1
|
Xiang Y, Naik S, Zhao L, Shi J, Ke H. Emerging phosphodiesterase inhibitors for treatment of neurodegenerative diseases. Med Res Rev 2024; 44:1404-1445. [PMID: 38279990 DOI: 10.1002/med.22017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/29/2024]
Abstract
Neurodegenerative diseases (NDs) cause progressive loss of neuron structure and ultimately lead to neuronal cell death. Since the available drugs show only limited symptomatic relief, NDs are currently considered as incurable. This review will illustrate the principal roles of the signaling systems of cyclic adenosine and guanosine 3',5'-monophosphates (cAMP and cGMP) in the neuronal functions, and summarize expression/activity changes of the associated enzymes in the ND patients, including cyclases, protein kinases, and phosphodiesterases (PDEs). As the sole enzymes hydrolyzing cAMP and cGMP, PDEs are logical targets for modification of neurodegeneration. We will focus on PDE inhibitors and their potentials as disease-modifying therapeutics for the treatment of Alzheimer's disease, Parkinson's disease, and Huntington's disease. For the overlapped but distinct contributions of cAMP and cGMP to NDs, we hypothesize that dual PDE inhibitors, which simultaneously regulate both cAMP and cGMP signaling pathways, may have complementary and synergistic effects on modifying neurodegeneration and thus represent a new direction on the discovery of ND drugs.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Swapna Naik
- Department of Pharmacology, Yale Cancer Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Rombaut B, Schepers M, Tiane A, Mussen F, Koole L, Kessels S, Trippaers C, Jacobs R, Wouters K, Willems E, van Veggel L, Koulousakis P, Deluyker D, Bito V, Prickaerts J, Wens I, Brône B, van den Hove DLA, Vanmierlo T. Early Inhibition of Phosphodiesterase 4B (PDE4B) Instills Cognitive Resilience in APPswe/PS1dE9 Mice. Cells 2024; 13:1000. [PMID: 38920631 PMCID: PMC11201979 DOI: 10.3390/cells13121000] [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: 03/13/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia activity can drive excessive synaptic loss during the prodromal phase of Alzheimer's disease (AD) and is associated with lowered cyclic adenosine monophosphate (cAMP) due to cAMP phosphodiesterase 4B (PDE4B). This study aimed to investigate whether long-term inhibition of PDE4B by A33 (3 mg/kg/day) can prevent synapse loss and its associated cognitive decline in APPswe/PS1dE9 mice. This model is characterized by a chimeric mouse/human APP with the Swedish mutation and human PSEN1 lacking exon 9 (dE9), both under the control of the mouse prion protein promoter. The effects on cognitive function of prolonged A33 treatment from 20 days to 4 months of age, was assessed at 7-8 months. PDE4B inhibition significantly improved both the working and spatial memory of APPswe/PSdE9 mice after treatment ended. At the cellular level, in vitro inhibition of PDE4B induced microglial filopodia formation, suggesting that regulation of PDE4B activity can counteract microglia activation. Further research is needed to investigate if this could prevent microglia from adopting their 'disease-associated microglia (DAM)' phenotype in vivo. These findings support the possibility that PDE4B is a potential target in combating AD pathology and that early intervention using A33 may be a promising treatment strategy for AD.
Collapse
Affiliation(s)
- Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Femke Mussen
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Lisa Koole
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Sofie Kessels
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Chloë Trippaers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Ruben Jacobs
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Center+ (MUMC+), 6229 ER Maastricht, The Netherlands;
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Emily Willems
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Lieve van Veggel
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Philippos Koulousakis
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Dorien Deluyker
- UHasselt, Cardio & Organ Systems (COST), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium; (D.D.); (V.B.)
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium; (D.D.); (V.B.)
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Inez Wens
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Bert Brône
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Daniel L. A. van den Hove
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, 97070 Wuerzburg, Germany
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| |
Collapse
|
3
|
Tropea MR, Gulisano W, Vacanti V, Arancio O, Puzzo D, Palmeri A. Nitric oxide/cGMP/CREB pathway and amyloid-beta crosstalk: From physiology to Alzheimer's disease. Free Radic Biol Med 2022; 193:657-668. [PMID: 36400326 DOI: 10.1016/j.freeradbiomed.2022.11.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/30/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Abstract
The nitric oxide (NO)/cGMP pathway has been extensively studied for its pivotal role in synaptic plasticity and memory processes, resulting in an increase of cAMP response element-binding (CREB) phosphorylation, and consequent synthesis of plasticity-related proteins. The NO/cGMP/CREB signaling is downregulated during aging and neurodegenerative disorders and is affected by Amyloid-β peptide (Aβ) and tau protein, whose increase and deposition is considered the key pathogenic event of Alzheimer's disease (AD). On the other hand, in physiological conditions, the crosstalk between the NO/cGMP/PKG/CREB pathway and Aβ ensures long-term potentiation and memory formation. This review summarizes the current knowledge on the interaction between the NO/cGMP/PKG/CREB pathway and Aβ in the healthy and diseased brain, offering a new perspective to shed light on AD pathophysiology. We will focus on the synaptic mechanisms underlying Aβ physiological interplay with cGMP pathway and how this balance is corrupted in AD, as high levels of Aβ interfere with NO production and cGMP molecular signaling leading to cognitive impairment. Finally, we will discuss results from preclinical and clinical studies proposing the increase of cGMP signaling as a therapeutic strategy in the treatment of AD.
Collapse
Affiliation(s)
- Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Valeria Vacanti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, USA; Department of Pathology & Cell Biology and Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy; Oasi Research Institute-IRCCS, Troina (EN), 94018, Italy.
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, 95123, Italy
| |
Collapse
|
4
|
Soluble guanylate cyclase stimulator riociguat improves spatial memory in mice via peripheral mechanisms. Neurosci Lett 2022; 788:136840. [PMID: 35985509 DOI: 10.1016/j.neulet.2022.136840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/10/2022] [Accepted: 08/11/2022] [Indexed: 01/02/2023]
Abstract
Soluble guanylate cyclase (sGC) - cyclic guanosine monophosphate (cGMP) signalling is important for healthy memory function and a healthy vascular system. Targeting sGC-cGMP signalling can therefore be a potential strategy to enhance memory processes. sGC can be targeted by using agonists, such as sGC stimulator riociguat. Therefore, this study aimed to target sGC using riociguat to investigate its acute effects on memory function and neuronal plasticity in mice. The effects of riociguat on long-term memory and a biperiden-induced memory deficit model for assessing short-term memory were tested in the object location task, and working memory was tested in the Y-maze continuous alternation task. Pharmacokinetic measurements were performed within brain tissue of mice, and hippocampal plasticity measures were assessed using western blotting. Acute oral administration with a low dose of 0.03 mg/kg riociguat was able to enhance working-, short-, and long-term spatial memory. Under cerebral vasoconstriction higher doses of riociguat were still effective on memory. Pharmacokinetic measurements revealed poor brain penetration of riociguat and its metabolite M-1. Increased activation of VASP was found, while no effects were found on other memory-related hippocampal plasticity measures. Memory enhancing effects of riociguat are most likely regulated by vascular peripheral effects on cGMP signalling. Yet, further research is needed to investigate the possible contribution of hemodynamic or metabolic effects of sGC stimulators on memory performance.
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: 0.3] [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
|
Zhang Y, Qian L, Liu Y, Liu Y, Yu W, Zhao Y. CircRNA-ceRNA Network Revealing the Potential Regulatory Roles of CircRNA in Alzheimer's Disease Involved the cGMP-PKG Signal Pathway. Front Mol Neurosci 2021; 14:665788. [PMID: 34093124 PMCID: PMC8176118 DOI: 10.3389/fnmol.2021.665788] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a chronic progressive neurodegenerative disease. The characteristic pathologies include extracellular senile plaques formed by β-amyloid protein deposition, neurofibrillary tangles formed by hyperphosphorylation of tau protein, and neuronal loss with glial cell hyperplasia. Circular RNAs (circRNAs) are rich in miRNA-binding sites (miRNA response elements, MREs), which serve as miRNA sponges or competitive endogenous RNAs (ceRNAs). Although several research groups have identified dysregulated circRNAs in the cerebral cortex of SAMP8 mice or APP/PS1 mice using deep RNA-seq analysis, we need to further explore circRNA expression patterns, targets, functions and the signaling pathways involved in the pathogenesis of AD and in particular the hippocampal circRNA expression profiles in AD. Methods: We used deep RNA sequencing to investigate circRNA-ceRNA network patterns in the hippocampus of APP/PS1 mice. Results: In our study, 70 dysregulated circRNAs, 39 dysregulated miRNAs and 121 dysregulated mRNAs were identified between the APP/PS1 group and the wild-type group at 8 months in the hippocampus of the mice. Through correlation analysis, we identified 11 dysregulated circRNAs, 7 dysregulated miRNAs and 8 dysregulated mRNAs forming 16 relationships in the circRNA-miRNA-mRNA regulatory network. Gene ontology (GO) analysis indicated that the dysregulated circRNAs were most enriched in biological metabolic processes. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the dysregulation of circRNAs was enriched in the cGMP-PKG signaling pathway, cAMP signaling pathway, Hippo signaling pathway, platelet activation, long-term potentiation and axon guidance. In addition, our findings preliminarily verified that the novel_circ_0003012/mmu-miR-298-3p/Smoc2 signaling axis may regulate the pathophysiology of AD by affecting the cGMP-PKG signaling pathway. Conclusions: These newly identified circRNAs in networks and signaling pathways reveal potential diagnostic or therapeutic targets for AD.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Lili Qian
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yingying Liu
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Ying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Yanfang Zhao
- Institute of Biomedical Research, School for Life Sciences, Shandong University of Technology, Zibo, China
| |
Collapse
|
7
|
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
|
8
|
Paes D, Xie K, Wheeler DG, Zook D, Prickaerts J, Peters M. Inhibition of PDE2 and PDE4 synergistically improves memory consolidation processes. Neuropharmacology 2021; 184:108414. [PMID: 33249120 DOI: 10.1016/j.neuropharm.2020.108414] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 10/21/2020] [Accepted: 11/23/2020] [Indexed: 01/10/2023]
Abstract
Phosphodiesterases (PDE) are the only enzymes that degrade cAMP and cGMP which are second messengers crucial to memory consolidation. Different PDE inhibitors have been developed and tested for their memory-enhancing potential, but the occurrence of side effects has hampered clinical progression. As separate inhibition of the PDE2 and PDE4 enzyme family has been shown to enhance memory, we investigated whether concurrent treatment with a PDE2 and PDE4 inhibitor can have synergistic effects on memory consolidation processes. We found that combined administration of PF-999 (PDE2 inhibitor) and roflumilast (PDE4 inhibitor) increases the phosphorylation of the AMPA receptor subunit GluR1 and induces CRE-mediated gene expression. Moreover, when combined sub-effective and effective doses of PF-999 and roflumilast were administered after learning, time-dependent forgetting was abolished in an object location memory task. Pharmacokinetic assessment indicated that combined treatment does not alter exposure of the individual compounds. Taken together, these findings suggest that combined PDE2 and PDE4 inhibition has synergistic effects on memory consolidation processes at sub-effective doses, which could therefore provide a therapeutic strategy with an improved safety profile.
Collapse
Affiliation(s)
- Dean Paes
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6229, ER Maastricht, the Netherlands
| | - Keqiang Xie
- In Vitro Pharmacology, Dart Neuroscience, LLC, 12278 Scripps Summit Drive, San Diego, CA, 92131, USA
| | - Damian G Wheeler
- Target Discovery & Behavioral Pharmacology, Dart Neuroscience, LLC, 12278 Scripps Summit Drive, San Diego, CA, 92131, USA
| | - Douglas Zook
- DMPK, Dart Neuroscience, LLC, 12278 Scripps Summit Drive, San Diego, CA, 92131, USA
| | - Jos Prickaerts
- School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6229, ER Maastricht, the Netherlands
| | - Marco Peters
- Target Discovery & Behavioral Pharmacology, Dart Neuroscience, LLC, 12278 Scripps Summit Drive, San Diego, CA, 92131, USA; Neurobiology and Behavior & Center for the Neurobiology of Learning and Memory, University of California Irvine, 213 Qureshey Research Lab, Irvine, CA, 92697, USA.
| |
Collapse
|
9
|
Contributions of animal models of cognitive disorders to neuropsychopharmacology. Therapie 2021; 76:87-99. [PMID: 33589315 DOI: 10.1016/j.therap.2021.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/30/2021] [Indexed: 12/18/2022]
Abstract
Cognitive disorders and symptoms are key features of many mental and neurological diseases, with a large spectrum of impaired domains. Because of their possible evolution and detrimental functioning impact, they are a major pharmacological target for both symptomatic and disease-modifier drugs, while few cognitive enhancers have been marketed with an insufficient efficiency. It explains the need to model these cognitive disorders beyond the modelization of mental or neurological diseases themselves. According to the experimental strategy used to induce cognitive impairment, three categories of models have been identified: neurotransmission-driven models; pathophysiology-driven models; environment-driven models. These three categories of models reflect different levels of integration of endogenous and exogenous mechanisms underlying cognitive disorders in humans. Their comprehensive knowledge and illustration of their pharmacological modulation could help to propose a renewing strategy of drug development in central nervous system (CNS) field at a time when the academic and industrial invest seems to be declining despite the medical and social burden of brain diseases.
Collapse
|
10
|
Chen L, Liu K, Wang Y, Liu N, Yao M, Hu J, Wang G, Sun Y, Pan J. Phosphodiesterase-2 inhibitor reverses post-traumatic stress induced fear memory deficits and behavioral changes via cAMP/cGMP pathway. Eur J Pharmacol 2021; 891:173768. [PMID: 33271150 DOI: 10.1016/j.ejphar.2020.173768] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 01/28/2023]
Abstract
Phosphodiesterase 2 is one of the phosphodiesterase (PDEs) family members that regulate cyclic nucleotide (namely cAMP and cGMP) concentrations. The present study determined whether PDE2 inhibition could rescue post-traumatic stress disorder (PTSD)-like symptoms. Mice were subjected to single prolonged stress (SPS) and treated with selective PDE2 inhibitor Bay 60-7550 (0.3, 1, or 3 mg/kg, i.p.). The behavioral tests such as forced swimming, sucrose preference test, open field, elevated plus maze, and contextual fear paradigm were conducted to determine the effects of Bay 60-7550 on SPS-induced depression- and anxiety-like behavior and fear memory deficits. The results suggested that Bay 60-7550 reversed SPS-induced depression- and anxiety-like behavior and fear memory deficits. Moreover, Bay 60-7550 prevented SPS-induced changes in the adrenal gland index, synaptic proteins synaptophysin and PSD95 expression, PKA, PKG, pCREB, and BDNF levels in the hippocampus and amygdala. These effects were completely prevented by PKG inhibitor KT5823. While PKA inhibitor H89 also prevented Bay 60-7550-induced pCREB and BDNF expression, but only partially prevented the effects on PSD95 expression in the hippocampus. These findings suggest that Bay 60-7550 protects mice against PTSD-like stress induced traumatic injury by activation of cGMP- or cAMP-related neuroprotective molecules, such as synaptic proteins, pCREB and BDNF.
Collapse
Affiliation(s)
- Ling Chen
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, PR China; Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Kaiping Liu
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yulu Wang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Na Liu
- Department of Traditional Medical Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, China
| | - Minjie Yao
- Department of Orthopedics, The People's Hospital of Yichun City, Yichun, Jiangxi Province, China
| | - Jinlan Hu
- Department of Anesthesiology, Shanghai Minhang TCM Hospital, Shanghai, China
| | - Gang Wang
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, PR China.
| | - Yindi Sun
- Department of Traditional Medical Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, China.
| | - Jianchun Pan
- Brain Institute, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
11
|
Yi P, Zhang Z, Huang S, Huang J, Peng W, Yang J. Integrated meta-analysis, network pharmacology, and molecular docking to investigate the efficacy and potential pharmacological mechanism of Kai-Xin-San on Alzheimer's disease. PHARMACEUTICAL BIOLOGY 2020; 58:932-943. [PMID: 32956608 PMCID: PMC7534219 DOI: 10.1080/13880209.2020.1817103] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/09/2020] [Accepted: 08/15/2020] [Indexed: 05/02/2023]
Abstract
CONTEXT Kai-Xin-San (KXS) has been used to treat Alzheimer's disease (AD) for thousands of years. However, no quantitative data regarding AD treatment using KXS are available. Moreover, its active compounds and mechanism of action for the treatment of AD remain largely unclear. OBJECTIVES To evaluate the efficacy and the potential pharmacological mechanisms of KXS in AD treatment. MATERIALS AND METHODS A systematic collection of KXS experiments was conducted from PubMed, Web of Science, Embase, CNKI, VIP, and Wanfang Data up to February, 2020. Review Manager 5 software was used for meta-analysis. In network pharmacology, components of KXS were screened, AD-related genes were then identified and the 'component-target-pathway' network constructed. Molecular docking was finally employed for in silico simulation matching between representative KXS compounds and their target genes. RESULTS Meta-analysis revealed that KXS improves the cognitive benefits in AD models by reducing the time of escape latency (SMD = -16.84) as well as increasing the number of cross-platform (SMD = 2.56) and proportion of time in the target quadrant (SMD = 7.52). Network pharmacology identified 25 KXS active compounds and 44 genes targets. DRD2, MAOA, ACHE, ADRA2A and CHRM2 were core target proteins. Besides, 22 potential pathways of KXS were identified, like cholinergic synapses, the cGMP/PKG pathway and calcium signalling. Molecular docking showed that stigmasterol, aposcopolamine and inermin can closely bind three targets (ACHE, ADRA2A and CHRM2). DISCUSSION AND CONCLUSION These findings suggest that KXS exerts effect on AD through multi-target, multi-component and multi-pathway mechanism. Future studies may explore the active components of KXS.
Collapse
Affiliation(s)
- Pengji Yi
- Department of Integrated Traditional Chinese and Western Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese and Western Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
| | - Siqi Huang
- Department of Integrated Traditional Chinese and Western Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiahua Huang
- Hunan Academy of Chinese Medicine, Changsha, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese and Western Medicine, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jingjing Yang
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
- Xiangya Nursing School, Central South University, Changsha, China
| |
Collapse
|
12
|
Sadek MS, Cachorro E, El-Armouche A, Kämmerer S. Therapeutic Implications for PDE2 and cGMP/cAMP Mediated Crosstalk in Cardiovascular Diseases. Int J Mol Sci 2020; 21:E7462. [PMID: 33050419 PMCID: PMC7590001 DOI: 10.3390/ijms21207462] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterases (PDEs) are the principal superfamily of enzymes responsible for degrading the secondary messengers 3',5'-cyclic nucleotides cAMP and cGMP. Their refined subcellular localization and substrate specificity contribute to finely regulate cAMP/cGMP gradients in various cellular microdomains. Redistribution of multiple signal compartmentalization components is often perceived under pathological conditions. Thereby PDEs have long been pursued as therapeutic targets in diverse disease conditions including neurological, metabolic, cancer and autoimmune disorders in addition to numerous cardiovascular diseases (CVDs). PDE2 is a unique member of the broad family of PDEs. In addition to its capability to hydrolyze both cAMP and cGMP, PDE2 is the sole isoform that may be allosterically activated by cGMP increasing its cAMP hydrolyzing activity. Within the cardiovascular system, PDE2 serves as an integral regulator for the crosstalk between cAMP/cGMP pathways and thereby may couple chronically adverse augmented cAMP signaling with cardioprotective cGMP signaling. This review provides a comprehensive overview of PDE2 regulatory functions in multiple cellular components within the cardiovascular system and also within various subcellular microdomains. Implications for PDE2- mediated crosstalk mechanisms in diverse cardiovascular pathologies are discussed highlighting the prospective use of PDE2 as a potential therapeutic target in cardiovascular disorders.
Collapse
Affiliation(s)
| | | | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| | - Susanne Kämmerer
- Department of Pharmacology and Toxicology, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (M.S.S.); (E.C.)
| |
Collapse
|
13
|
Li YJ, Yang LP, Hou JL, Li XM, Chen L, Zhu JH, Wang QY, Li G, Zhao PY, Liu XH, Shi ZJ. Prenatal Stress Impairs Postnatal Learning and Memory Development via Disturbance of the cGMP-PKG Pathway and Oxidative Phosphorylation in the Hippocampus of Rats. Front Mol Neurosci 2020; 13:158. [PMID: 33013315 PMCID: PMC7509422 DOI: 10.3389/fnmol.2020.00158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/03/2020] [Indexed: 12/30/2022] Open
Abstract
Clinical and animal studies have found that prenatal stress can lead to pathological changes in embryos and fetuses. However, the mechanisms through which this occurs have not been made clear. In the present study, pregnant rats were subjected to chronic psychological stress during gestational days using an improved communication box system, and the changes in behavioral performance and proteins in the hippocampus of offspring were analyzed. It was found that prenatal stress caused postnatal growth retardation and impairment in spatial learning and memory. Furthermore, in isobaric tags for relative and absolute quantitation-based proteomics analyses, 158 significantly differentially expressed proteins (DEPs) were found between the two groups. Further analyses showed that these DEPs are involved in different molecular function categories and participate in several biological processes, such as energy metabolism, learning or memory, and synaptic plasticity. Moreover, the enrichment of pathways showed that the learning and memory impairment was primarily connected with the cyclic guanosine monophosphate–protein kinase G (cGMP–PKG) pathway and oxidative phosphorylation. At the same time, the cGMP level and the expression of PKG protein were significantly decreased, and the neuronal mitochondria appeared to have a swollen and irregular shape in the hippocampus of offspring of stressed rats. These results suggest that the chronic psychological stress that pregnant rats were subjected to during gestational days may have impaired the spatial learning and memory of offspring. This affected the hippocampal oxidative phosphorylation and inhibited the cGMP–PKG pathway.
Collapse
Affiliation(s)
- Yu-Jie Li
- Pharmacology Laboratory, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Li-Ping Yang
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jun-Lin Hou
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xin-Min Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lei Chen
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiang-Hui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qi-Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Gai Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Pei-Yuan Zhao
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xi-Hong Liu
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhan-Jiang Shi
- Department of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
14
|
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: 6.6] [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
|
15
|
Argyrousi EK, Heckman PRA, Prickaerts J. Role of cyclic nucleotides and their downstream signaling cascades in memory function: Being at the right time at the right spot. Neurosci Biobehav Rev 2020; 113:12-38. [PMID: 32044374 DOI: 10.1016/j.neubiorev.2020.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
A plethora of studies indicate the important role of cAMP and cGMP cascades in neuronal plasticity and memory function. As a result, altered cyclic nucleotide signaling has been implicated in the pathophysiology of mnemonic dysfunction encountered in several diseases. In the present review we provide a wide overview of studies regarding the involvement of cyclic nucleotides, as well as their upstream and downstream molecules, in physiological and pathological mnemonic processes. Next, we discuss the regulation of the intracellular concentration of cyclic nucleotides via phosphodiesterases, the enzymes that degrade cAMP and/or cGMP, and via A-kinase-anchoring proteins that refine signal compartmentalization of cAMP signaling. We also provide an overview of the available data pointing to the existence of specific time windows in cyclic nucleotide signaling during neuroplasticity and memory formation and the significance to target these specific time phases for improving memory formation. Finally, we highlight the importance of emerging imaging tools like Förster resonance energy transfer imaging and optogenetics in detecting, measuring and manipulating the action of cyclic nucleotide signaling cascades.
Collapse
Affiliation(s)
- Elentina K Argyrousi
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Pim R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands.
| |
Collapse
|
16
|
Radiosynthesis and Biological Investigation of a Novel Fluorine-18 Labeled Benzoimidazotriazine- Based Radioligand for the Imaging of Phosphodiesterase 2A with Positron Emission Tomography. Molecules 2019; 24:molecules24224149. [PMID: 31731831 PMCID: PMC6891464 DOI: 10.3390/molecules24224149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022] Open
Abstract
A specific radioligand for the imaging of cyclic nucleotide phosphodiesterase 2A (PDE2A) via positron emission tomography (PET) would be helpful for research on the physiology and disease-related changes in the expression of this enzyme in the brain. In this report, the radiosynthesis of a novel PDE2A radioligand and the subsequent biological evaluation were described. Our prospective compound 1-(2-chloro-5-methoxy phenyl)-8-(2-fluoropyridin-4-yl)-3- methylbenzo[e]imidazo[5,1-c][1,2,4]triazine, benzoimidazotriazine (BIT1) (IC50 PDE2A = 3.33 nM; 16-fold selectivity over PDE10A) was fluorine-18 labeled via aromatic nucleophilic substitution of the corresponding nitro precursor using the K[18F]F-K2.2.2-carbonate complex system. The new radioligand [18F]BIT1 was obtained with a high radiochemical yield (54 ± 2%, n = 3), a high radiochemical purity (≥99%), and high molar activities (155–175 GBq/μmol, n = 3). In vitro autoradiography on pig brain cryosections exhibited a heterogeneous spatial distribution of [18F]BIT1 corresponding to the known pattern of expression of PDE2A. The investigation of in vivo metabolism of [18F]BIT1 in a mouse revealed sufficient metabolic stability. PET studies in mouse exhibited a moderate brain uptake of [18F]BIT1 with a maximum standardized uptake value of ~0.7 at 5 min p.i. However, in vivo blocking studies revealed a non-target specific binding of [18F]BIT1. Therefore, further structural modifications are needed to improve target selectivity.
Collapse
|
17
|
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: 7.0] [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
|
18
|
Nakashima M, Suzuki N, Shiraishi E, Iwashita H. TAK-915, a phosphodiesterase 2A inhibitor, ameliorates the cognitive impairment associated with aging in rodent models. Behav Brain Res 2019; 376:112192. [PMID: 31521738 DOI: 10.1016/j.bbr.2019.112192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 08/24/2019] [Accepted: 08/28/2019] [Indexed: 10/26/2022]
Abstract
Changes in the cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) signaling are implicated in older people with dementia. Drugs that modulate the cAMP/cGMP levels in the brain might therefore provide new therapeutic options for the treatment of cognitive impairment in aging and elderly with dementia. Phosphodiesterase 2A (PDE2A), which is highly expressed in the forebrain, is one of the key phosphodiesterase enzymes that hydrolyze cAMP and cGMP. In this study, we investigated the effects of PDE2A inhibition on the cognitive functions associated with aging, such as spatial learning, episodic memory, and attention, in rats with a selective, brain penetrant PDE2A inhibitor, 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). Repeated treatment with TAK-915 (3 mg/kg/day, p.o. for 4 days) significantly reduced escape latency in aged rats in the Morris water maze task compared to the vehicle treatment. In the novel object recognition task, TAK-915 (1, 3, and 10 mg/kg, p.o.) dose-dependently attenuated the non-selective muscarinic antagonist scopolamine-induced memory deficits in rats. In addition, oral administration of TAK-915 at 10 mg/kg significantly improved the attentional performance in middle-aged, poorly performing rats in the 5-choice serial reaction time task. These findings suggest that PDE2A inhibition in the brain has the potential to ameliorate the age-related cognitive decline.
Collapse
Affiliation(s)
- Masato Nakashima
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Japan
| | - Noriko Suzuki
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Japan
| | - Eri Shiraishi
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Japan
| | - Hiroki Iwashita
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Japan.
| |
Collapse
|
19
|
Schepers M, Tiane A, Paes D, Sanchez S, Rombaut B, Piccart E, Rutten BPF, Brône B, Hellings N, Prickaerts J, Vanmierlo T. Targeting Phosphodiesterases-Towards a Tailor-Made Approach in Multiple Sclerosis Treatment. Front Immunol 2019; 10:1727. [PMID: 31396231 PMCID: PMC6667646 DOI: 10.3389/fimmu.2019.01727] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) characterized by heterogeneous clinical symptoms including gradual muscle weakness, fatigue, and cognitive impairment. The disease course of MS can be classified into a relapsing-remitting (RR) phase defined by periods of neurological disabilities, and a progressive phase where neurological decline is persistent. Pathologically, MS is defined by a destructive immunological and neuro-degenerative interplay. Current treatments largely target the inflammatory processes and slow disease progression at best. Therefore, there is an urgent need to develop next-generation therapeutic strategies that target both neuroinflammatory and degenerative processes. It has been shown that elevating second messengers (cAMP and cGMP) is important for controlling inflammatory damage and inducing CNS repair. Phosphodiesterases (PDEs) have been studied extensively in a wide range of disorders as they breakdown these second messengers, rendering them crucial regulators. In this review, we provide an overview of the role of PDE inhibition in limiting pathological inflammation and stimulating regenerative processes in MS.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Assia Tiane
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Dean Paes
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Selien Sanchez
- Department of Morphology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Ben Rombaut
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Elisabeth Piccart
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bart P F Rutten
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bert Brône
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Niels Hellings
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Tim Vanmierlo
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
20
|
Morsy A, Trippier PC. Current and Emerging Pharmacological Targets for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2019; 72:S145-S176. [PMID: 31594236 DOI: 10.3233/jad-190744] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
No cure or disease-modifying therapy for Alzheimer's disease (AD) has yet been realized. However, a multitude of pharmacological targets have been identified for possible engagement to enable drug discovery efforts for AD. Herein, we review these targets comprised around three main therapeutic strategies. First is an approach that targets the main pathological hallmarks of AD: amyloid-β (Aβ) oligomers and hyperphosphorylated tau tangles which primarily focuses on reducing formation and aggregation, and/or inducing their clearance. Second is a strategy that modulates neurotransmitter signaling. Comprising this strategy are the cholinesterase inhibitors and N-methyl-D-aspartate receptor blockade treatments that are clinically approved for the symptomatic treatment of AD. Additional targets that aim to stabilize neuron signaling through modulation of neurotransmitters and their receptors are also discussed. Finally, the third approach comprises a collection of 'sensitive targets' that indirectly influence Aβ or tau accumulation. These targets are proteins that upon Aβ accumulation in the brain or direct Aβ-target interaction, a modification in the target's function is induced. The process occurs early in disease progression, ultimately causing neuronal dysfunction. This strategy aims to restore normal target function to alleviate Aβ-induced toxicity in neurons. Overall, we generally limit our analysis to targets that have emerged in the last decade and targets that have been validated using small molecules in in vitro and/or in vivo models. This review is not an exhaustive list of all possible targets for AD but serves to highlight the most promising and critical targets suitable for small molecule drug intervention.
Collapse
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
21
|
Lee CC, Chang CP, Lin CJ, Lai HL, Kao YH, Cheng SJ, Chen HM, Liao YP, Faivre E, Buée L, Blum D, Fang JM, Chern Y. Adenosine Augmentation Evoked by an ENT1 Inhibitor Improves Memory Impairment and Neuronal Plasticity in the APP/PS1 Mouse Model of Alzheimer's Disease. Mol Neurobiol 2018; 55:8936-8952. [PMID: 29616397 DOI: 10.1007/s12035-018-1030-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment and synaptic dysfunction. Adenosine is an important homeostatic modulator that controls the bioenergetic network in the brain through regulating receptor-evoked signaling pathways, bioenergetic machineries, and epigenetic-mediated gene regulation. Equilibrative nucleoside transporter 1 (ENT1) is a major adenosine transporter that recycles adenosine from the extracellular space. In the present study, we report that a small adenosine analogue (designated J4) that inhibited ENT1 prevented the decline in spatial memory in an AD mouse model (APP/PS1). Electrophysiological and biochemical analyses further demonstrated that chronic treatment with J4 normalized the impaired basal synaptic transmission and long-term potentiation (LTP) at Schaffer collateral synapses as well as the aberrant expression of synaptic proteins (e.g., NR2A and NR2B), abnormal neuronal plasticity-related signaling pathways (e.g., PKA and GSK3β), and detrimental elevation in astrocytic A2AR expression in the hippocampus and cortex of APP/PS1 mice. In conclusion, our findings suggest that modulation of adenosine homeostasis by J4 is beneficial in a mouse model of AD. Our study provides a potential therapeutic strategy to delay the progression of AD.
Collapse
Affiliation(s)
- Chia-Chia Lee
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Chun-Jung Lin
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Hsing-Lin Lai
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Yu-Han Kao
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Sin-Jhong Cheng
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan.,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Yu-Ping Liao
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan
| | - Emilie Faivre
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - Luc Buée
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - David Blum
- Université de Lille, Inserm, CHU-Lille, LabEx DISTALZ, Jean-Pierre Aubert research centre UMR-S1172, Lille, France
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei, 115, Taiwan.
| |
Collapse
|
22
|
Wu Y, Li Z, Huang YY, Wu D, Luo HB. Novel Phosphodiesterase Inhibitors for Cognitive Improvement in Alzheimer's Disease. J Med Chem 2018; 61:5467-5483. [PMID: 29363967 DOI: 10.1021/acs.jmedchem.7b01370] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is one of the greatest public health challenges. Phosphodiesterases (PDEs) are a superenzyme family responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Since several PDE subfamilies are highly expressed in the human brain, the inhibition of PDEs is involved in neurodegenerative processes by regulating the concentration of cAMP and/or cGMP. Currently, PDEs are considered as promising targets for the treatment of AD since many PDE inhibitors have exhibited remarkable cognitive improvement effects in preclinical studies and over 15 of them have been subjected to clinical trials. The aim of this review is to summarize the outstanding progress that has been made by PDE inhibitors as anti-AD agents with encouraging results in preclinical studies and clinical trials. The binding affinity, pharmacokinetics, underlying mechanisms, and limitations of these PDE inhibitors in the treatment of AD are also reviewed and discussed.
Collapse
Affiliation(s)
- Yinuo Wu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Zhe Li
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Yi-You Huang
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Deyan Wu
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , P. R. China
| |
Collapse
|
23
|
He B, Xu F, Xiao F, Yan T, Wu B, Bi K, Jia Y. Neuroprotective effects of nootkatone from Alpiniae oxyphyllae Fructus against amyloid-β-induced cognitive impairment. Metab Brain Dis 2018; 33:251-259. [PMID: 29177693 DOI: 10.1007/s11011-017-0154-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/14/2017] [Indexed: 11/30/2022]
Abstract
The sesquiterpene nootkatone (NKT), isolated from Alpiniae oxyphyllae Fructus, was shown to possess protective effects on neurons. In our study, by using an Alzheimer's disease (AD) model of mice induced by intracerebroventricular (i.c.v.) injection of Aβ1-42 oligomers, we investigated the effects of NKT on memory impairment and further evaluated the pathological changes of mice. AD mice were treated by i.c.v. injection of NKT (at a dose of 0.02 mg/kg and 0.20 mg/kg) or vehicle (PBS) into the lateral ventricle once daily for 5 consecutive days. The behavioral tasks were performed, and levels of some biochemical indicators and histopathological changes of the brain were evaluated to elucidate the mechanism of NKT in the treatment of AD. The results revealed that NKT significantly improved the neurobehavioral performance of the AD mice in the Y-maze and Morris water maze tests. More importantly, NKT treatment decreased the malondialdehyde (MDA), Aβ as well as the acetylcholin esterase (AChE) levels in the mice brain, while increased the glutathione peroxidase (GSH-Px) levels with improved histopathological changes in the hippocampus. These findings provided evidences for the beneficial role of NKT in Aβ1-42-induced mice AD model linking to anti-oxidative and anti-AChE activities with inhibitory effect against Aβ accumulation.
Collapse
Affiliation(s)
- Bosai He
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, People's Republic of China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
- Jiangsu Kangyuan Pharmaceutical Co. Ltd, Lianyungang, People's Republic of China
| | - Feng Xiao
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, People's Republic of China
| | - Tingxu Yan
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, People's Republic of China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
24
|
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: 2.7] [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
|
25
|
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: 13.6] [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
|
26
|
Physiological and pathological processes of synaptic plasticity and memory in drug discovery: Do not forget the dose-response curve. Eur J Pharmacol 2017; 817:59-70. [DOI: 10.1016/j.ejphar.2017.05.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/20/2017] [Accepted: 05/30/2017] [Indexed: 01/24/2023]
|
27
|
Inhibition of PDE2 reverses beta amyloid induced memory impairment through regulation of PKA/PKG-dependent neuro-inflammatory and apoptotic pathways. Sci Rep 2017; 7:12044. [PMID: 28935920 PMCID: PMC5608906 DOI: 10.1038/s41598-017-08070-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/29/2017] [Indexed: 11/08/2022] Open
Abstract
Beta amyloid peptides (Aβ) are known risk factors involved in cognitive impairment, neuroinflammatory and apoptotic processes in Alzheimer’s disease (AD). Phosphodiesterase 2 (PDE2) inhibitors increase the intracellular cAMP and/or cGMP activities, which may ameliorate cognitive deficits associated with AD. However, it remains unclear whether PDE2 mediated neuroapoptotic and neuroinflammatory events, as well as cognitive performance in AD are related to cAMP/cGMP-dependent pathways. The present study investigated how the selective PDE2 inhibitor BAY60-7550 (BAY) affected Aβ-induced learning and memory impairment in two classic rodent models. IL-22 and IL-17, Bax and Bcl-2, PKA/PKG and the brain derived neurotropic factor (BDNF) levels in hippocampus and cortex were detected with immunoblotting assay. The results showed that BAY reversed Aβ-induced cognitive impairment as shown in the water maze test and step-down test. Moreover, BAY treatment reversed the Aβ-induced changes in IL-22 and IL-17 and the ratio of Bax/Bcl-2. Changes in cAMP/cGMP levels, PKA/PKG and BDNF expression were also prevented by BAY. These effects of BAY on memory performance and related neurochemical changes were partially blocked by the PKG inhibitor KT 5823. These findings indicated that the protective effects of BAY against Aβ-induced memory deficits might involve the regulation of neuroinflammation and neuronal apoptotic events.
Collapse
|
28
|
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: 123] [Impact Index Per Article: 15.4] [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
|
29
|
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: 6.4] [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
|
30
|
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: 3.1] [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
|
31
|
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: 2.3] [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
|
32
|
Snyder GL, Vanover KE. PDE Inhibitors for the Treatment of Schizophrenia. ADVANCES IN NEUROBIOLOGY 2017; 17:385-409. [DOI: 10.1007/978-3-319-58811-7_14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
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: 2.4] [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
|
34
|
Wang JH, Lei X, Cheng XR, Zhang XR, Liu G, Cheng JP, Xu YR, Zeng J, Zhou WX, Zhang YX. LW-AFC, a new formula derived from Liuwei Dihuang decoction, ameliorates behavioral and pathological deterioration via modulating the neuroendocrine-immune system in PrP-hAβPPswe/PS1 ΔE9 transgenic mice. Alzheimers Res Ther 2016; 8:57. [PMID: 27964740 PMCID: PMC5154149 DOI: 10.1186/s13195-016-0226-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 11/24/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Accumulating evidence implicates the neuroendocrine immunomodulation (NIM) network in the physiopathological mechanism of Alzheimer's disease (AD). Notably, we previously revealed that the NIM network is dysregulated in the PrP-hAβPPswe/PS1ΔE9 (APP/PS1) transgenic mouse model of AD. METHODS After treatment with a novel Liuwei Dihuang formula (LW-AFC), mice were cognitively evaluated in behavioral experiments. Neuron loss, amyloid-β (Aβ) deposition, and Aβ level were analyzed using Nissl staining, immunofluorescence, and an AlphaLISA assay, respectively. Multiplex bead analysis, a radioimmunoassay, immunochemiluminometry, and an enzyme-linked immunosorbent assay (ELISA) were used to measure cytokine and hormone levels. Lymphocyte subsets were detected using flow cytometry. Data between two groups were compared using a Student's t test. Comparison of the data from multiple groups against one group was performed using a one-way analysis of variance (ANOVA) followed by a Dunnett's post hoc test or a two-way repeated-measures analysis of variance with a Tukey multiple comparisons test. RESULTS LW-AFC ameliorated the cognitive impairment observed in APP/PS1 mice, including the impairment of object recognition memory, spatial learning and memory, and active and passive avoidance. In addition, LW-AFC alleviated the neuron loss in the hippocampus, suppressed Aβ deposition in the brain, and reduced the concentration of Aβ1-42 in the hippocampus and plasma of APP/PS1 mice. LW-AFC treatment also significantly decreased the secretion of corticotropin-releasing hormone and gonadotropin-releasing hormone in the hypothalamus, and adrenocorticotropic hormone, luteinizing hormone, and follicle-stimulating hormone in the pituitary. Moreover, LW-AFC increased CD8+CD28+ T cells, and reduced CD4+CD25+Foxp3+ T cells in the spleen lymphocytes, downregulated interleukin (IL)-1β, IL-2, IL-6, IL-23, granulocyte-macrophage colony stimulating factor, and tumor necrosis factor-α and -β, and upregulated IL-4 and granulocyte colony stimulating factor in the plasma of APP/PS1 mice. CONCLUSIONS LW-AFC ameliorated the behavioral and pathological deterioration of APP/PS1 transgenic mice via the restoration of the NIM network to a greater extent than either memantine or donepezil, which supports the use of LW-AFC as a potential agent for AD therapy.
Collapse
Affiliation(s)
- Jian-Hui Wang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Xi Lei
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
- Guangxi Medical University, Nanning, 530021, China
| | - Xiao-Rui Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| | - Xiao-Rui Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Gang Liu
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Jun-Ping Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Yi-Ran Xu
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Ju Zeng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Wen-Xia Zhou
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| | - Yong-Xiang Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| |
Collapse
|
35
|
Soares LM, Meyer E, Milani H, Steinbusch HWM, Prickaerts J, de Oliveira RMW. The phosphodiesterase type 2 inhibitor BAY 60-7550 reverses functional impairments induced by brain ischemia by decreasing hippocampal neurodegeneration and enhancing hippocampal neuronal plasticity. Eur J Neurosci 2016; 45:510-520. [PMID: 27813297 DOI: 10.1111/ejn.13461] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/26/2016] [Accepted: 10/31/2016] [Indexed: 01/08/2023]
Abstract
Cognitive and affective impairments are the most characterized consequences following cerebral ischemia. BAY 60-7550, a selective phosphodiesterase type 2 inhibitor (PDE2-I), presents memory-enhancing and anxiolytic-like properties. The behavioral effects of BAY 60-7550 have been associated with its ability to prevent hydrolysis of both cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) thereby interfering with neuronal plasticity. Here, we hypothesize that PDE2-I treatment could promote functional recovery after brain ischemia. Mice C57Bl/6 were submitted to bilateral common carotid artery occlusion (BCCAO), an experimental model of transient brain ischemia, for 20 min. During 21 days after reperfusion, the animals were tested in a battery of behavioral tests including the elevated zero maze (EZM), object location task (OLT) and forced swim test (FST). The effects of BAY 60-7550 were evaluated on neuronal nuclei (NeuN), caspase-9, cAMP response element-binding protein (CREB), phosphorylated CREB (pCREB) and brain-derived neurotrophic factor (BDNF) expression in the hippocampus. BCCAO increased anxiety levels, impaired hippocampus-dependent cognitive function and induced despair-like behavior in mice. Hippocampal neurodegeneration was evidenced by a decrease in NeuN and increase incaspase-9 protein levels in BCCAO mice. Ischemic mice also showed low BDNF protein levels in the hippocampus. Repeated treatment with BAY 60-7550 attenuated the behavioral impairments induced by BCCAO in mice. Concomitantly, BAY 60-7550 enhanced expression of pCREB and BDNF protein levels in the hippocampus of ischemic mice. The present findings suggest that chronic inhibition of PDE2 provides functional recovery in BCCAO mice possibly by augmenting hippocampal neuronal plasticity.
Collapse
Affiliation(s)
- Ligia Mendes Soares
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, CEP 87020-900, Maringá, Paraná, Brazil
| | - Erika Meyer
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, CEP 87020-900, Maringá, Paraná, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, CEP 87020-900, Maringá, Paraná, Brazil
| | - Harry W M Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Rúbia M Weffort de Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790, CEP 87020-900, Maringá, Paraná, Brazil
| |
Collapse
|
36
|
Fontana R, Agostini M, Murana E, Mahmud M, Scremin E, Rubega M, Sparacino G, Vassanelli S, Fasolato C. Early hippocampal hyperexcitability in PS2APP mice: role of mutant PS2 and APP. Neurobiol Aging 2016; 50:64-76. [PMID: 27889678 DOI: 10.1016/j.neurobiolaging.2016.10.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 10/17/2016] [Accepted: 10/28/2016] [Indexed: 12/27/2022]
Abstract
Alterations of brain network activity are observable in Alzheimer's disease (AD) together with the occurrence of mild cognitive impairment, before overt pathology. However, in humans as well in AD mouse models, identification of early biomarkers of network dysfunction is still at its beginning. We performed in vivo recordings of local field potential activity in the dentate gyrus of PS2APP mice expressing the human amyloid precursor protein (APP) Swedish mutation and the presenilin-2 (PS2) N141I. From a frequency-domain analysis, we uncovered network hyper-synchronicity as early as 3 months, when intracellular accumulation of amyloid beta was also observable. In addition, at 6 months of age, we identified network hyperactivity in the beta/gamma frequency bands, along with increased theta-beta and theta-gamma phase-amplitude cross-frequency coupling, in coincidence with the histopathological traits of the disease. Although hyperactivity and hypersynchronicity were respectively detected in mice expressing the PS2-N141I or the APP Swedish mutant alone, the increase in cross-frequency coupling specifically characterized the 6-month-old PS2APP mice, just before the surge of the cognitive decline.
Collapse
Affiliation(s)
- Roberto Fontana
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Mario Agostini
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Emanuele Murana
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Mufti Mahmud
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Elena Scremin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Maria Rubega
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Giovanni Sparacino
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
37
|
Translational Assays for Assessment of Cognition in Rodent Models of Alzheimer’s Disease and Dementia. J Mol Neurosci 2016; 60:371-382. [DOI: 10.1007/s12031-016-0837-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023]
|
38
|
Effects of neuregulin-1 administration on neurogenesis in the adult mouse hippocampus, and characterization of immature neurons along the septotemporal axis. Sci Rep 2016; 6:30467. [PMID: 27469430 PMCID: PMC4965755 DOI: 10.1038/srep30467] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
Adult hippocampal neurogenesis is associated with learning and affective behavioural regulation. Its diverse functionality is segregated along the septotemporal axis from the dorsal to ventral hippocampus. However, features distinguishing immature neurons in these regions have yet to be characterized. Additionally, although we have shown that administration of the neurotrophic factor neuregulin-1 (NRG1) selectively increases proliferation and overall neurogenesis in the mouse ventral dentate gyrus (DG), likely through ErbB3, NRG1's effects on intermediate neurogenic stages in immature neurons are unknown. We examined whether NRG1 administration increases DG ErbB3 phosphorylation. We labeled adultborn cells using BrdU, then administered NRG1 to examine in vivo neurogenic effects on immature neurons with respect to cell survival, morphology, and synaptogenesis. We also characterized features of immature neurons along the septotemporal axis. We found that neurogenic effects of NRG1 are temporally and subregionally specific to proliferation in the ventral DG. Particular morphological features differentiate immature neurons in the dorsal and ventral DG, and cytogenesis differed between these regions. Finally, we identified synaptic heterogeneity surrounding the granule cell layer. These results indicate neurogenic involvement of NRG1-induced antidepressant-like behaviour is particularly associated with increased ventral DG cell proliferation, and identify novel distinctions between dorsal and ventral hippocampal neurogenic development.
Collapse
|
39
|
Tang W, Cheng J, Wang ZY, Chen KY, Han ZM, Wang QH, Yao YY. The Synergistic Roles of the Chronic Prenatal and Offspring Stress Exposures in Impairing Offspring Learning and Memory. J Alzheimers Dis 2016; 53:221-36. [PMID: 27128656 DOI: 10.3233/jad-160011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In Alzheimer's disease (AD), extensive experimental studies have demonstrated a negative impact of chronic stress during various stages of life (including prenatal phase) on some aspects of AD pathology. Nevertheless, presently, few studies have been involved in the learning and memory impairments, as well as neuropathology elicited by the chronic prenatal stress (CPS) and the chronic offspring stress (COS) exposures simultaneously, particularly for the adult male APPswe/PS1dE9 murine offspring. Therefore, the aim of the present study was to investigate the influence of CPS on learning and memory impairments induced by COS in 6-month-old male APPswe/PS1dE9 offspring mice and the related mechanism. Our study firstly demonstrates that 14-day exposure to CPS could exacerbate the learning and memory impairments, as well as neuropathological damages in the CA3 regions of the hippocampus and cortex neurons, which is induced by the 28-day exposure to COS in 6-month-old male APPswe/PS1dE9 offspring mice. In addition, CPS could potentiate the production of AβPP, Aβ42, and corticosterone in 6-month-old male APPswe/PS1dE9 offspring that also suffer COS. In conclusion, our novel findings strongly implicate the synergistic roles of the CPS and COS exposures in impairing offspring learning and memory. Moreover, CPS potentiating the production of Aβ42 might be mediated by glucocorticoids through increasing the expression of APP and BACE1 gene.
Collapse
Affiliation(s)
- Wei Tang
- Department of Laboratory Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Department of Clinical Laboratory Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Juan Cheng
- Department of Laboratory Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zheng-Yu Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Ke-Yang Chen
- Department of Clinical Laboratory Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Zhen-Min Han
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Qi-Hong Wang
- Hefei Meikang Medical Equipment Co., Ltd., Hefei, Anhui, China
| | - Yu-You Yao
- Department of Clinical Laboratory Medicine, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
40
|
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
|
41
|
Fernández-Fernández D, Rosenbrock H, Kroker KS. Inhibition of PDE2A, but not PDE9A, modulates presynaptic short-term plasticity measured by paired-pulse facilitation in the CA1 region of the hippocampus. Synapse 2015; 69:484-96. [DOI: 10.1002/syn.21840] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/28/2015] [Accepted: 05/28/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Diego Fernández-Fernández
- Department of CNS Diseases Research; Boehringer Ingelheim Pharma GmbH & Co KG; Biberach (Riss) 88397 Germany
| | - Holger Rosenbrock
- Department of CNS Diseases Research; Boehringer Ingelheim Pharma GmbH & Co KG; Biberach (Riss) 88397 Germany
| | - Katja S. Kroker
- Department of Drug Discovery Support; Boehringer Ingelheim Pharma GmbH & Co KG; Biberach (Riss) 88397 Germany
| |
Collapse
|
42
|
Yang G, Song Y, Zhou X, Deng Y, Liu T, Weng G, Yu D, Pan S. MicroRNA-29c targets β-site amyloid precursor protein-cleaving enzyme 1 and has a neuroprotective role in vitro and in vivo. Mol Med Rep 2015; 12:3081-8. [PMID: 25955795 DOI: 10.3892/mmr.2015.3728] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/24/2015] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD), characterized by β-amyloid deposition and neurodegeneration, is the most common cause of dementia worldwide. Emerging evidence suggests that ectopic expression of micro (mi)RNAs is involved in the pathogenesis of AD. There is increasing evidence that miRNAs expressed in the brain are involved in neuronal development, survival and apoptosis. The expression of β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is regulated by dysregulated miRNAs in the brain. The present study determined the expression levels of the miRNA-29 (miR-29) family in peripheral blood samples of patients with AD and demonstrated a marked decrease in the expression of miR-29c compared with age-matched controls. In addition, a significant increase in the expression of BACE1 was observed in the peripheral blood of patients with AD. Correlation analysis revealed that the expression of miR-29c was negatively correlated with the protein expression of BACE1 in the peripheral blood samples from patients with AD. The present study also investigated the role of miR-29 on hippocampal neurons in vitro and in vivo. The results demonstrated that the upregulation of miR-29c promoted learning and memory behaviors in SAMP8 mice, at least partially, by increasing the activity of protein kinase A/cAMP response element-binding protein, involved in neuroprotection. This evidence suggested that miR-29c may be a promising potential therapeutic target against AD.
Collapse
Affiliation(s)
- Guoshuai Yang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yanmin Song
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaoyan Zhou
- Department of Neurology, Haikou People's Hospital, Haikou, Hainan 570208, P.R. China
| | - Yidong Deng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tao Liu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guohu Weng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dan Yu
- Department of Neurology, Haikou People's Hospital, Haikou, Hainan 570208, P.R. China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
43
|
Bollen E, Akkerman S, Puzzo D, Gulisano W, Palmeri A, D'Hooge R, Balschun D, Steinbusch HWM, Blokland A, Prickaerts J. Object memory enhancement by combining sub-efficacious doses of specific phosphodiesterase inhibitors. Neuropharmacology 2015; 95:361-6. [PMID: 25896769 DOI: 10.1016/j.neuropharm.2015.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/13/2015] [Accepted: 04/03/2015] [Indexed: 01/17/2023]
Abstract
The second messengers cGMP and cAMP have a vital role in synaptic plasticity and memory processes. As such, phosphodiesterases inhibitors (PDE-Is), which prevent the breakdown of these cyclic nucleotides, represent a potential treatment strategy in memory decline. Recently it has been demonstrated that cGMP and cAMP signaling act in sequence during memory consolidation, with early cGMP signaling requiring subsequent cAMP signaling. Here, we sought to confirm this relationship, and to evaluate its therapeutic implications. Combining sub-efficacious doses of the cGMP-specific PDE type 5 inhibitor vardenafil (0.1 mg/kg) and cAMP-specific PDE type 4 inhibitor rolipram (0.01 mg/kg) during the early and late memory consolidation phase, respectively, led to improved memory performance in a 24 h interval object recognition task. Similarly, such a sub-efficacious combination treatment enhanced the transition of early-phase long-term potentiation (LTP) to late-phase LTP in hippocampal slices. In addition, both object memory and LTP were improved after administration of two sub-efficacious doses of the dual substrate PDE type 2 inhibitor BAY60 7550 (0.3 mg/kg) at the early and late consolidation phase, respectively. Taken together, combinations of sub-efficacious doses of cAMP- and cGMP-specific PDE-Is have an additive effect on long-term synaptic plasticity and memory formation and might prove a superior alternative to single PDE-I treatment.
Collapse
Affiliation(s)
- E Bollen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - S Akkerman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - D Puzzo
- Department of Biomedical and Biotechnological Sciences - Section of Physiology, University of Catania, 95125 Catania, Italy
| | - W Gulisano
- Department of Biomedical and Biotechnological Sciences - Section of Physiology, University of Catania, 95125 Catania, Italy
| | - A Palmeri
- Department of Biomedical and Biotechnological Sciences - Section of Physiology, University of Catania, 95125 Catania, Italy
| | - R D'Hooge
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, University of Leuven, 3000 Leuven, Belgium
| | - D Balschun
- Laboratory of Biological Psychology, Faculty of Psychology and Educational Sciences, University of Leuven, 3000 Leuven, Belgium
| | - H W M Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - A Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - J Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
44
|
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: 1.3] [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
|
45
|
Kametani F, Haga S. Accumulation of carboxy-terminal fragments of APP increases phosphodiesterase 8B. Neurobiol Aging 2014; 36:634-7. [PMID: 25457556 DOI: 10.1016/j.neurobiolaging.2014.09.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/19/2014] [Accepted: 09/30/2014] [Indexed: 11/17/2022]
Abstract
The long-standing "amyloid hypothesis" that Alzheimer's disease is caused by the production and aggregation of amyloid-β faces serious challenges by data recently obtained from neuroimaging studies and amyloid-β amyloid-focused clinical trials. Meanwhile, accumulation of carboxy-terminal fragments (CTFs) of the amyloid precursor protein (APP) may be neurotoxic and may impair synaptic plasticity and long-term memory in Alzheimer's disease, as suggested in murine models. To clarify these issues, we carried out a proteomic analysis of Chinese hamster ovary cells expressing APP CTFs and found that APP-CTF accumulation induced an increase in the level of phosphodiesterase 8B, suggesting that the hydrolysis of cyclic AMP was enhanced.
Collapse
Affiliation(s)
- Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Seiichi Haga
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
46
|
Phosphodiesterase 2A localized in the spinal cord contributes to inflammatory pain processing. Anesthesiology 2014; 121:372-82. [PMID: 24758774 DOI: 10.1097/aln.0000000000000270] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Phosphodiesterase 2A (PDE2A) is an evolutionarily conserved enzyme that catalyzes the degradation of the cyclic nucleotides, cyclic adenosine monophosphate, and/or cyclic guanosine monophosphate. Recent studies reported the expression of PDE2A in the dorsal horn of the spinal cord, pointing to a potential contribution to the processing of pain. However, the functions of PDE2A in spinal pain processing in vivo remained elusive. METHODS Immunohistochemistry, laser microdissection, and quantitative real-time reverse transcription polymerase chain reaction experiments were performed to characterize the localization and regulation of PDE2A protein and messenger RNA in the mouse spinal cord. Effects of the selective PDE2A inhibitor, BAY 60-7550 (Cayman Chemical, Ann Arbor, MI), in animal models of inflammatory pain (n = 6 to 10), neuropathic pain (n = 5 to 6), and after intrathecal injection of cyclic nucleotides (n = 6 to 8) were examined. Also, cyclic adenosine monophosphate and cyclic guanosine monophosphate levels in spinal cord tissues were measured by liquid chromatography tandem mass spectrometry. RESULTS The authors here demonstrate that PDE2A is distinctly expressed in neurons of the superficial dorsal horn of the spinal cord, and that its spinal expression is upregulated in response to hind paw inflammation. Administration of the selective PDE2A inhibitor, BAY 60-7550, increased the nociceptive behavior of mice in animal models of inflammatory pain. Moreover, BAY 60-7550 increased the pain hypersensitivity induced by intrathecal delivery of cyclic adenosine monophosphate, but not of cyclic guanosine monophosphate, and it increased the cyclic adenosine monophosphate levels in spinal cord tissues. CONCLUSION Our findings indicate that PDE2A contributes to the processing of inflammatory pain in the spinal cord.
Collapse
|
47
|
Xu Y, Pan J, Sun J, Ding L, Ruan L, Reed M, Yu X, Klabnik J, Lin D, Li J, Chen L, Zhang C, Zhang H, O'Donnell JM. Inhibition of phosphodiesterase 2 reverses impaired cognition and neuronal remodeling caused by chronic stress. Neurobiol Aging 2014; 36:955-70. [PMID: 25442113 DOI: 10.1016/j.neurobiolaging.2014.08.028] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 12/22/2022]
Abstract
Chronic stress and neuronal vulnerability have recently been recognized as factors contributing to cognitive disorders. One way to modify neuronal vulnerability is through mediation of phosphodiesterase 2 (PDE2), an enzyme that exerts its action on cognitive processes via the control of intracellular second messengers, cGMP and, to a lesser extent, cAMP. This study explored the effects of a PDE2 inhibitor, Bay 60-7550, on stress-induced learning and memory dysfunction in terms of its ramification on behavioral, morphologic, and molecular changes. Bay 60-7550 reversed stress-induced cognitive impairment in the Morris water maze, novel object recognition, and location tasks (object recognition test and/or object location test), effects prevented by treatment with 7-NI, a selective inhibitor of neuronal nitric oxide synthase; MK801, a glutamate receptor (NMDAR) inhibitor; myr-AIP, a CaMKII inhibitor; and KT5823, a protein kinase G inhibitor. Bay 60-7550 also ameliorated stress-induced structural remodeling in the CA1 of the hippocampus, leading to increases in dendritic branching, length, and spine density. However, the neuroplasticity initiated by Bay 60-7550 was not seen in the presence of 7-NI, MK801, myr-AIP, or KT5823. PDE2 inhibition reduced stress-induced extracellular-regulated protein kinase activation and attenuated stress-induced decreases in transcription factors (e.g., Elk-1, TORC1, and CREB phosphorylation) and plasticity-related proteins (e.g., Egr-1 and brain-derived neurotrophic factor). Pretreatment with inhibitors of NMDA, CaMKII, neuronal nitric oxide synthase, and protein kinase G (or protein kinase A) blocked the effects of Bay 60-7550 on cGMP or cAMP signaling. These findings indicate that the effect of PDE2 inhibition on stress-induced memory impairment is potentially mediated via modulation of neuroplasticity-related NMDAR-CaMKII-cGMP/cAMP signaling.
Collapse
Affiliation(s)
- Ying Xu
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Jianchun Pan
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jiao Sun
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Lina Ruan
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Miranda Reed
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Xuefeng Yu
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jonathan Klabnik
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - Dan Lin
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Jianxin Li
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Ling Chen
- Brain Institute, Department of Pharmacy, School of Pharmacy, Wenzhou Medical College, Wenzhou, China
| | - Chong Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Hanting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University, Morgantown, WV, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
48
|
am Ende CW, Kormos BL, Humphrey JM. The State of the Art in Selective PDE2A Inhibitor Design. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/9783527682348.ch06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
49
|
Gupta S, Sharma B. Protective effects of phosphodiesterase-1 (PDE1) and ATP sensitive potassium (KATP) channel modulators against 3-nitropropionic acid induced behavioral and biochemical toxicities in experimental Huntington׳s disease. Eur J Pharmacol 2014; 732:111-22. [DOI: 10.1016/j.ejphar.2014.03.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/15/2014] [Accepted: 03/24/2014] [Indexed: 12/16/2022]
|
50
|
Persistence of behaviours in the Forced Swim Test in 3xTg-AD mice at advanced stages of disease. Behav Processes 2014; 106:118-21. [PMID: 24857980 DOI: 10.1016/j.beproc.2014.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 03/04/2014] [Accepted: 05/13/2014] [Indexed: 01/19/2023]
Abstract
Forced Swimming Test (FST) models behavioural despair in animals by loss of motivation to respond or the refusal to escape. The present study characterizes the behavioural responses of 12-month-old male 3xTg-AD mice in FST as compared to age-matched no-transgenic (NTg) mice. Paradoxical results were consistently found from what would be expected from their BPSD (Behavioural and Psychological Symptoms of Dementia)-like profile. The comprehensive analysis of the ethogram shown in the FST considered the intervals of the test (0-2 and 2-6min), all the elicited behavioural responses (immobility, swimming and climbing) and their features (total duration, frequency of episodes and mean duration). Both genotypes showed equal number of swimming episodes and climbing attempts during the first interval, that resulted in high swimming times, short climbing and scarce immobility. Thereafter, the NTg mice showed a behavioural shift over time and the immobility response showed up. In contrast, all the measures consistently evidenced that 3xTg-AD persisted with the previous behavioural pattern. Genotype differences consisted in less number of episodes of immobility and swimming, and a low immobility time in favour of swimming. No differences were found in 'climbing' attempts. The behavioural response observed is discussed as a lack of ability of 3xTg-AD mice to shift behaviour over time that may result of poorest cognitive flexibility and copying with stress strategies more than behavioural despair per se.
Collapse
|