, we found that host defense to Ma549 was correlated with defense to the bacterium Pseudomonas aeruginosa Pa14, and several previously published DGRP phenotypes including oxidative stress sensitivity, starvation stress resistance, hemolymph glucose levels, and sleep indices. We identified polymorphisms associated with differences between lines in both their mean survival times and microenvironmental plasticity, suggesting that lines differ in their ability to adapt to variable pathogen exposures. The majority of polymorphisms increasing resistance to Ma549 were sex biased, located in non-coding regions, had moderately large effect and were rare, suggesting that there is a general cost to defense. Nevertheless, host defense was not negatively correlated with overall longevity and fecundity. In contrast to Ma549, minor alleles were concentrated in the most Pa14-susceptible as well as the most Pa14-resistant lines. A pathway based analysis revealed a network of Pa14 and Ma549-resistance genes that are functionally connected through processes that encompass phagocytosis and engulfment, cell mobility, intermediary metabolism, protein phosphorylation, axon guidance, response to DNA damage, and drug metabolism. Functional testing with insertional mutagenesis lines indicates that 12/13 candidate genes tested influence susceptibility to Ma549. Many candidate genes have homologs identified in studies of human disease, suggesting that genes affecting variation in susceptibility are conserved across species.
Collapse
Affiliation(s)
- Jonathan B. Wang
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Hsiao-Ling Lu
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Raymond J. St. Leger
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Peng Y, Xia P, Zhang J, Villeneuve DL, Zhang J, Wang Z, Wei S, Yu H, Zhang X. Toxicogenomic Assessment of 6-OH-BDE47-Induced Developmental Toxicity in Chicken Embryos. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2016; 50:12493-12503. [PMID: 27749045 DOI: 10.1021/acs.est.6b04467] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hydroxylated polybrominated diphenyl ethers (OH-PBDEs) are analogs of PBDEs with hundreds of possible structures and are frequently detected in the environment. However, the in vivo evidence on the toxicity of OH-PBDEs is still very limited. Here, the developmental toxicity of 6-OH-BDE47, a predominant congener of OH-PBDEs detected in the environment, in chicken embryos was assessed using a toxicogenomic approach. Fertilized chicken eggs were dosed via in ovo administration of 0.006 to 0.474 nmol 6-OH-BDE47/g egg followed by 18 days of incubation. Significant embryo lethality (LD50 = 1.940 nmol/g egg) and increased hepatic somatic index (HSI) were caused by 6-OH-BDE47 exposure. The functional enrichment of differentially expressed genes (DEGs) was associated with oxidative phosphorylation, generation of precursor metabolites and energy, and electron transport chains, which suggest that 6-OH-BDE47 exposure may disrupt the embryo development by altering the function of energy production in mitochondria. Moreover, aryl hydrocarbon receptor (AhR)-mediated responses including up-regulation of CYP1A4 were observed in the livers of embryos exposed to 6-OH-BDE47. Overall, this study confirmed the embryo lethality by 6-OH-BDE47 and further improved the mechanistic understanding of OH-PBDEs-caused toxicity. Ecological risk assessment via application of both no-observed-effect level (NOEL) and the sensitive NOTEL (transcriptional NOEL) suggested that OH-PBDEs might cause ecological risk to wild birds.
Collapse
Affiliation(s)
- Ying Peng
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Pu Xia
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Junjiang Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Daniel L Villeneuve
- Mid-Continent Ecology Division, United States Environmental Protection Agency , Duluth, Minnesota 55804, United States
| | - Jiamin Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Zhihao Wang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Si Wei
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Hongxia Yu
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| | - Xiaowei Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University , Nanjing 210023, PR China
| |
Collapse
|
19
|
Huang M, Shao Y, Hou J, Cui W, Liang B, Huang Y, Li Z, Wu Y, Zhu X, Liu P, Wan Y, Ke H, Luo HB. Structural Asymmetry of Phosphodiesterase-9A and a Unique Pocket for Selective Binding of a Potent Enantiomeric Inhibitor. Mol Pharmacol 2015; 88:836-45. [PMID: 26316540 DOI: 10.1124/mol.115.099747] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/24/2015] [Indexed: 01/21/2023] Open
Abstract
Phosphodiesterase-9 (PDE9) inhibitors have been studied as potential therapeutics for treatment of central nervous system diseases and diabetes. Here, we report the discovery of a new category of PDE9 inhibitors by rational design on the basis of the crystal structures. The best compound, (S)-6-((1-(4-chlorophenyl)ethyl)amino)-1-cyclopentyl-1,5,6,7-tetrahydro-4H-pyrazolo[3,4-day]pyrimidin-4-one [(S)-C33], has an IC50 value of 11 nM against PDE9 and the racemic C33 has bioavailability of 56.5% in the rat pharmacokinetic model. The crystal structures of PDE9 in the complex with racemic C33, (R)-C33, and (S)-C33 reveal subtle conformational asymmetry of two M-loops in the PDE9 dimer and different conformations of two C33 enantiomers. The structures also identified a small hydrophobic pocket that interacts with the tyrosyl tail of (S)-C33 but not with (R)-C33, and is thus possibly useful for improvement of selectivity of PDE9 inhibitors. The asymmetry of the M-loop and the different interactions of the C33 enantiomers imply the necessity to consider the whole PDE9 dimer in the design of inhibitors.
Collapse
Affiliation(s)
- Manna Huang
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Yongxian Shao
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Jianying Hou
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Wenjun Cui
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Beibei Liang
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Yingchun Huang
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Zhe Li
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Yinuo Wu
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Xinhai Zhu
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Peiqing Liu
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Yiqian Wan
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Hengming Ke
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| | - Hai-Bin Luo
- School of Chemistry and Chemical Engineering (M.H., J.H., X.Z. Yiq.W.), School of Pharmaceutical Sciences (Y.S., Z.L., Yin.W., P.L., H.-B.L.), Sun Yat-Sen University, Guangzhou, PR China; and Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina (W.C., B.L., Y.H., H.K.)
| |
Collapse
|
20
|
Cheng ZB, Lu X, Bao JM, Han QH, Dong Z, Tang GH, Gan LS, Luo HB, Yin S. (±)-Torreyunlignans A-D, rare 8-9' linked neolignan enantiomers as phosphodiesterase-9A inhibitors from Torreya yunnanensis. JOURNAL OF NATURAL PRODUCTS 2014; 77:2651-2657. [PMID: 25495612 DOI: 10.1021/np500528u] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
(±)-Torreyunlignans A-D (1a/1b-4a/4b), four pairs of new 8-9' linked neolignan enantiomers featuring a rare (E)-2-styryl-1,3-dioxane moiety, were isolated from the trunk of Torreya yunnanensis. The structures were determined by combined spectroscopic and chemical methods, and the absolute configurations were elucidated by ECD calculations. The compounds were screened by using tritium-labeled adenosine 3',5'-cyclic monophosphate ([(3)H]-cGMP) as a substrate for inhibitory affinities against phosphodiesterase-9A (PDE9A), which is a potential target for the treatment of diabetes and Alzheimer's disease. All of the enantiomers exhibited inhibition against PDE9A with IC50 values ranging from 5.6 to 15.0 μM. This is the first report of PDE9A inhibitors from nature.
Collapse
Affiliation(s)
- Zhong-Bin Cheng
- School of Pharmaceutical Sciences, Sun Yat-sen University , Guangzhou, Guangdong 510006, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|