1
|
Kochoian BA, Bure C, Papa SM. Targeting Striatal Glutamate and Phosphodiesterases to Control L-DOPA-Induced Dyskinesia. Cells 2023; 12:2754. [PMID: 38067182 PMCID: PMC10706484 DOI: 10.3390/cells12232754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
A large body of work during the past several decades has been focused on therapeutic strategies to control L-DOPA-induced dyskinesias (LIDs), common motor complications of long-term L-DOPA therapy in Parkinson's disease (PD). Yet, LIDs remain a clinical challenge for the management of patients with advanced disease. Glutamatergic dysregulation of striatal projection neurons (SPNs) appears to be a key contributor to altered motor responses to L-DOPA. Targeting striatal hyperactivity at the glutamatergic neurotransmission level led to significant preclinical and clinical trials of a variety of antiglutamatergic agents. In fact, the only FDA-approved treatment for LIDs is amantadine, a drug with NMDAR antagonistic actions. Still, novel agents with improved pharmacological profiles are needed for LID therapy. Recently other therapeutic targets to reduce dysregulated SPN activity at the signal transduction level have emerged. In particular, mechanisms regulating the levels of cyclic nucleotides play a major role in the transduction of dopamine signals in SPNs. The phosphodiesterases (PDEs), a large family of enzymes that degrade cyclic nucleotides in a specific manner, are of special interest. We will review the research for antiglutamatergic and PDE inhibition strategies in view of the future development of novel LID therapies.
Collapse
Affiliation(s)
- Brik A. Kochoian
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Cassandra Bure
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Stella M. Papa
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30329, USA
| |
Collapse
|
2
|
Landucci E, Ribaudo G, Anyanwu M, Oselladore E, Giannangeli M, Mazzantini C, Lana D, Giovannini MG, Memo M, Pellegrini-Giampietro DE, Gianoncelli A. Virtual Screening-Accelerated Discovery of a Phosphodiesterase 9 Inhibitor with Neuroprotective Effects in the Kainate Toxicity In Vitro Model. ACS Chem Neurosci 2023; 14:3826-3838. [PMID: 37726213 PMCID: PMC10587872 DOI: 10.1021/acschemneuro.3c00431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023] Open
Abstract
In the central nervous system, some specific phosphodiesterase (PDE) isoforms modulate pathways involved in neuronal plasticity. Accumulating evidence suggests that PDE9 may be a promising therapeutic target for neurodegenerative diseases. In the current study, computational techniques were used to identify a nature-inspired PDE9 inhibitor bearing the scaffold of an isoflavone, starting from a database of synthetic small molecules using a ligand-based approach. Furthermore, docking studies supported by molecular dynamics investigations allowed us to evaluate the features of the ligand-target complex. In vitro assays confirmed the computational results, showing that the selected compound inhibits the enzyme in the nanomolar range. Additionally, we evaluated the expression of gene and protein levels of PDE9 in organotypic hippocampal slices, observing an increase following exposure to kainate (KA). Importantly, the PDE9 inhibitor reduced CA3 damage induced by KA in a dose-dependent manner in organotypic hippocampal slices. Taken together, these observations strongly support the potential of the identified nature-inspired PDE9 inhibitor and suggest that such a molecule could represent a promising lead compound to develop novel therapeutic tools against neurological diseases..
Collapse
Affiliation(s)
- Elisa Landucci
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Giovanni Ribaudo
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Margrate Anyanwu
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Erika Oselladore
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Matteo Giannangeli
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Costanza Mazzantini
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Daniele Lana
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Maria Grazia Giovannini
- Department
of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze 50139, Italy
| | - Maurizio Memo
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | | | - Alessandra Gianoncelli
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| |
Collapse
|
3
|
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
|
4
|
Hou X, Rong C, Zhang Q, Song S, Cong Y, Zhang HT. Cyclic Nucleotide Phosphodiesterases in Alcohol Use Disorders: Involving Gut Microbiota. Int J Neuropsychopharmacol 2022; 26:70-79. [PMID: 36087271 PMCID: PMC9850663 DOI: 10.1093/ijnp/pyac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 01/22/2023] Open
Abstract
Alcohol abuse is 1 of the most significant public health problems in the world. Chronic, excessive alcohol consumption not only causes alcohol use disorder (AUD) but also changes the gut and lung microbiota, including bacterial and nonbacterial types. Both types of microbiota can release toxins, further damaging the gastrointestinal and respiratory tracts; causing inflammation; and impairing the functions of the liver, lung, and brain, which in turn deteriorate AUD. Phosphodiesterases (PDEs) are critical in the control of intracellular cyclic nucleotides, including cyclic adenosine monophosphate and cyclic guanosine monophosphate. Inhibition of certain host PDEs reduces alcohol consumption and attenuates alcohol-related impairment. These PDEs are also expressed in the microbiota and may play a role in controlling microbiota-associated inflammation. Here, we summarize the influences of alcohol on gut/lung bacterial and nonbacterial microbiota as well as on the gut-liver/brain/lung axis. We then discuss the relationship between gut and lung microbiota-mediated PDE signaling and AUD consequences in addition to highlighting PDEs as potential targets for treatment of AUD.
Collapse
Affiliation(s)
- Xueqin Hou
- Correspondence: Xueqin Hou, PhD, Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China ()
| | | | - Qiwei Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Shuangshuang Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Yifan Cong
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Han-Ting Zhang
- Han-Ting Zhang, MD, PhD, Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266073, P.R. China ()
| |
Collapse
|
5
|
Yan Y, Gao S, Avasthi S, Zhao Y, Ye J, Tao Y, Wang W, Zhu X, Du F, O'Donnell JM, Xu Y. Protective effects of phosphodiesterase 2 inhibitor against Aβ 1-42 induced neuronal toxicity. Neuropharmacology 2022; 213:109128. [PMID: 35588859 DOI: 10.1016/j.neuropharm.2022.109128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 01/12/2023]
Abstract
Our previous study suggested that inhibition of Phosphodiesterase 2 ameliorates memory loss upon exposure to oxidative stress. While whether memory enhancing effects of PDE2 inhibition on Alzheimer's disease mouse model are involved in antioxidant defense and neuronal remodeling, are largely unexplored. The present study addressed whether and how PDE2 inhibitor Bay 60-7550 rescued Aβ oligomers (Aβo)-induced neuronal damage and memory impairment. The results suggested that exposure of primary cortical neurons to Aβo induced neuronal cells damage and increased PDE2 expression, which were paralleled to an increase in the oxidative parameter malondialdehyde (MDA) level and cellular apoptosis. However, this Aβo-induced oxidative damage was blocked by pre-treatment with protein kinase A or G (PKA or PKG) inhibitor, suggesting the involvement of cAMP/cGMP signaling. Moreover, microinjection of Aβo into the prefrontal cortex of mice increased the MDA level; while Bay 60-7550 reversed this effect and increased antioxidant and anti-apoptotic factors, i.e. increased trolox-equivalent-antioxidant capacity and Bcl-2/Bax ratio. Bay 60-7550 also rescued Aβo-induced synaptic atrophy and memory deficits, as evidenced by the increased synaptic proteins' levels and spine density in the prefrontal cortex, and improved cognitive behaviors by decreased working memory errors in the eight-arm maze and increased discrimination index in the novel object recognition test. These findings suggest that inhibition of PDE2 contributes to antioxidant defense and neuronal remodeling by regulation of cAMP/cGMP signaling, which provide a theoretical basis for the future use of PDE2 inhibitors as the anti-AD drugs.
Collapse
Affiliation(s)
- Yuqing Yan
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shichao Gao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Shivani Avasthi
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Yuhan Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Jianghong Ye
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Yuanxiang Tao
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA
| | - Wei Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Fu Du
- FD Neurotechnologies, Inc., Elicott City, MD, 21041, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Ying Xu
- Department of Anesthesiology, Rutgers New Jersey Medical School, The State University of New Jersey, Newark, NJ, 07101, USA.
| |
Collapse
|
6
|
Jehle A, Garaschuk O. The Interplay between cGMP and Calcium Signaling in Alzheimer's Disease. Int J Mol Sci 2022; 23:7048. [PMID: 35806059 PMCID: PMC9266933 DOI: 10.3390/ijms23137048] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger and a key molecule in many important signaling cascades in the body and brain, including phototransduction, olfaction, vasodilation, and functional hyperemia. Additionally, cGMP is involved in long-term potentiation (LTP), a cellular correlate of learning and memory, and recent studies have identified the cGMP-increasing drug Sildenafil as a potential risk modifier in Alzheimer's disease (AD). AD development is accompanied by a net increase in the expression of nitric oxide (NO) synthases but a decreased activity of soluble guanylate cyclases, so the exact sign and extent of AD-mediated imbalance remain unclear. Moreover, human patients and mouse models of the disease present with entangled deregulation of both cGMP and Ca2+ signaling, e.g., causing changes in cGMP-mediated Ca2+ release from the intracellular stores as well as Ca2+-mediated cGMP production. Still, the mechanisms governing such interplay are poorly understood. Here, we review the recent data on mechanisms underlying the brain cGMP signaling and its interconnection with Ca2+ signaling. We also discuss the recent evidence stressing the importance of such interplay for normal brain function as well as in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
7
|
Masilamoni GJ, Sinon CG, Kochoian BA, Singh A, Mcriner AJ, Leventhal L, Papa SM. Phosphodiesterase 9 inhibition prolongs the antiparkinsonian action of l-DOPA in parkinsonian non-human primates. Neuropharmacology 2022; 212:109060. [DOI: 10.1016/j.neuropharm.2022.109060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/05/2022] [Accepted: 04/09/2022] [Indexed: 10/18/2022]
|
8
|
Xi M, Sun T, Chai S, Xie M, Chen S, Deng L, Du K, Shen R, Sun H. Therapeutic potential of phosphodiesterase inhibitors for cognitive amelioration in Alzheimer's disease. Eur J Med Chem 2022; 232:114170. [DOI: 10.1016/j.ejmech.2022.114170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 02/07/2023]
|
9
|
Gorny N, Kelly MP. Alterations in cyclic nucleotide signaling are implicated in healthy aging and age-related pathologies of the brain. VITAMINS AND HORMONES 2021; 115:265-316. [PMID: 33706951 DOI: 10.1016/bs.vh.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is not only important to consider how hormones may change with age, but also how downstream signaling pathways that couple to hormone receptors may change. Among these hormone-coupled signaling pathways are the 3',5'-cyclic guanosine monophosphate (cGMP) and 3',5'-cyclic adenosine monophosphate (cAMP) intracellular second messenger cascades. Here, we test the hypothesis that dysfunction of cAMP and/or cGMP synthesis, execution, and/or degradation occurs in the brain during healthy and pathological diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Although most studies report lower cyclic nucleotide signaling in the aged brain, with further reductions noted in the context of age-related diseases, there are select examples where cAMP signaling may be elevated in select tissues. Thus, therapeutics would need to target cAMP/cGMP in a tissue-specific manner if efficacy for select symptoms is to be achieved without worsening others.
Collapse
Affiliation(s)
- Nicole Gorny
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michy P Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
10
|
Jiang H, Zheng Y, Ni J, Xu Y. BAY 73-6691 Alters Neuron Plasticity and Phosphorylation of Tau Through Regulation of Cyclic Guanosine Monophosphate/Protein Kinase G/Cyclic Adenosine Monophosphate Response Element-Binding Protein Pathway. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alzheimer’s disease (AD) is one of neurodegenerative diseases characterized by cognitive and memory decline, accompanying with neurofibrillary tangles (NFTs) made of hyperphosphorylated tau protein and senile plaques (SP) accumulated by β-amyloid protein (Aβ).
BAY 73-6691, an inhibitor of phosphodiesterase-9 (PDE-9), can improve learning and memory of elderly rats. However, the effects of BAY 73-6691 on neuroapoptotic and neuroinflammatory events, as well as synaptic plasticity of differentiated PC12 cells are remain unclear. In this work, we screened
apoptotic cells induced by Aβ25-35 via flow cytometry. TNF-α, IL-1β, IL-6 secreted by PC12 cells were estimated by ELISA kits. The levels of cGMP, PKG and CREB mediated by BAY 73-6691 were assessed. Moreover, we conducted western blots analysis
to evaluate the phosphorylation of tau and synaptic related proteins. Results showed that BAY 73-6691 could reduce Aβ25-35-triggered neuroapoptosis and neuroinflammation. Phosphorylation of tau was inhibited by BAY 73-6691, whereas sildenafil citrate (SC, an inhibitor
of cGMP) partially weakened the effect of BAY 73-6691. Additionally, synaptic plasticity restored by BAY 73-6691 was also suppressed via SC. Taken together, BAY 73-6691 exhibited neuro protective effects, and altered tau phosphorylation as well as synaptic related proteins through cGMP/PKG/CREB
pathway.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P. R. China
| | - Yan Zheng
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P. R. China
| | - Jie Ni
- Department of Emergency, Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210000, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, 210000, China
| |
Collapse
|
11
|
Ribaudo G, Memo M, Gianoncelli A. A Perspective on Natural and Nature-Inspired Small Molecules Targeting Phosphodiesterase 9 (PDE9): Chances and Challenges against Neurodegeneration. Pharmaceuticals (Basel) 2021; 14:ph14010058. [PMID: 33451065 PMCID: PMC7828511 DOI: 10.3390/ph14010058] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/24/2022] Open
Abstract
As life expectancy increases, dementia affects a growing number of people worldwide. Besides current treatments, phosphodiesterase 9 (PDE9) represents an alternative target for developing innovative small molecules to contrast neurodegeneration. PDE inhibition promotes neurotransmitter release, amelioration of microvascular dysfunction, and neuronal plasticity. This review will provide an update on natural and nature-inspired PDE9 inhibitors, with a focus on the structural features of PDE9 that encourage the development of isoform-selective ligands. The expression in the brain, the presence within its structure of a peculiar accessory pocket, the asymmetry between the two subunits composing the protein dimer, and the selectivity towards chiral species make PDE9 a suitable target to develop specific inhibitors. Additionally, the world of natural compounds is an ideal source for identifying novel, possibly asymmetric, scaffolds, and xanthines, flavonoids, neolignans, and their derivatives are currently being studied. In this review, the available literature data were interpreted and clarified, from a structural point of view, taking advantage of molecular modeling: 3D structures of ligand-target complexes were retrieved, or built, and discussed.
Collapse
|
12
|
Memory Enhancers for Alzheimer's Dementia: Focus on cGMP. Pharmaceuticals (Basel) 2021; 14:ph14010061. [PMID: 33451088 PMCID: PMC7828493 DOI: 10.3390/ph14010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Cyclic guanosine-3',5'-monophosphate, better known as cyclic-GMP or cGMP, is a classical second messenger involved in a variety of intracellular pathways ultimately controlling different physiological functions. The family of guanylyl cyclases that includes soluble and particulate enzymes, each of which comprises several isoforms with different mechanisms of activation, synthesizes cGMP. cGMP signaling is mainly executed by the activation of protein kinase G and cyclic nucleotide gated channels, whereas it is terminated by its hydrolysis to GMP operated by both specific and dual-substrate phosphodiesterases. In the central nervous system, cGMP has attracted the attention of neuroscientists especially for its key role in the synaptic plasticity phenomenon of long-term potentiation that is instrumental to memory formation and consolidation, thus setting off a "gold rush" for new drugs that could be effective for the treatment of cognitive deficits. In this article, we summarize the state of the art on the neurochemistry of the cGMP system and then review the pre-clinical and clinical evidence on the use of cGMP enhancers in Alzheimer's disease (AD) therapy. Although preclinical data demonstrates the beneficial effects of cGMP on cognitive deficits in AD animal models, the results of the clinical studies carried out to date are not conclusive. More trials with a dose-finding design on selected AD patient's cohorts, possibly investigating also combination therapies, are still needed to evaluate the clinical potential of cGMP enhancers.
Collapse
|
13
|
Sharma VK, Singh TG, Singh S. Cyclic Nucleotides Signaling and Phosphodiesterase Inhibition: Defying Alzheimer's Disease. Curr Drug Targets 2020; 21:1371-1384. [PMID: 32718286 DOI: 10.2174/1389450121666200727104728] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Defects in brain functions associated with aging and neurodegenerative diseases benefit insignificantly from existing options, suggesting that there is a lack of understanding of pathological mechanisms. Alzheimer's disease (AD) is such a nearly untreatable, allied to age neurological deterioration for which only the symptomatic cure is available and the agents able to mould progression of the disease, is still far away. The altered expression of phosphodiesterases (PDE) and deregulated cyclic nucleotide signaling in AD has provoked a new thought of targeting cyclic nucleotide signaling in AD. Targeting cyclic nucleotides as an intracellular messenger seems to be a viable approach for certain biological processes in the brain and controlling substantial. Whereas, the synthesis, execution, and/or degradation of cyclic nucleotides has been closely linked to cognitive deficits. In relation to cognition, the cyclic nucleotides (cAMP and cGMP) have an imperative execution in different phases of memory, including gene transcription, neurogenesis, neuronal circuitry, synaptic plasticity and neuronal survival, etc. AD is witnessed by impairments of these basic processes underlying cognition, suggesting a crucial role of cAMP/cGMP signaling in AD populations. Phosphodiesterase inhibitors are the exclusive set of enzymes to facilitate hydrolysis and degradation of cAMP and cGMP thereby, maintains their optimum levels initiating it as an interesting target to explore. The present work reviews a neuroprotective and substantial influence of PDE inhibition on physiological status, pathological progression and neurobiological markers of AD in consonance with the intensities of cAMP and cGMP.
Collapse
Affiliation(s)
- Vivek K Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India,Govt. College of Pharmacy, Rohru, District Shimla, Himachal Pradesh-171207, India
| | - Thakur G Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
14
|
Jankowska A, Wesołowska A, Pawłowski M, Chłoń-Rzepa G. Multifunctional Ligands Targeting Phosphodiesterase as the Future Strategy for the Symptomatic and Disease-Modifying Treatment of Alzheimer’s Disease. Curr Med Chem 2020; 27:5351-5373. [DOI: 10.2174/0929867326666190620095623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer’s Disease (AD) is a chronic neurodegenerative disorder characterized by cognitive
impairments such as memory loss, decline in language skills, and disorientation that affects
over 46 million people worldwide. Patients with AD also suffer from behavioral and psychological
symptoms of dementia that deteriorate their quality of life and lead to premature death. Currently
available drugs provide modest symptomatic relief but do not reduce pathological hallmarks (senile
plaques and neurofibrillary tangles) and neuroinflammation, both of which are integral parts of dementia.
A large body of evidence indicates that impaired signaling pathways of cyclic-3′,5′-
Adenosine Monophosphate (cAMP) and cyclic-3′,5′-guanosine Monophosphate (cGMP) may contribute
to the development and progression of AD. In addition, Phosphodiesterase (PDE) inhibitors,
commonly known as cAMP and/or cGMP modulators, were found to be involved in the phosphorylation
of tau; aggregation of amyloid beta; neuroinflammation; and regulation of cognition, mood,
and emotion processing. The purpose of this review was to update the most recent reports on the
development of novel multifunctional ligands targeting PDE as potential drugs for both symptomatic
and disease-modifying therapy of AD. This review collected the chemical structures of representative
multifunctional ligands, results of experimental in vitro and in vivo pharmacological studies,
and current opinions regarding the potential utility of these compounds for the comprehensive
therapy of AD. Finally, the multiparameter predictions of drugability of the representative compounds
were calculated and discussed.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
15
|
Zhang P, Jiang MY, Le ML, Zhang B, Zhou Q, Wu Y, Zhang C, Luo HB. Design, synthesis and evaluation of pyrazolopyrimidinone derivatives as novel PDE9A inhibitors for treatment of Alzheimer’s disease. Bioorg Med Chem Lett 2020; 30:127254. [DOI: 10.1016/j.bmcl.2020.127254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 01/24/2023]
|
16
|
Ribaudo G, Ongaro A, Zagotto G, Memo M, Gianoncelli A. Therapeutic Potential of Phosphodiesterase Inhibitors against Neurodegeneration: The Perspective of the Medicinal Chemist. ACS Chem Neurosci 2020; 11:1726-1739. [PMID: 32401481 PMCID: PMC8007108 DOI: 10.1021/acschemneuro.0c00244] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
Increasing human
life expectancy prompts the development of novel
remedies for cognitive decline: 44 million people worldwide are affected
by dementia, and this number is predicted to triple by 2050. Acetylcholinesterase
and N-methyl-d-aspartate receptors represent
the targets of currently available drugs for Alzheimer’s disease,
which are characterized by limited efficacy. Thus, the search for
therapeutic agents with alternative or combined mechanisms of action
is wide open. Since variations in 3′,5′-cyclic adenosine
monophosphate, 3′,5′-cyclic guanosine monophosphate,
and/or nitric oxide levels interfere with downstream pathways involved
in memory processes, evidence supporting the potential of phosphodiesterase
(PDE) inhibitors in contrasting neurodegeneration should be
critically considered. For the preparation of this Review, more than
140 scientific papers were retrieved by searching PubMed and Scopus
databases. A systematic approach was adopted when overviewing the
different PDE isoforms, taking into account details on brain localization,
downstream molecular mechanisms, and inhibitors currently under study,
according to available in vitro and in vivo data. In the context of drug repurposing, a section focusing on
PDE5 was introduced. Original computational studies were performed
to rationalize the emerging evidence that suggests the role of PDE5
inhibitors as multi-target agents against neurodegeneration.
Moreover, since such compounds must cross the blood–brain barrier
and reach inhibitory concentrations in the central nervous system
to exert their therapeutic activity, physicochemical parameters
were analyzed and discussed. Taken together, literature and computational
data suggest that some PDE5 inhibitors, such as tadalafil, represent
promising candidates.
Collapse
Affiliation(s)
- Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Alberto Ongaro
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Giuseppe Zagotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Alessandra Gianoncelli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
17
|
Abstract
Nitric oxide/cyclic guanosine monophosphate (cGMP) signaling is compromised in Alzheimer’s disease (AD), and phosphodiesterase 5 (PDE5), which degrades cGMP, is upregulated. Sildenafil inhibits PDE5 and increases cGMP levels. Integrating previous findings, we determine that most doses of sildenafil (especially low doses) likely activate peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) via protein kinase G-mediated cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) phosphorylation and/or Sirtuin-1 activation and PGC1α deacetylation. Via PGC1α signaling, low-dose sildenafil likely suppresses β-secretase 1 expression and amyloid-β (Aβ) generation, upregulates antioxidant enzymes, and induces mitochondrial biogenesis. Plus, sildenafil should increase brain perfusion, insulin sensitivity, long-term potentiation, and neurogenesis while suppressing neural apoptosis and inflammation. A systematic review of sildenafil in AD was undertaken. In vitro, sildenafil protected neural mitochondria from Aβ and advanced glycation end products. In transgenic AD mice, sildenafil was found to rescue deficits in CREB phosphorylation and memory, upregulate brain-derived neurotrophic factor, reduce reactive astrocytes and microglia, decrease interleukin-1β, interleukin-6, and tumor necrosis factor-α, decrease neural apoptosis, increase neurogenesis, and reduce tau hyperphosphorylation. All studies that tested Aβ levels reported significant improvements except the two that used the highest dosage, consistent with the dose-limiting effect of cGMP-induced phosphodiesterase 2 (PDE2) activation and cAMP depletion on PGC1α signaling. In AD patients, a single dose of sildenafil decreased spontaneous neural activity, increased cerebral blood flow, and increased the cerebral metabolic rate of oxygen. A randomized control trial of sildenafil (ideally with a PDE2 inhibitor) in AD patients is warranted.
Collapse
|
18
|
PDE9 inhibition promotes proliferation of neural stem cells via cGMP-PKG pathway following oxygen-glucose deprivation/reoxygenation injury in vitro. Neurochem Int 2019; 133:104630. [PMID: 31821840 DOI: 10.1016/j.neuint.2019.104630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023]
Abstract
Cerebral ischemia is one of leading causes of death and long-term disability worldwide. Stem cell-based therapy is promising some valuable strategies for the structural and functional recovery after ischemic insult. The inhibition of phosphodiesterases (PDEs) has wide spectrum neuroprotective properties by stimulating proliferation of neural stem cells (NSCs). However, the potential role of PDE9 on NSCs proliferation after cerebral ischemia is not well investigated. The present study aimed to assess the contribution of PDE9 inhibition on the proliferation of NSCs and to determine the details of its underlying mechanisms against cerebral ischemia. The survival and proliferation of NSCs were assessed by CCK-8 assay and BrdU immunofluorescence staining, respectively. PDE9 activity and cGMP level were measured by ELISA kits. The protein expression of PKG and BDNF was detected by Western blot. Exposing NSCs of cultured primary hippocampus to oxygen-glucose deprivation/reoxygenation (OGD/R) significantly decreased the survival rate, but increased the proliferation of NSCs. Meanwhile, PDE9 activity was decreased, cGMP level was increased, PKG and BDNF protein expression was increased. PF-04447953, a PDE9 inhibitor, increased the survival rate of NSCs, moreover, PDE9 activity reduced more, and NSCs proliferation, cGMP level, PKG and BDNF protein expression were increased further, compared with OGD/R model group. These effects of PF-04447953, except for PDE9 activity and cGMP level, were reversed by treatment with KT5823, a PKG inhibitor. Taken together, the inhibition of PDE9 can promote the proliferation of NSCs following OGD/R injury, which may be, at least partly, mediated by cGMP-PKG pathway.
Collapse
|
19
|
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
|
20
|
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
|
21
|
Sharma P, Srivastava P, Seth A, Tripathi PN, Banerjee AG, Shrivastava SK. Comprehensive review of mechanisms of pathogenesis involved in Alzheimer's disease and potential therapeutic strategies. Prog Neurobiol 2018; 174:53-89. [PMID: 30599179 DOI: 10.1016/j.pneurobio.2018.12.006] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/04/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
Abstract
AD is a progressive neurodegenerative disorder and a leading cause of dementia in an aging population worldwide. The enormous challenge which AD possesses to global healthcare makes it as urgent as ever for the researchers to develop innovative treatment strategies to fight this disease. An in-depth analysis of the extensive available data associated with the AD is needed for a more comprehensive understanding of underlying molecular mechanisms and pathophysiological pathways associated with the onset and progression of the AD. The currently understood pathological and biochemical manifestations include cholinergic, Aβ, tau, excitotoxicity, oxidative stress, ApoE, CREB signaling pathways, insulin resistance, etc. However, these hypotheses have been criticized with several conflicting reports for their involvement in the disease progression. Several issues need to be addressed such as benefits to cost ratio with cholinesterase therapy, the dilemma of AChE selectivity over BChE, BBB permeability of peptidic BACE-1 inhibitors, hurdles related to the implementation of vaccination and immunization therapy, and clinical failure of candidates related to newly available targets. The present review provides an insight to the different molecular mechanisms involved in the development and progression of the AD and potential therapeutic strategies, enlightening perceptions into structural information of conventional and novel targets along with the successful applications of computational approaches for the design of target-specific inhibitors.
Collapse
Affiliation(s)
- Piyoosh Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pavan Srivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ankit Seth
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Prabhash Nath Tripathi
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Anupam G Banerjee
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushant K Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
| |
Collapse
|
22
|
Lai B, Li M, Hu WL, Li W, Gan WB. The Phosphodiesterase 9 Inhibitor PF-04449613 Promotes Dendritic Spine Formation and Performance Improvement after Motor Learning. Dev Neurobiol 2018; 78:859-872. [PMID: 30022611 PMCID: PMC6158093 DOI: 10.1002/dneu.22623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022]
Abstract
The cyclic nucleotide cGMP is an intracellular second messenger with important roles in neuronal functions and animals' behaviors. The phosphodiesterases (PDEs) are a family of enzymes that hydrolyze the second messengers cGMP and cAMP. Inhibition of phosphodiesterase 9 (PDE9), a main isoform of PDEs hydrolyzing cGMP, has been shown to improve learning and memory as well as cognitive function in rodents. However, the role of PDE9 in regulating neuronal structure and function in vivo remains unclear. Here we used in vivo two-photon microscopy to investigate the effect of a selective PDE9 inhibitor PF-04449613 on the activity and plasticity of dendritic spines of layer V pyramidal neurons in the mouse primary motor cortex. We found that administration of PF-04449613 increased calcium activity of dendrites and dendritic spines of layer V pyramidal neurons in mice under resting and running conditions. Chronic treatment of PF-04449613 over weeks increased dendritic spine formation and elimination under basal conditions. Furthermore, PF-04449613 treatment over 1-7 days increased the formation and survival of new spines as well as performance improvement after rotarod motor training. Taken together, our studies suggest that elevating the level of cGMP with the PDE9 inhibitor PF-04449613 increases synaptic calcium activity and learning-dependent synaptic plasticity, thereby contributing to performance improvement after learning. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 00: 000-000, 2018.
Collapse
Affiliation(s)
- Baoling Lai
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
- Molecular Neurobiology Program, Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Miao Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
| | - Wan-Ling Hu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
| | - Wei Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
| | - Wen-Biao Gan
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
- Molecular Neurobiology Program, Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
23
|
Real-Time Imaging Reveals Augmentation of Glutamate-Induced Ca 2+ Transients by the NO-cGMP Pathway in Cerebellar Granule Neurons. Int J Mol Sci 2018; 19:ijms19082185. [PMID: 30049956 PMCID: PMC6121606 DOI: 10.3390/ijms19082185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 01/11/2023] Open
Abstract
Dysfunctions of NO-cGMP signaling have been implicated in various neurological disorders. We have studied the potential crosstalk of cGMP and Ca2+ signaling in cerebellar granule neurons (CGNs) by simultaneous real-time imaging of these second messengers in living cells. The NO donor DEA/NO evoked cGMP signals in the granule cell layer of acute cerebellar slices from transgenic mice expressing a cGMP sensor protein. cGMP and Ca2+ dynamics were visualized in individual CGNs in primary cultures prepared from 7-day-old cGMP sensor mice. DEA/NO increased the intracellular cGMP concentration and augmented glutamate-induced Ca2+ transients. These effects of DEA/NO were absent in CGNs isolated from knockout mice lacking NO-sensitive guanylyl cyclase. Furthermore, application of the cGMP analogues 8-Br-cGMP and 8-pCPT-cGMP, which activate cGMP effector proteins such as cyclic nucleotide-gated cation channels and cGMP-dependent protein kinases (cGKs), also potentiated glutamate-induced Ca2+ transients. Western blot analysis failed to detect cGK type I or II in our primary CGNs. The addition of phosphodiesterase (PDE) inhibitors during cGMP imaging showed that CGNs degrade cGMP mainly via Zaprinast-sensitive PDEs, most likely PDE5 and/or PDE10, but not via PDE1, 2, or 3. In sum, these data delineate a cGK-independent NO-cGMP signaling cascade that increases glutamate-induced Ca2+ signaling in CGNs. This cGMP–Ca2+ crosstalk likely affects neurotransmitter-stimulated functions of CGNs.
Collapse
|
24
|
Heckman PRA, Blokland A, Bollen EPP, Prickaerts J. Phosphodiesterase inhibition and modulation of corticostriatal and hippocampal circuits: Clinical overview and translational considerations. Neurosci Biobehav Rev 2018; 87:233-254. [PMID: 29454746 DOI: 10.1016/j.neubiorev.2018.02.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/20/2022]
Abstract
The corticostriatal and hippocampal circuits contribute to the neurobiological underpinnings of several neuropsychiatric disorders, including Alzheimer's disease, Parkinson's disease and schizophrenia. Based on biological function, these circuits can be clustered into motor circuits, associative/cognitive circuits and limbic circuits. Together, dysfunctions in these circuits produce the wide range of symptoms observed in related neuropsychiatric disorders. Intracellular signaling in these circuits is largely mediated through the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway with an additional role for the cyclic guanosine monophosphate (cGMP)/ protein kinase G (PKG) pathway, both of which can be regulated by phosphodiesterase inhibitors (PDE inhibitors). Through their effects on cAMP response element-binding protein (CREB) and Dopamine- and cAMP-Regulated PhosphoProtein MR 32 kDa (DARPP-32), cyclic nucleotide pathways are involved in synaptic transmission, neuron excitability, neuroplasticity and neuroprotection. In this clinical review, we provide an overview of the current clinical status, discuss the general mechanism of action of PDE inhibitors in relation to the corticostriatal and hippocampal circuits and consider several translational challenges.
Collapse
Affiliation(s)
- P R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands.
| | - A Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands
| | - E P P Bollen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - J Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
25
|
Padovan-Neto FE, West AR. Regulation of Striatal Neuron Activity by Cyclic Nucleotide Signaling and Phosphodiesterase Inhibition: Implications for the Treatment of Parkinson's Disease. ADVANCES IN NEUROBIOLOGY 2018; 17:257-283. [PMID: 28956336 DOI: 10.1007/978-3-319-58811-7_10] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cyclic nucleotide phosphodiesterase (PDE) enzymes catalyze the hydrolysis and inactivation of cyclic nucleotides (cAMP/cGMP) in the brain. Several classes of PDE enzymes with distinct tissue distributions, cyclic nucleotide selectivity, and regulatory factors are highly expressed in brain regions subserving cognitive and motor processes known to be disrupted in neurodegenerative diseases such as Parkinson's disease (PD). Furthermore, small-molecule inhibitors of several different PDE family members alter cyclic nucleotide levels and favorably enhance motor performance and cognition in animal disease models. This chapter will explore the roles and therapeutic potential of non-selective and selective PDE inhibitors on neural processing in fronto-striatal circuits in normal animals and models of DOPA-induced dyskinesias (LIDs) associated with PD. The impact of selective PDE inhibitors and augmentation of cAMP and cGMP signaling on the membrane excitability of striatal medium-sized spiny projection neurons (MSNs) will be discussed. The effects of cyclic nucleotide signaling and PDE inhibitors on synaptic plasticity of striatonigral and striatopallidal MSNs will be also be reviewed. New data on the efficacy of PDE10A inhibitors for reversing behavioral and electrophysiological correlates of L-DOPA-induced dyskinesias in a rat model of PD will also be presented. Together, these data will highlight the potential of novel PDE inhibitors for treatment of movement disorders such as PD which are associated with abnormal corticostriatal transmission.
Collapse
Affiliation(s)
- Fernando E Padovan-Neto
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Anthony R West
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
26
|
The Role of Phosphodiesterase-2 in Psychiatric and Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2018; 17:307-347. [PMID: 28956338 DOI: 10.1007/978-3-319-58811-7_12] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cyclic nucleotide PDEs are a super-family of enzymes responsible for regulating intracellular levels of the second messengers cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Through their catalysis, PDEs are able to exert tight regulation over these important intracellular signaling cascades. Previously, PDEs have been implicated in learning and memory, as well as in mood disorders, such as anxiety and depression. PDE2 is of special interest due to its high level of expression in the forebrain, specifically in the isocortex, entorhinal cortex, striatum, hippocampus, amygdala, and medial habenula. Many of these brain regions are considered participants of the limbic system, which is known as the emotional regulatory center of the brain, and is important for modulating emotion and long-term memory. Therefore, PDE2s coincidental expression in these areas suggests an important role for PDE2 in these behaviors, and researchers are continuing to uncover the complex connections. It was shown that PDE2 inhibitors have pro-cognitive effects in tests of memory, including the object recognition test. PDE2 inhibitors are also protective against cognitive deficits in various models of cognitive impairment. Additionally, PDE2 inhibitors are protective against many different forms of stress-induced anxiety-like and depression-like behaviors. Currently, there is a great need for novel therapeutics for the treatment of mood and cognitive disorders, especially anxiety and depression, and other neurodegenerative diseases, such as Alzheimer's disease, and PDE2 is emerging as a viable target for future drug development for many of these diseases.
Collapse
|
27
|
Kelly MP. Cyclic nucleotide signaling changes associated with normal aging and age-related diseases of the brain. Cell Signal 2018; 42:281-291. [PMID: 29175000 PMCID: PMC5732030 DOI: 10.1016/j.cellsig.2017.11.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/21/2017] [Indexed: 01/23/2023]
Abstract
Deficits in brain function that are associated with aging and age-related diseases benefit very little from currently available therapies, suggesting a better understanding of the underlying molecular mechanisms is needed to develop improved drugs. Here, we review the literature to test the hypothesis that a break down in cyclic nucleotide signaling at the level of synthesis, execution, and/or degradation may contribute to these deficits. A number of findings have been reported in both the human and animal model literature that point to brain region-specific changes in Galphas (a.k.a. Gαs or Gsα), adenylyl cyclase, 3',5'-adenosine monophosphate (cAMP) levels, protein kinase A (PKA), cAMP response element binding protein (CREB), exchange protein activated by cAMP (Epac), hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs), atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), soluble and particulate guanylyl cyclase, 3',5'-guanosine monophosphate (cGMP), protein kinase G (PKG) and phosphodiesterases (PDEs). Among the most reproducible findings are 1) elevated circulating ANP and BNP levels being associated with cognitive dysfunction or dementia independent of cardiovascular effects, 2) reduced basal and/or NMDA-stimulated cGMP levels in brain with aging or Alzheimer's disease (AD), 3) reduced adenylyl cyclase activity in hippocampus and specific cortical regions with aging or AD, 4) reduced expression/activity of PKA in temporal cortex and hippocampus with AD, 5) reduced phosphorylation of CREB in hippocampus with aging or AD, 6) reduced expression/activity of the PDE4 family in brain with aging, 7) reduced expression of PDE10A in the striatum with Huntington's disease (HD) or Parkinson's disease, and 8) beneficial effects of select PDE inhibitors, particularly PDE10 inhibitors in HD models and PDE4 and PDE5 inhibitors in aging and AD models. Although these findings generally point to a reduction in cyclic nucleotide signaling being associated with aging and age-related diseases, there are exceptions. In particular, there is evidence for increased cAMP signaling specifically in aged prefrontal cortex, AD cerebral vessels, and PD hippocampus. Thus, if cyclic nucleotide signaling is going to be targeted effectively for therapeutic gain, it will have to be manipulated in a brain region-specific manner.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, VA Bldg 1, 3rd Floor, D-12, Columbia, SC 29209, United States.
| |
Collapse
|
28
|
Zhang C, Zhou Q, Wu XN, Huang YD, Zhou J, Lai Z, Wu Y, Luo HB. Discovery of novel PDE9A inhibitors with antioxidant activities for treatment of Alzheimer's disease. J Enzyme Inhib Med Chem 2017; 33:260-270. [PMID: 29271265 PMCID: PMC7011943 DOI: 10.1080/14756366.2017.1412315] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Phosphodiesterase-9 (PDE9) is a promising target for treatment of Alzheimer’s disease (AD). To discover multifunctional anti-AD agents with capability of PDE9 inhibition and antioxidant activity, a series of novel pyrazolopyrimidinone derivatives, coupling with the pharmacophore of antioxidants such as ferulic and lipolic acids have been designed with the assistance of molecular docking and dynamics simulations. Twelve out of 14 synthesised compounds inhibited PDE9A with IC50 below 200 nM, and showed good antioxidant capacities in the ORAC assay. Compound 1h, the most promising multifunctional anti-AD agent, had IC50 of 56 nM against PDE9A and good antioxidant ability (ORAC (trolox) = 3.3). The selectivity of 1h over other PDEs was acceptable. In addition, 1h showed no cytotoxicity to human neuroblastoma SH-SY5Y cells. The analysis on structure-activity relationship (SAR) and binding modes of the compounds may provide insight into further modification.
Collapse
Affiliation(s)
- Chen Zhang
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Qian Zhou
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Xu-Nian Wu
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Ya-Dan Huang
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Jie Zhou
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Zengwei Lai
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China.,b State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources , Guangxi Normal University , Guilin , PR China
| | - Yinuo Wu
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China
| | - Hai-Bin Luo
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , PR China.,c Collaborative Innovation Center of High Performance Computing , National University of Defence Technology , Changsha , PR China
| |
Collapse
|
29
|
Mikami S, Kawasaki M, Ikeda S, Negoro N, Nakamura S, Nomura I, Ashizawa T, Kokubo H, Hoffman ID, Zou H, Oki H, Uchiyama N, Hiura Y, Miyamoto M, Itou Y, Nakashima M, Iwashita H, Taniguchi T. Discovery of a Novel Series of Pyrazolo[1,5-a]pyrimidine-Based Phosphodiesterase 2A Inhibitors Structurally Different from N-((1S)-1-(3-Fluoro-4-(trifluoromethoxy)phenyl)-2-methoxyethyl)-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915), for the Treatment of Cognitive Disorders. Chem Pharm Bull (Tokyo) 2017; 65:1058-1077. [PMID: 29093293 DOI: 10.1248/cpb.c17-00564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been hypothesized that selective inhibition of phosphodiesterase (PDE) 2A could potentially be a novel approach to treat cognitive impairment in neuropsychiatric and neurodegenerative disorders through augmentation of cyclic nucleotide signaling pathways in brain regions associated with learning and memory. Following our earlier work, this article describes a drug design strategy for a new series of lead compounds structurally distinct from our clinical candidate 2 (TAK-915), and subsequent medicinal chemistry efforts to optimize potency, selectivity over other PDE families, and other preclinical properties including in vitro phototoxicity and in vivo rat plasma clearance. These efforts resulted in the discovery of N-((1S)-2-hydroxy-2-methyl-1-(4-(trifluoromethoxy)phenyl)propyl)-6-methyl-5-(3-methyl-1H-1,2,4-triazol-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxamide (20), which robustly increased 3',5'-cyclic guanosine monophosphate (cGMP) levels in the rat brain following an oral dose, and moreover, attenuated MK-801-induced episodic memory deficits in a passive avoidance task in rats. These data provide further support to the potential therapeutic utility of PDE2A inhibitors in enhancing cognitive performance.
Collapse
Affiliation(s)
- Satoshi Mikami
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Masanori Kawasaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Shuhei Ikeda
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Nobuyuki Negoro
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Shinji Nakamura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Izumi Nomura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Tomoko Ashizawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Hironori Kokubo
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | | | | | - Hideyuki Oki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Noriko Uchiyama
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Yuuto Hiura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Maki Miyamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Yuuki Itou
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Masato Nakashima
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Hiroki Iwashita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| | - Takahiko Taniguchi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company, Limited
| |
Collapse
|
30
|
Yu YF, Huang YD, Zhang C, Wu XN, Zhou Q, Wu D, Wu Y, Luo HB. Discovery of Novel Pyrazolopyrimidinone Derivatives as Phosphodiesterase 9A Inhibitors Capable of Inhibiting Butyrylcholinesterase for Treatment of Alzheimer's Disease. ACS Chem Neurosci 2017; 8:2522-2534. [PMID: 28783948 DOI: 10.1021/acschemneuro.7b00268] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Discovery of multitarget-directed ligands (MTDLs), targeting different factors simultaneously to control the complicated pathogenesis of Alzheimer's disease (AD), has become an important research area in recent years. Both phosphodiesterase 9A (PDE9A) and butyrylcholinesterase (BuChE) inhibitors could participate in different processes of AD to attenuate neuronal injuries and improve cognitive impairments. However, research on MTDLs combining the inhibition of PDE9A and BuChE simultaneously has not been reported yet. In this study, a series of novel pyrazolopyrimidinone-rivastigmine hybrids were designed, synthesized, and evaluated in vitro. Most compounds exhibited remarkable inhibitory activities against both PDE9A and BuChE. Compounds 6c and 6f showed the best IC50 values against PDE9A (6c, 14 nM; 6f, 17 nM) together with the considerable inhibition against BuChE (IC50, 6c, 3.3 μM; 6f, 0.97 μM). Their inhibitory potencies against BuChE were even higher than the anti-AD drug rivastigmine. It is worthy mentioning that both showed moderate selectivity for BuChE over acetylcholinesterase (AChE). Molecular docking studies revealed their binding patterns and explained the influence of configuration and substitutions on the inhibition of PDE9A and BuChE. Furthermore, compounds 6c and 6f exhibited negligible toxicity, which made them suitable for the further study of AD in vivo.
Collapse
Affiliation(s)
- Yan-Fa Yu
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Ya-Dan Huang
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Chen Zhang
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Xu-Nian Wu
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Qian Zhou
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Deyan Wu
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Yinuo Wu
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Hai-Bin Luo
- School of Pharmaceutical
Sciences, Sun Yat-Sen University, Guangzhou 510006, PR China
- Collaborative Innovation Center of High
Performance Computing, National University of Defense Technology, Changsha 410073, China
| |
Collapse
|
31
|
Mikami S, Nakamura S, Ashizawa T, Nomura I, Kawasaki M, Sasaki S, Oki H, Kokubo H, Hoffman ID, Zou H, Uchiyama N, Nakashima K, Kamiguchi N, Imada H, Suzuki N, Iwashita H, Taniguchi T. Discovery of Clinical Candidate N-((1S)-1-(3-Fluoro-4-(trifluoromethoxy)phenyl)-2-methoxyethyl)-7-methoxy-2-oxo-2,3-dihydropyrido[2,3-b]pyrazine-4(1H)-carboxamide (TAK-915): A Highly Potent, Selective, and Brain-Penetrating Phosphodiesterase 2A Inhibitor for the Treatment of Cognitive Disorders. J Med Chem 2017; 60:7677-7702. [PMID: 28796496 DOI: 10.1021/acs.jmedchem.7b00807] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Phosphodiesterase (PDE) 2A inhibitors have emerged as a novel mechanism with potential therapeutic option to ameliorate cognitive dysfunction in schizophrenia or Alzheimer's disease through upregulation of cyclic nucleotides in the brain and thereby achieve potentiation of cyclic nucleotide signaling pathways. This article details the expedited optimization of our recently disclosed pyrazolo[1,5-a]pyrimidine lead compound 4b, leading to the discovery of clinical candidate 36 (TAK-915), which demonstrates an appropriate combination of potency, PDE selectivity, and favorable pharmacokinetic (PK) properties, including brain penetration. Successful identification of 36 was realized through application of structure-based drug design (SBDD) to further improve potency and PDE selectivity, coupled with prospective design focused on physicochemical properties to deliver brain penetration. Oral administration of 36 demonstrated significant elevation of 3',5'-cyclic guanosine monophosphate (cGMP) levels in mouse brains and improved cognitive performance in a novel object recognition task in rats. Consequently, compound 36 was advanced into human clinical trials.
Collapse
Affiliation(s)
- Satoshi Mikami
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shinji Nakamura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomoko Ashizawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Izumi Nomura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masanori Kawasaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Shigekazu Sasaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideyuki Oki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hironori Kokubo
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Isaac D Hoffman
- Takeda California, Inc. , 10410 Science Center Drive, San Diego, California 92121, United States
| | - Hua Zou
- Takeda California, Inc. , 10410 Science Center Drive, San Diego, California 92121, United States
| | - Noriko Uchiyama
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kosuke Nakashima
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naomi Kamiguchi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Haruka Imada
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Noriko Suzuki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroki Iwashita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takahiko Taniguchi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited , 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
32
|
Prickaerts J, Heckman PRA, Blokland A. Investigational phosphodiesterase inhibitors in phase I and phase II clinical trials for Alzheimer's disease. Expert Opin Investig Drugs 2017; 26:1033-1048. [PMID: 28772081 DOI: 10.1080/13543784.2017.1364360] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Phosphodiesterase (PDE) inhibitors improve signaling pathways in brain circuits by increasing intracellular cyclic adenosine monophosphate (cAMP) and/or cyclic guanosine monophosphate (cGMP). In the last decade, the first clinical studies investigating selective PDE inhibitors in Alzheimer's disease (AD) have been initiated, based on their positive effects on cognitive processes and neuroprotection in numerous animal studies. Areas covered: This article reviews the clinical studies investigating the pro-cognitive/neuroprotective effects of PDE inhibitors in patients with AD, as well as in age-associated memory impaired elderly and patients with mild cognitive impairment (MCI), the prodromal stage of AD. PDE inhibitors will also be discussed with respect to adverse effects including safety and tolerability. Expert opinion: The limited available data of clinical studies with PDE inhibitors tested in different populations of AD patients do not allow the drawing of any concrete conclusion yet. Currently, studies with a PDE3 (cilostazol) or PDE9 inhibitor (BI 409,306) are still ongoing in patients with MCI or AD, respectively. Studies with PDE4 inhibitors (HT-0712, roflumilast and BPN14770) in healthy elderly and elderly with age-associated memory impairments indicate that the optimum dose and/or inhibiting the most relevant PDE isoform hold great promise when tested in the appropriate population of patients with MCI or AD eventually.
Collapse
Affiliation(s)
- Jos Prickaerts
- a Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience , Maastricht University , Maastricht , The Netherlands
| | - Pim R A Heckman
- a Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience , Maastricht University , Maastricht , The Netherlands.,b Department of Neuropsychology and Psychopharmacology , Maastricht University , Maastricht , The Netherlands
| | - Arjan Blokland
- b Department of Neuropsychology and Psychopharmacology , Maastricht University , Maastricht , The Netherlands
| |
Collapse
|
33
|
Knott EP, Assi M, Rao SNR, Ghosh M, Pearse DD. Phosphodiesterase Inhibitors as a Therapeutic Approach to Neuroprotection and Repair. Int J Mol Sci 2017; 18:E696. [PMID: 28338622 PMCID: PMC5412282 DOI: 10.3390/ijms18040696] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
A wide diversity of perturbations of the central nervous system (CNS) result in structural damage to the neuroarchitecture and cellular defects, which in turn are accompanied by neurological dysfunction and abortive endogenous neurorepair. Altering intracellular signaling pathways involved in inflammation and immune regulation, neural cell death, axon plasticity and remyelination has shown therapeutic benefit in experimental models of neurological disease and trauma. The second messengers, cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP), are two such intracellular signaling targets, the elevation of which has produced beneficial cellular effects within a range of CNS pathologies. The only known negative regulators of cyclic nucleotides are a family of enzymes called phosphodiesterases (PDEs) that hydrolyze cyclic nucleotides into adenosine monophosphate (AMP) or guanylate monophosphate (GMP). Herein, we discuss the structure and physiological function as well as the roles PDEs play in pathological processes of the diseased or injured CNS. Further we review the approaches that have been employed therapeutically in experimental paradigms to block PDE expression or activity and in turn elevate cyclic nucleotide levels to mediate neuroprotection or neurorepair as well as discuss both the translational pathway and current limitations in moving new PDE-targeted therapies to the clinic.
Collapse
Affiliation(s)
- Eric P Knott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Sudheendra N R Rao
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Neuroscience Program, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- Bruce Wayne Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
34
|
Hesse R, Lausser L, Gummert P, Schmid F, Wahler A, Schnack C, Kroker KS, Otto M, Tumani H, Kestler HA, Rosenbrock H, von Arnim CAF. Reduced cGMP levels in CSF of AD patients correlate with severity of dementia and current depression. Alzheimers Res Ther 2017; 9:17. [PMID: 28274265 PMCID: PMC5343324 DOI: 10.1186/s13195-017-0245-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/13/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder, primarily affecting memory. That disorder is thought to be a consequence of neuronal network disturbances and synapse loss. Decline in cognitive function is associated with a high burden of neuropsychiatric symptoms (NPSs) such as depression. The cyclic nucleotides cyclic adenosine-3',5'-monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) are essential second messengers that play a crucial role in memory processing as well as synaptic plasticity and are potential therapeutic targets. Biomarkers that are able to monitor potential treatment effects and that reflect the underlying pathology are of crucial interest. METHODS In this study, we measured cGMP and cAMP in cerebrospinal fluid (CSF) in a cohort of 133 subjects including 68 AD patients and 65 control subjects. To address the association with disease progression we correlated cognitive status with cyclic nucleotide levels. Because a high burden of NPSs is associated with decrease in cognitive function, we performed an exhaustive evaluation of AD-relevant marker combinations in a depressive subgroup. RESULTS We show that cGMP, but not cAMP, levels in the CSF of AD patients are significantly reduced compared with the control group. Reduced cGMP levels in AD patients correlate with memory impairment based on Mini-Mental State Examination score (r = 0.17, p = 0.048) and tau as a marker of neurodegeneration (r = -0.28, p = 0.001). Moreover, we were able to show that AD patients suffering from current depression show reduced cGMP levels (p = 0.07) and exhibit a higher degree of cognitive impairment than non-depressed AD patients. CONCLUSION These results provide further evidence for an involvement of cGMP in AD pathogenesis and accompanying co-morbidities, and may contribute to elucidating synaptic plasticity alterations during disease progression.
Collapse
Affiliation(s)
- Raphael Hesse
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Ludwig Lausser
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Pauline Gummert
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Florian Schmid
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Anke Wahler
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Cathrin Schnack
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Katja S. Kroker
- Department of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Holger Rosenbrock
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | | |
Collapse
|
35
|
Teich AF, Sakurai M, Patel M, Holman C, Saeed F, Fiorito J, Arancio O. PDE5 Exists in Human Neurons and is a Viable Therapeutic Target for Neurologic Disease. J Alzheimers Dis 2017; 52:295-302. [PMID: 26967220 PMCID: PMC4927884 DOI: 10.3233/jad-151104] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Phosphodiesterase 5 (PDE5) is a critical component of the cGMP-PKG axis of cellular signaling in neurons, and inhibition of PDE5 has been shown to be therapeutic in a wide range of neurologic conditions in animal models. However, enthusiasm for PDE5 inhibitors in humans is limited by data suggesting that PDE5 may not exist in human neurons. Here, we first show that past attempts to quantify PDE5 mRNA were flawed due to the use of incorrect primers, and that when correct primers are used, PDE5 mRNA is detectable in human brain tissue. We then show that PDE5 protein exists in human brain by western blot and ELISA. Most importantly, we performed immunohistochemistry and demonstrate that PDE5 is present in human neurons. We hope that this work will trigger a renewed interest in the development of PDE5 inhibitors for neurologic disease.
Collapse
Affiliation(s)
- Andrew F Teich
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Mikako Sakurai
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Mitesh Patel
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Cameron Holman
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Faisal Saeed
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Jole Fiorito
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| |
Collapse
|
36
|
Can Cyclic Nucleotide Phosphodiesterase Inhibitors Be Drugs for Parkinson's Disease? Mol Neurobiol 2017; 55:822-834. [PMID: 28062949 DOI: 10.1007/s12035-016-0355-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) has no known cure; available therapies are only capable of offering temporary, symptomatic relief to the patients. Varied therapeutic strategies that are clinically used for PD are pharmacological therapies including dopamine replacement therapies (with or without adjuvant), postsynaptic dopamine receptor stimulation, dopamine catabolism inhibitors and also anticholinergics. Surgical therapies like deep brain stimulation and ablative surgical techniques are also employed. Phosphodiesterases (PDEs) are enzymes that degrade the phosphodiester bond in the second messenger molecules, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). A number of PDE families are highly expressed in the striatum including PDE1-4, PDE7, PDE9 and PDE10. There are growing evidences to suggest that these enzymes play a critical role in modulating cAMP-mediated dopamine signalling at the postsynaptic region. Therefore, it is clear that PDEs, given the broad range of subtypes and their varied tissue- and region-specific distributions, will be able to provide a range of possibilities as drug targets. There is no phosphodiesterase inhibitor currently approved for use against PD. The development of small molecule inhibitors against cyclic nucleotide PDE is a particularly hot area of investigation, and a lot of research and development is geared in this direction with major players in the pharmaceutical industry investing heavily in developing such potential drug entities. This review, while critically assessing the existing body of literature on brain PDEs with particular interest in the striatum in the context of motor function regulation, indicates it is certainly likely that PDE inhibitors could be developed as therapeutic agents against PD.
Collapse
|
37
|
Boland K, Moschetti V, Dansirikul C, Pichereau S, Gheyle L, Runge F, Zimdahl-Gelling H, Sand M. A phase I, randomized, proof-of-clinical-mechanism study assessing the pharmacokinetics and pharmacodynamics of the oral PDE9A inhibitor BI 409306 in healthy male volunteers. Hum Psychopharmacol 2017; 32. [PMID: 28120486 DOI: 10.1002/hup.2569] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/14/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase (PDE) inhibitors are hypothesized to improve cognition in schizophrenia and Alzheimer disease by increasing cGMP levels in certain brain regions. This phase I, randomized, parallel-group, double-blind, placebo-controlled study provides proof-of-mechanism evidence for BI 409306, a novel, oral PDE9A inhibitor. METHODS In healthy males, exposure of BI 409306 (25-, 50-, 100-, and 200-mg single dose) and placebo was assessed in plasma and cerebrospinal fluid (CSF). Effect of BI 409306 on CSF cGMP levels was evaluated, and adverse events (AEs) were monitored. RESULTS In all enrolled subjects (N = 20), plasma BI 409306 concentration increased rapidly (median tmax : 0.75-1.25 hr) followed by rapid increases in CSF (median tmax : 1.5-2.0 hr). Maximum CSF cGMP concentrations were achieved within 2 to 5 hr, declining to baseline levels 10 to 14 hr after dosing. Dose-dependent increases in plasma and CSF exposure and CSF cGMP were shown. BI 409306 was safe and well tolerated. Most AEs were mild to moderate in intensity and study procedure-related. CONCLUSIONS BI 409306 increased rapidly in plasma and was subsequently detected in CSF, resulting in dose-dependent increases in cGMP levels in CSF. Results indicate BI 409306 efficiently crosses the blood-CSF barrier, with an acceptable level of AEs.
Collapse
Affiliation(s)
- Katja Boland
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | | - Solen Pichereau
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Lien Gheyle
- Clinical Research, SGS, Life Science Services, Antwerpen, Belgium
| | - Frank Runge
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Michael Sand
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| |
Collapse
|
38
|
Dorner-Ciossek C, Kroker KS, Rosenbrock H. Role of PDE9 in Cognition. ADVANCES IN NEUROBIOLOGY 2017; 17:231-254. [DOI: 10.1007/978-3-319-58811-7_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Fusco FR, Paldino E. Role of Phosphodiesterases in Huntington’s Disease. ADVANCES IN NEUROBIOLOGY 2017; 17:285-304. [DOI: 10.1007/978-3-319-58811-7_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
40
|
Heckman PRA, Blokland A, Prickaerts J. From Age-Related Cognitive Decline to Alzheimer's Disease: A Translational Overview of the Potential Role for Phosphodiesterases. ADVANCES IN NEUROBIOLOGY 2017; 17:135-168. [PMID: 28956332 DOI: 10.1007/978-3-319-58811-7_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Phosphodiesterase inhibitors (PDE-Is) are pharmacological compounds enhancing cAMP and/or cGMP signaling. Both these substrates affect neural communication by influencing presynaptic neurotransmitter release and postsynaptic intracellular pathways after neurotransmitter binding to its receptor. Both cAMP and cGMP play an important role in a variety of cellular functions including neuroplasticity and neuroprotection. This chapter provides a translational overview of the effects of different classes of PDE-Is on cognition enhancement in age-related cognitive decline and Alzheimer's disease (AD). The most effective PDE-Is in preclinical models of aging and AD appear to be PDE2-Is, PDE4-Is and PDE5-Is. Clinical studies are relatively sparse and so far PDE1-Is and PDE4-Is showed some promising results. In the future, the demonstration of clinical proof of concept and the generation of isoform selective PDE-Is are the hurdles to overcome in developing safe and efficacious novel PDE-Is for the treatment of age-related cognitive decline and cognitive dysfunction in AD.
Collapse
Affiliation(s)
- Pim R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
- Department of Neuropsychology and Psychopharmacology, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200, MD, Maastricht, The Netherlands.
| |
Collapse
|
41
|
Pharmacological Rescue of Long-Term Potentiation in Alzheimer Diseased Synapses. J Neurosci 2016; 37:1197-1212. [PMID: 27986924 DOI: 10.1523/jneurosci.2774-16.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/05/2016] [Accepted: 12/09/2016] [Indexed: 01/05/2023] Open
Abstract
Long-term potentiation (LTP) is an activity-dependent and persistent increase in synaptic transmission. Currently available techniques to measure LTP are time-intensive and require highly specialized expertise and equipment, and thus are not well suited for screening of multiple candidate treatments, even in animal models. To expand and facilitate the analysis of LTP, here we use a flow cytometry-based method to track chemically induced LTP by detecting surface AMPA receptors in isolated synaptosomes: fluorescence analysis of single-synapse long-term potentiation (FASS-LTP). First, we demonstrate that FASS-LTP is simple, sensitive, and models electrically induced LTP recorded in intact circuitries. Second, we conducted FASS-LTP analysis in two well-characterized Alzheimer's disease (AD) mouse models (3xTg and Tg2576) and, importantly, in cryopreserved human AD brain samples. By profiling hundreds of synaptosomes, our data provide the first direct evidence to support the idea that synapses from AD brain are intrinsically defective in LTP. Third, we used FASS-LTP for drug evaluation in human synaptosomes. Testing a panel of modulators of cAMP and cGMP signaling pathways, FASS-LTP identified vardenafil and Bay-73-6691 (phosphodiesterase-5 and -9 inhibitors, respectively) as potent enhancers of LTP in synaptosomes from AD cases. These results indicate that our approach could provide the basis for protocols to study LTP in both healthy and diseased human brains, a previously unattainable goal. SIGNIFICANCE STATEMENT Learning and memory depend on the ability of synapses to strengthen in response to activity. Long-term potentiation (LTP) is a rapid and persistent increase in synaptic transmission that is thought to be affected in Alzheimer's disease (AD). However, direct evidence of LTP deficits in human AD brain has been elusive, primarily due to methodological limitations. Here, we analyze LTP in isolated synapses from AD brain using a novel approach that allows testing LTP in cryopreserved brain. Our analysis of hundreds of synapses supports the idea that AD-diseased synapses are intrinsically defective in LTP. Further, we identified pharmacological agents that rescue LTP in AD, thus opening up a new avenue for drug screening and evaluation of strategies for alleviating memory impairments.
Collapse
|
42
|
Trabanco AA, Buijnsters P, Rombouts FJR. Towards selective phosphodiesterase 2A (PDE2A) inhibitors: a patent review (2010 - present). Expert Opin Ther Pat 2016; 26:933-46. [PMID: 27321640 DOI: 10.1080/13543776.2016.1203902] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The cyclic nucleotides cAMP and cGMP are ubiquitous intracellular second messengers regulating a large variety of biological processes. The intracellular concentration of these biologically relevant molecules is modulated by the activity of phosphodiesterases (PDEs), a class of enzymes that is grouped in 11 families. The expression of PDEs is tissue- and cell-specific allowing spatiotemporal integration of multiple signaling cascades. PDE2A is a dual substrate enzyme and is expressed in both the periphery and in the central nervous system, however its expression is highest in the brain, where it is mainly localized in the cortex, hippocampus, and striatum. This suggests that this enzyme may regulate intraneuronal cGMP and cAMP levels in brain areas involved in emotion, perception, concentration, learning and memory. AREAS COVERED This review covers the patent applications published between January 2010 and February 2016 on phosphodiesterase 2A inhibitors. EXPERT OPINION Recent publications in the literature and in filed patent applications demonstrate the interest of pharmaceutical companies for PDE2A. This has increased the insights of its possible therapeutic role but the few clinical trials were terminated. Based on the ongoing interest in the field it is likely that new clinical trials can be expected and will unravel the therapeutic potential of PDE2A inhibition.
Collapse
Affiliation(s)
- Andrés A Trabanco
- a A Division of Janssen-Cilag S.A., Neuroscience Medicinal Chemistry Department , Janssen Research and Development , Toledo , Spain
| | - Peter Buijnsters
- b A Division of Janssen Pharmaceutica N.V., Neuroscience Medicinal Chemistry Department , Janssen Research and Development , Beerse , Belgium
| | - Frederik J R Rombouts
- b A Division of Janssen Pharmaceutica N.V., Neuroscience Medicinal Chemistry Department , Janssen Research and Development , Beerse , Belgium
| |
Collapse
|
43
|
Su T, Zhang T, Xie S, Yan J, Wu Y, Li X, Huang L, Luo HB. Discovery of novel PDE9 inhibitors capable of inhibiting Aβ aggregation as potential candidates for the treatment of Alzheimer's disease. Sci Rep 2016; 6:21826. [PMID: 26911795 PMCID: PMC4766439 DOI: 10.1038/srep21826] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 02/01/2016] [Indexed: 12/20/2022] Open
Abstract
Recently, phosphodiesterase-9 (PDE9) inhibitors and biometal-chelators have received much attention as potential therapeutics for the treatment of Alzheimer’s disease (AD). Here, we designed, synthesized, and evaluated a novel series of PDE9 inhibitors with the ability to chelate metal ions. The bioassay results showed that most of these molecules strongly inhibited PDE9 activity. Compound 16 showed an IC50 of 34 nM against PDE9 and more than 55-fold selectivity against other PDEs. In addition, this compound displayed remarkable metal-chelating capacity and a considerable ability to halt copper redox cycling. Notably, in comparison to the reference compound clioquinol, it inhibited metal-induced Aβ1-42 aggregation more effectively and promoted greater disassembly of the highly structured Aβ fibrils generated through Cu2+-induced Aβ aggregation. These activities of 16, together with its favorable blood-brain barrier permeability, suggest that 16 may be a promising compound for treatment of AD.
Collapse
Affiliation(s)
- Tao Su
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shishun Xie
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jun Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yinuo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ling Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
44
|
Ugarte A, Gil-Bea F, García-Barroso C, Cedazo-Minguez Á, Ramírez MJ, Franco R, García-Osta A, Oyarzabal J, Cuadrado-Tejedor M. Decreased levels of guanosine 3', 5'-monophosphate (cGMP) in cerebrospinal fluid (CSF) are associated with cognitive decline and amyloid pathology in Alzheimer's disease. Neuropathol Appl Neurobiol 2016; 41:471-82. [PMID: 25488891 DOI: 10.1111/nan.12203] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/03/2014] [Indexed: 01/03/2023]
Abstract
AIMS Levels of the cyclic nucleotides guanosine 3', 5'-monophosphate (cGMP) or adenosine 3', 5'-monophosphate (cAMP) that play important roles in memory processes are not characterized in Alzheimer's disease (AD). The aim of this study was to analyse the levels of these nucleotides in cerebrospinal fluid (CSF) samples from patients diagnosed with clinical and prodromal stages of AD and study the expression level of the enzymes that hydrolyzed them [phosphodiesterases (PDEs)] in the brain of AD patients vs. METHODS For cGMP and cAMP CSF analysis, the cohort (n = 79) included cognitively normal participants (subjective cognitive impairment), individuals with stable mild cognitive impairment or AD converters (sMCI and cMCI), and mild AD patients. A high throughput liquid chromatography-tandem mass spectrometry method was used. Interactions between CSF cGMP or cAMP with mini-mental state examination (MMSE) score, CSF Aβ(1-42) and CSF p-tau were analysed. For PDE4, 5, 9 and 10 expression analysis, brains of AD patients vs. controls (n = 7 and n = 8) were used. RESULTS cGMP, and not cAMP levels, were significantly lower in the CSF of patients diagnosed with mild AD when compared with nondemented controls. CSF levels of cGMP showed a significant association with MMSE-diagnosed clinical dementia and with CSF biomarker Aβ42 in AD patients. Significant increase in PDE5 expression was detected in temporal cortex of AD patients compared with that of age-matched healthy control subjects. No changes in the expression of others PDEs were detected. CONCLUSIONS These results support the potential involvement of cGMP in the pathological and clinical development of AD. The cGMP reduction in early stages of AD might participate in the aggravation of amyloid pathology and cognitive decline.
Collapse
Affiliation(s)
- Ana Ugarte
- Small Molecule Discovery Platform, Molecular Therapeutics Program, University of Navarra, Pamplona, Spain
| | - Francisco Gil-Bea
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Carolina García-Barroso
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Ángel Cedazo-Minguez
- KI-Alzheimer's Disease Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - M Javier Ramírez
- Department of Pharmacology, University of Navarra, Pamplona, Spain
| | - Rafael Franco
- Department of Biochemistry and Molecular Biology, Universitat de Barcelona, Barcelona, Spain
| | - Ana García-Osta
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, University of Navarra, Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Neurobiology of Alzheimer's Disease, Neurosciences Division, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.,Department of Anatomy, University of Navarra, Pamplona, Spain
| |
Collapse
|
45
|
Purin-6-One Derivatives as Phosphodiesterase-2 Inhibitors. J CHEM-NY 2016. [DOI: 10.1155/2016/6878353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A series of purin-6-one derivatives were synthesized, and theirin vitroinhibitory activity against phosphodiesterase-2 (PDE2) was evaluated by using a fluorescence polarization assay. Three compounds, that are,2j,2p, and2q, showed significant inhibitory activity against PDE2 with IC50values of 1.73, 0.18, and 3.43 μM, respectively. Structure-activity relationship (SAR) analysis was performed to explore the relationship between the chemical structures of these compounds and their inhibitory activity. Compounds2j,2p, and2qwere further selected for molecular docking study. The docking results suggested that these ligands bind with hydrophobic pockets of the catalytic active site of PDE2, where a Tyr655 residue was found to be important in binding with compound2p, the most potent inhibitor identified in this study. Our present study provides useful information for the future design of novel PDE2 inhibitors.
Collapse
|
46
|
Redrobe JP, Rasmussen LK, Christoffersen CT, Bundgaard C, Jørgensen M. Characterisation of Lu AF33241: A novel, brain-penetrant, dual inhibitor of phosphodiesterase (PDE) 2A and PDE10A. Eur J Pharmacol 2015; 761:79-85. [DOI: 10.1016/j.ejphar.2015.04.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/01/2015] [Accepted: 04/16/2015] [Indexed: 11/26/2022]
|
47
|
Herbs to curb cyclic nucleotide phosphodiesterase and their potential role in Alzheimer's disease. Mech Ageing Dev 2015; 149:75-87. [PMID: 26050556 DOI: 10.1016/j.mad.2015.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/22/2015] [Accepted: 05/27/2015] [Indexed: 01/02/2023]
Abstract
Cyclic nucleotides viz., cAMP/cGMP has been well known to play important role in cellular function and deficiency in their levels has been implicated in the pathogenesis of various neurodegenerative disorders including Alzheimer's disease (AD). Phosphodiesterases (PDE) are the enzymes involved in the metabolism of cyclic nucleotides and the inhibition of phosphodiesterases is considered to be viable strategy to restore the level of cyclic nucleotides and their functions in the brain. Various synthetic PDE inhibitors had been used clinically for various disorders and also suggested to be useful candidates for treating neurological disorders. However, side effects of these synthetic PDE inhibitors have limited their use in clinical practice. Natural plant extracts or their bio-active compounds are considered to be safe and are widely acceptable. During the last decade, many plant extracts or their bio-active compounds were tested pre-clinically for PDE inhibitory activity and are reported to be equally potent in inhibiting PDE's, as that of synthetic compounds. The present review is aimed to discuss the potential plant extract/compounds with PDE inhibitory activity and critically discuss their potential role in Alzheimer's disease.
Collapse
|
48
|
Umar T, Hoda N. Selective inhibitors of phosphodiesterases: therapeutic promise for neurodegenerative disorders. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00419e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PDE inhibitors: significant contributors to the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Tarana Umar
- Department of Chemistry
- Jamia Millia Islamia
- Central University
- New Delhi
- 110025 India
| | - Nasimul Hoda
- Department of Chemistry
- Jamia Millia Islamia
- Central University
- New Delhi
- 110025 India
| |
Collapse
|
49
|
Redrobe JP, Jørgensen M, Christoffersen CT, Montezinho LP, Bastlund JF, Carnerup M, Bundgaard C, Lerdrup L, Plath N. In vitro and in vivo characterisation of Lu AF64280, a novel, brain penetrant phosphodiesterase (PDE) 2A inhibitor: potential relevance to cognitive deficits in schizophrenia. Psychopharmacology (Berl) 2014; 231:3151-67. [PMID: 24577516 DOI: 10.1007/s00213-014-3492-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/05/2014] [Indexed: 12/25/2022]
Abstract
Here, we present the pharmacological characterisation of Lu AF64280, a novel, selective, brain penetrant phosphodiesterase (PDE) 2A inhibitor, in in vitro/in vivo assays indicative of PDE2A inhibition, and in vivo models/assays relevant to cognitive processing or antipsychotic-like activity. The in vitro selectivity of Lu AF64280 was determined against a panel of PDE enzymes and 3',5'-cyclic guanosine monophosphate (cGMP) levels in the hippocampus were determined using in vivo microdialysis. Lu AF64280 potently inhibited hPDE2A (Ki = 20 nM), 50-fold above moderate inhibition of both hPDE9A (Ki = 1,000 nM) and hPDE10A (Ki = 1,800 nM), and displayed a >250-fold selectivity over all other full-length human recombinant PDE family members (Ki above 5,000 nM). Lu AF64280 (20 mg/kg) significantly increased cGMP levels in the hippocampus (p < 0.01 versus vehicle-treated mice), attenuated sub-chronic phencyclidine-induced deficits in novel object exploration in rats (10 mg/kg, p < 0.001 versus vehicle-treated), blocked early postnatal phencyclidine-induced deficits in the intradimensional/extradimensional shift task in rats (1 and 10 mg/kg, p < 0.001 versus vehicle-treated) and attenuated spontaneous P20-N40 auditory gating deficits in DBA/2 mice (20 mg/kg, p < 0.05 versus vehicle-treated). In contrast, Lu AF64280 failed to attenuate phencyclidine-induced hyperactivity in mice, and was devoid of antipsychotic-like activity in the conditioned avoidance response paradigm in rats, at any dose tested. Lu AF64280 represents a novel tool compound for selective PDE2A inhibition that substantiates a critical role of this enzyme in cognitive processes under normal and pathological conditions.
Collapse
Affiliation(s)
- John P Redrobe
- Neuroscience Research DK, H. Lundbeck A/S, Ottiliavej 9, Valby, 2500, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Akar F, Mutlu O, Komsuoglu Celikyurt I, Ulak G, Erden F, Bektas E, Tanyeri P. Zaprinast and rolipram enhances spatial and emotional memory in the elevated plus maze and passive avoidance tests and diminishes exploratory activity in naive mice. Med Sci Monit Basic Res 2014; 20:105-11. [PMID: 25057848 PMCID: PMC4117679 DOI: 10.12659/msmbr.891149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Phosphodiesterase (PDE) inhibitors in the central nervous system have been shown to stimulate neuronal functions and increase neurogenesis in Alzheimer disease (AD) patients. Material/Methods The aim of this study was to investigate the effects of zaprinast, a PDE5 inhibitor, and rolipram, a PDE4 inhibitor, on learning and memory in elevated plus maze (EPM) and passive avoidance (PA) tests in naive mice. Male Balb-c mice received short-term treatment with zaprinast (3 and 10 mg/kg) and rolipram (0.05 and 0.1 mg/kg) before the acquisition trial of the EPM and PA tests. The exploratory activity of the animals was also investigated in the Hughes box test. Results Both zaprinast (10 mg/kg) and rolipram (0.1 mg/kg) significantly decreased second-day latency compared to the control group in the EPM test, while only rolipram (0.1 mg/kg) significantly increased second-day latency in the PA test. Both zaprinast (10 mg/kg) and rolipram (0.1 mg/kg) significantly decreased the number of entries to new areas and time spent in new areas in the Hughes box test. Conclusions Our study revealed that both zaprinast and rolipram enhanced spatial memory in EPM, while rolipram seemed to have more emotional memory-enhancing effects in the PA test compared to zaprinast. Both zaprinast and rolipram diminished exploratory activity in the Hughes box test, which can be attributed to the drugs’ anxiogenic effects.
Collapse
Affiliation(s)
- Furuzan Akar
- Department of Pharmacology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | - Oguz Mutlu
- Department of Pharmacology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | | | - Guner Ulak
- Department of Pharmacology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | - Faruk Erden
- Department of Pharmacology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | - Emine Bektas
- Department of Pharmacology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | - Pelin Tanyeri
- Department of Pharmacology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| |
Collapse
|