51
|
Gulisano W, Melone M, Ripoli C, Tropea MR, Li Puma DD, Giunta S, Cocco S, Marcotulli D, Origlia N, Palmeri A, Arancio O, Conti F, Grassi C, Puzzo D. Neuromodulatory Action of Picomolar Extracellular Aβ42 Oligomers on Presynaptic and Postsynaptic Mechanisms Underlying Synaptic Function and Memory. J Neurosci 2019; 39:5986-6000. [PMID: 31127002 PMCID: PMC6650983 DOI: 10.1523/jneurosci.0163-19.2019] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/09/2019] [Accepted: 04/28/2019] [Indexed: 01/01/2023] Open
Abstract
Failure of anti-amyloid-β peptide (Aβ) therapies against Alzheimer's disease (AD), a neurodegenerative disorder characterized by high amounts of the peptide in the brain, raised the question of the physiological role of Aβ released at low concentrations in the healthy brain. To address this question, we studied the presynaptic and postsynaptic mechanisms underlying the neuromodulatory action of picomolar amounts of oligomeric Aβ42 (oAβ42) on synaptic glutamatergic function in male and female mice. We found that 200 pm oAβ42 induces an increase of frequency of miniature EPSCs and a decrease of paired pulse facilitation, associated with an increase in docked vesicle number, indicating that it augments neurotransmitter release at presynaptic level. oAβ42 also produced postsynaptic changes as shown by an increased length of postsynaptic density, accompanied by an increased expression of plasticity-related proteins such as cAMP-responsive element binding protein phosphorylated at Ser133, calcium-calmodulin-dependent kinase II phosphorylated at Thr286, and brain-derived neurotrophic factor, suggesting a role for Aβ in synaptic tagging. These changes resulted in the conversion of early into late long-term potentiation through the nitric oxide/cGMP/protein kinase G intracellular cascade consistent with a cGMP-dependent switch from short- to long-term memory observed in vivo after intrahippocampal administration of picomolar amounts of oAβ42 These effects were present upon extracellular but not intracellular application of the peptide and involved α7 nicotinic acetylcholine receptors. These observations clarified the physiological role of oAβ42 in synaptic function and memory formation providing solid fundamentals for investigating the pathological effects of high Aβ levels in the AD brains.SIGNIFICANCE STATEMENT High levels of oligomeric amyloid-β42 (oAβ42) induce synaptic dysfunction leading to memory impairment in Alzheimer's disease (AD). However, at picomolar concentrations, the peptide is needed to ensure long-term potentiation (LTP) and memory. Here, we show that extracellular 200 pm oAβ42 concentrations increase neurotransmitter release, number of docked vesicles, postsynaptic density length, and expression of plasticity-related proteins leading to the conversion of early LTP into late LTP and of short-term memory into long-term memory. These effects require α7 nicotinic acetylcholine receptors and are mediated through the nitric oxide/cGMP/protein kinase G pathway. The knowledge of Aβ function in the healthy brain might be useful to understand the causes leading to its increase and detrimental effect in AD.
Collapse
Affiliation(s)
- Walter Gulisano
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
- Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), Ancona 60020, Italy
| | - Cristian Ripoli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Maria Rosaria Tropea
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Salvatore Giunta
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Daniele Marcotulli
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
| | - Nicola Origlia
- Neuroscience Institute, Italian National Research Council, Pisa 56100, Italy
| | - Agostino Palmeri
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York 10032
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona 60020, Italy
- Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), Ancona 60020, Italy
- Foundation for Molecular Medicine, Università Politecnica delle Marche, Ancona 60020, Italy, and
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
| | - Daniela Puzzo
- Department Biomedical and Biotechnological Sciences, University of Catania, Catania 95123, Italy,
- Oasi Research Institute-IRCCS, Troina, 94018, Italy
| |
Collapse
|
52
|
van Goethem NP, Paes D, Puzzo D, Fedele E, Rebosio C, Gulisano W, Palmeri A, Wennogle LP, Peng Y, Bertrand D, Prickaerts J. Antagonizing α7 nicotinic receptors with methyllycaconitine (MLA) potentiates receptor activity and memory acquisition. Cell Signal 2019; 62:109338. [PMID: 31176021 DOI: 10.1016/j.cellsig.2019.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
α7 nicotinic acetylcholine receptors (α7nAChRs) have been targeted to improve cognition in different neurological and psychiatric disorders. Nevertheless, no α7nAChR activating ligand has been clinically approved. Here, we investigated the effects of antagonizing α7nAChRs using the selective antagonist methyllycaconitine (MLA) on receptor activity in vitro and cognitive functioning in vivo. Picomolar concentrations of MLA significantly potentiated receptor responses in electrophysiological experiments mimicking the in vivo situation. Furthermore, microdialysis studies showed that MLA administration substantially increased hippocampal glutamate efflux which is related to memory processes. Accordingly, pre-tetanus administration of low MLA concentrations produced longer lasting potentiation (long-term potentiation, LTP) in studies examining hippocampal plasticity. Moreover, low doses of MLA improved acquisition, but not consolidation memory processes in rats. While the focus to enhance cognition by modulating α7nAChRs lies on agonists and positive modulators, antagonists at low doses should provide a novel approach to improve cognition in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Nick P van Goethem
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200, MD, Maastricht, The Netherlands
| | - Dean Paes
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200, MD, Maastricht, The Netherlands
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95124 Catania, Italy
| | - Ernesto Fedele
- Department of Pharmacy, Section of Pharmacology and Toxicology, School of Medical and Pharmaceutical Sciences, Centre of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy; IRCCS Polyclinic Hospital San Martino, 16132 Genoa, Italy
| | - Claudia Rebosio
- Department of Pharmacy, Section of Pharmacology and Toxicology, School of Medical and Pharmaceutical Sciences, Centre of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95124 Catania, Italy
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95124 Catania, Italy
| | | | - Youyi Peng
- Intra-Cellular Therapies, Inc., New York 10016, United States
| | - Daniel Bertrand
- HiQScreen Sàrl, 6, rte de Compois, 1222, Vésenaz, Geneva, Switzerland
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200, MD, Maastricht, The Netherlands.
| |
Collapse
|
53
|
Zhang X, Pan L, Yu J, Huang H. One recombinant C-type lectin (LvLec) from white shrimp Litopenaeus vannamei affected the haemocyte immune response in vitro. FISH & SHELLFISH IMMUNOLOGY 2019; 89:35-42. [PMID: 30890430 DOI: 10.1016/j.fsi.2019.03.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 06/09/2023]
Abstract
C-type lectin has received widespread attention in animal immunomodulation functions since it was discovered, but it is still limited in crustaceans. The present study is to explore effects of one recombinant C-type lectin (LvLec protein) on haemocyte immune response in Litopenaeus vannamei (L. vannamei). The methods of keeping haemocyte immune activity were optimised by the Key Laboratory of Mariculture. The experiment was divided into four groups: control group, recombinant protein group (LvLec protein, 1.0 mg mL-1), Lipopolysaccharide group (LPS, 1.0 mg mL-1), and LPS combine with LvLec protein group (LPS + LvLec protein, 1.0 mg mL-1 + 1.0 mg mL-1), while each group processes 0, 3, 6, 9, 12, and 24 h respectively. The results showed that the haemocyte count reduced, while the exocytosis PO activity, hemagglutinating activity and phagocytic activity promoted, and the concentration of cGMP and PKA increased after LvLec protein treatment. However, the levels of antibacterial activity and bacteriolytic activity as well as the concentrations of cAMP and PKG did not change significantly after treating with LvLec protein, LPS or LPS + LvLec protein. Therefore, these results suggest that LvLec protein can stimulate the exocytosis PO activity through cGMP-PKA pathway to affect the phagocytic activity and hemagglutinating activity of L. vannamei haemocytes in vitro.
Collapse
Affiliation(s)
- Xin Zhang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China.
| | - Jinhong Yu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Hui Huang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| |
Collapse
|
54
|
Liu Q, Li X, Zhao Y, Cao K, Liu Y, Xiao R, Wang C, Li Y, Huang W, Wang X. Dopamine D1 receptor agonist treatment alleviates morphine-exposure-induced learning and memory impairments. Brain Res 2019; 1711:120-129. [DOI: 10.1016/j.brainres.2019.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 01/06/2023]
|
55
|
Li ZH, Cui D, Qiu CJ, Song XJ. Cyclic nucleotide signaling in sensory neuron hyperexcitability and chronic pain after nerve injury. NEUROBIOLOGY OF PAIN 2019; 6:100028. [PMID: 31223142 PMCID: PMC6565612 DOI: 10.1016/j.ynpai.2019.100028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 11/08/2022]
Abstract
Activation of cAMP-PKA and cGMP-PKG pathways contributes to injury-induced sensory neuron hyperexcitability. Activation of cAMP and cGMP contributes to the development of bone cancer pain. PAR2 activation mediates injury-induced cAMP-dependent sensory neuron hyperexcitability.
The cyclic nucleotide signaling, including cAMP-PKA and cGMP-PKG pathways, has been well known to play critical roles in regulating cellular growth, metabolism and many other intracellular processes. In recent years, more and more studies have uncovered the roles of cAMP and cGMP in the nervous system. The cAMP and cGMP signaling mediates chronic pain induced by different forms of injury and stress. Here we summarize the roles of cAMP-PKA and cGMP-PKG signaling pathways in the pathogenesis of chronic pain after nerve injury. In addition, acute dissociation and chronic compression of the dorsal root ganglion (DRG) neurons, respectively, leads to neural hyperexcitability possibly through PAR2 activation-dependent activation of cAMP-PKA pathway. Clinically, radiotherapy can effectively alleviate bone cancer pain at least partly through inhibiting the cancer cell-induced activation of cAMP-PKA pathway. Roles of cyclic nucleotide signaling in neuropathic and inflammatory pain are also seen in many other animal models and are involved in many pro-nociceptive mechanisms including the activation of hyperpolarization-activated cyclic nucleotide (HCN)-modulated ion channels and the exchange proteins directly activated by cAMP (EPAC). Further understanding the roles of cAMP and cGMP signaling in the pathogenesis of chronic pain is theoretically significant and clinically valuable for treatment of chronic pain.
Collapse
Affiliation(s)
- Ze-Hua Li
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Dong Cui
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Cheng-Jie Qiu
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xue-Jun Song
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
56
|
Blokland A, Van Duinen MA, Sambeth A, Heckman PRA, Tsai M, Lahu G, Uz T, Prickaerts J. Acute treatment with the PDE4 inhibitor roflumilast improves verbal word memory in healthy old individuals: a double-blind placebo-controlled study. Neurobiol Aging 2019; 77:37-43. [PMID: 30776650 DOI: 10.1016/j.neurobiolaging.2019.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 11/30/2022]
Abstract
There is ample evidence that phosphodiesterase 4 (PDE4) inhibition can improve memory performance in animal studies. In the present study, we examined the acute effects of the PDE4 inhibitor roflumilast on memory performance in healthy individuals (60-80 years of age). We tested the effects of acute roflumilast administration (100, 250, 1000 μg) in a double-blind, placebo-controlled, 4-way crossover design. Participants were first screened for their verbal word memory performance to ensure normal memory performance (within 0.5 standard deviation from norm score; n = 20) Drug effects on memory performance were tested in a verbal memory test and a spatial memory test. Reported side effects of drug treatment were registered. Roflumilast (100 μg) improved the delayed recall performance of the participants (Cohen's d, 0.69). No effects were observed in the spatial memory task. Roflumilast was well tolerated at this low dose. Although no clear adverse side effects were reported at the low dose, mild adverse events (including headache, dizziness, insomnia, and diarrhea) were reported after the 1000 μg dose. The present study provides first evidence that the PDE4 inhibitor roflumilast improves verbal memory performance in old participants. The current data encourage further development of PDE4 inhibitors for improving memory.
Collapse
Affiliation(s)
- Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, the Netherlands
| | - Marlies A Van Duinen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Anke Sambeth
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, the Netherlands
| | - Pim R A Heckman
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, the Netherlands; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Max Tsai
- Department of Clinical Development, Takeda Development Center Americas, Deerfield, IL, USA
| | - Gezim Lahu
- Department of Clinical Development, Takeda Development Center Americas, Deerfield, IL, USA
| | - Tolga Uz
- Department of Clinical Development, Takeda Development Center Americas, Deerfield, IL, USA
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
57
|
Hollas MA, Ben Aissa M, Lee SH, Gordon-Blake JM, Thatcher GRJ. Pharmacological manipulation of cGMP and NO/cGMP in CNS drug discovery. Nitric Oxide 2019; 82:59-74. [PMID: 30394348 PMCID: PMC7645969 DOI: 10.1016/j.niox.2018.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/14/2018] [Accepted: 10/25/2018] [Indexed: 12/21/2022]
Abstract
The development of small molecule modulators of NO/cGMP signaling for use in the CNS has lagged far behind the use of such clinical agents in the periphery, despite the central role played by NO/cGMP in learning and memory, and the substantial evidence that this signaling pathway is perturbed in neurodegenerative disorders, including Alzheimer's disease. The NO-chimeras, NMZ and Nitrosynapsin, have yielded beneficial and disease-modifying responses in multiple preclinical animal models, acting on GABAA and NMDA receptors, respectively, providing additional mechanisms of action relevant to synaptic and neuronal dysfunction. Several inhibitors of cGMP-specific phosphodiesterases (PDE) have replicated some of the actions of these NO-chimeras in the CNS. There is no evidence that nitrate tolerance is a phenomenon relevant to the CNS actions of NO-chimeras, and studies on nitroglycerin in the periphery continue to challenge the dogma of nitrate tolerance mechanisms. Hybrid nitrates have shown much promise in the periphery and CNS, but to date only one treatment has received FDA approval, for glaucoma. The potential for allosteric modulation of soluble guanylate cyclase (sGC) in brain disorders has not yet been fully explored nor exploited; whereas multiple applications of PDE inhibitors have been explored and many have stalled in clinical trials.
Collapse
Affiliation(s)
- Michael A Hollas
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Manel Ben Aissa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Sue H Lee
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Jesse M Gordon-Blake
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, USA.
| |
Collapse
|
58
|
Cao LM, Dong ZQ, Li Q, Chen X. Treadmill training improves neurological deficits and suppresses neuronal apoptosis in cerebral ischemic stroke rats. Neural Regen Res 2019; 14:1387-1393. [PMID: 30964064 PMCID: PMC6524516 DOI: 10.4103/1673-5374.253523] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Rehabilitation training is believed to be beneficial to patients with stroke, but its molecular mechanism is still unclear. Rat models of cerebral ischemic stroke were established by middle cerebral artery occlusion/reperfusion, and then received treadmill training of different intensities, twice a day for 30 minutes for 1 week. Low-intensity training was conducted at 5 m/min, with a 10-minute running, 10-minute rest, and 10-minute running cycle. In the moderate-intensity training, the intensity gradually increased from 5 m/min to 10 m/min in 5 minutes, with the same rest cycle as above. In high-intensity training, the intensity gradually increased from 5 m/min to 25 m/min in 5 minutes, with the same rest cycle as above. The Bederson scale was used to evaluate the improvement of motor function. Infarct volume was detected using 2,3,5-triphenyltetrazolium chloride staining. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining was applied to detect the apoptosis of nerve cells in brain tissue. Western blot assay was employed to analyze the activation of cyclic adenosine monophosphate (cAMP)/protein kinase A and Akt/glycogen synthase kinase-3β signaling pathways in rat brain tissue. All training intensities reduced the neurological deficit score, infarct volume, and apoptosis in nerve cells in brain tissue of stroke rats. Training intensities activated the cAMP/protein kinase A and Akt/glycogen synthase kinase-3 beta signaling pathways. This activation was more obvious with higher training intensities. These changes were reversed by intracerebroventricular injection of protein kinase A inhibitor Rp-cAMP. Our findings indicate that the neuroprotective effect of rehabilitation training is achieved via activation of the cAMP/protein kinase A and Akt/glycogen synthase kinase-3 beta signaling pathways. This study was approved by the Ethics Committee of Animal Experimentation in Shanghai No. 8 People’s Hospital, China.
Collapse
Affiliation(s)
- Li-Mei Cao
- Department of Neurology, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Zhi-Qiang Dong
- Department of Neurology, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Qiang Li
- Department of Neurology, Shanghai No. 8 People's Hospital, Shanghai, China
| | - Xu Chen
- Department of Neurology, Shanghai No. 8 People's Hospital, Shanghai, China
| |
Collapse
|
59
|
Borghans LGJM, Sambeth A, Prickaerts J, Ramaekers JG, Blokland A. The effects of the soluble guanylate cyclase stimulator riociguat on memory performance in healthy volunteers with a biperiden-induced memory impairment. Psychopharmacology (Berl) 2018; 235:2407-2416. [PMID: 29882087 PMCID: PMC6061766 DOI: 10.1007/s00213-018-4938-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/30/2018] [Indexed: 10/25/2022]
Abstract
RATIONALE After stimulation with nitric oxide, soluble guanylate cyclase (sGC) produces cyclic guanosine monophosphate (cGMP), which stimulates an important signalling pathway for long-term potentiation (LTP). By upregulating cGMP, LTP could be stimulated and thereby enhancing memory processes. The present study investigated the effects of the sGC stimulator riociguat on cognition in healthy volunteers. Participants were pre-treated with and without biperiden, which impairs memory performance, to investigate the memory-enhancing effects of riociguat. METHODS Twenty volunteers participated in a double-blind placebo-controlled six-way crossover design with a cognitive test battery including the verbal learning task (VLT), n-back task, spatial memory test, the attention network test, and a reaction time task. Treatments were placebo and riociguat 0.5 mg, placebo and riociguat 1.0 mg, biperiden 2.0 mg and placebo, biperiden 2.0 mg and riociguat 0.5 mg and biperiden 2.0 mg and riociguat 1.0 mg. RESULTS Blood pressure was found to be decreased and heart rate to be increased after administration of riociguat. Cognitive performance was not enhanced after administration of riociguat. Biperiden decreased episodic memory on the VLT, yet this deficit was not reversed by riociguat. CONCLUSION This supports the notion that biperiden might be a valuable pharmacological model to induce episodic memory impairments as observed in AD/MCI.
Collapse
Affiliation(s)
- Laura G. J. M. Borghans
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Anke Sambeth
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Faculty of Medicine, Health & Life Science, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Johannes G. Ramaekers
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
60
|
Zhang L, Seo JH, Li H, Nam G, Yang HO. The phosphodiesterase 5 inhibitor, KJH-1002, reverses a mouse model of amnesia by activating a cGMP/cAMP response element binding protein pathway and decreasing oxidative damage. Br J Pharmacol 2018; 175:3347-3360. [PMID: 29847860 DOI: 10.1111/bph.14377] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhibition of PDE5 improves synaptic plasticity and memory via enhancing cGMP expression, thus activating the cGMP/cAMP response element binding protein (CREB) signalling pathway. This study investigated the effects of a PDE5 inhibitor on scopolamine-induced cognitive dysfunction, using memory-related behavioural tests and biochemical assays. EXPERIMENTAL APPROACH In mice were pretreated with PDE5 inhibitor, amnesia was induced by scopolamine. The learning and memory abilities of mice were tested using the Morris water maze test, the Y-maze test, the passive avoidance test and the novel object recognition test in sequence. Expression of memory-related bio-molecules and oxidative stress parameters in brain tissue were measured using Western blot and spectrophotometry respectively. KEY RESULTS KJH-1002, a novel and potent inhibitor of PDE5 (IC50 0.059 ± 0.04 nmol·L-1 ), was synthesized. In the behavioural tests, it markedly improved the memory performance impaired by scopolamine, indicating a restoration of cognitive function in the mice. Moreover, KJH-1002 increased cGMP levels in the cortex and the scopolamine-reduced expression of phosphorylated CREB, Levels of ERK 1/2, Akt and brain-derived neurotrophic factor in the cortex and hippocampus were restored by KJH-1002 treatment. In addition, KJH-1002 administration increased the activities of SOD, glutathione peroxidase and glutathione reductase, and decreased the level of malondialdehyde. CONCLUSION AND IMPLICATIONS KJH-1002 restored cognitive function in scopolamine-induced amnesia mice by activating the cGMP/CREB signalling pathway and attenuating oxidative stress. The beneficial effects of KJH-1002 on cognition indicate its potential as a therapeutic candidate for Alzheimer's disease.
Collapse
Affiliation(s)
- Lijun Zhang
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Korea.,Division of Bio-medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Jae Hong Seo
- Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Gyeonggi-do, Republic of Korea
| | - Huan Li
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Korea.,Division of Bio-medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Ghilsoo Nam
- Division of Bio-medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea.,Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Hyun Ok Yang
- Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Korea.,Division of Bio-medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
61
|
Gulisano W, Tropea MR, Arancio O, Palmeri A, Puzzo D. Sub-efficacious doses of phosphodiesterase 4 and 5 inhibitors improve memory in a mouse model of Alzheimer's disease. Neuropharmacology 2018; 138:151-159. [PMID: 29885420 DOI: 10.1016/j.neuropharm.2018.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/30/2018] [Accepted: 06/03/2018] [Indexed: 01/06/2023]
Abstract
Cyclic nucleotides cAMP and cGMP cooperate to ensure memory acquisition and consolidation. Increasing their levels by phosphodiesterase inhibitors (PDE-Is) enhanced cognitive functions and rescued memory loss in different models of aging and Alzheimer's disease (AD). However, side effects due to the high doses used limited their application in humans. Based on previous studies suggesting that combinations of sub-efficacious doses of cAMP- and cGMP-specific PDE-Is improved synaptic plasticity and memory in physiological conditions, here we aimed to study whether this treatment was effective to counteract the AD phenotype in APPswe mice. We found that a 3-week chronic treatment with a combination of sub-efficacious doses of the cAMP-specific PDE4-I roflumilast (0.01 mg/kg) and the cGMP-specific PDE5-I vardenafil (0.1 mg/kg) improved recognition, spatial and contextual fear memory. Importantly, the cognitive enhancement persisted for 2 months beyond administration. This long-lasting action, and the possibility to minimize side effects due to the low doses used, might open feasible therapeutic strategies against AD.
Collapse
Affiliation(s)
- Walter Gulisano
- Dept. Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Maria Rosaria Tropea
- Dept. Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Ottavio Arancio
- Dept. of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Agostino Palmeri
- Dept. Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Daniela Puzzo
- Dept. Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy.
| |
Collapse
|
62
|
Nelissen E, Prickaerts J, Blokland A. Acute stress negatively affects object recognition early memory consolidation and memory retrieval unrelated to state-dependency. Behav Brain Res 2018; 345:9-12. [DOI: 10.1016/j.bbr.2018.02.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 01/27/2023]
|
63
|
Liu L, Zheng J, Huang XF, Zhu X, Ding SM, Ke HM, O'Donnell JM, Zhang HT, Song GQ, Xu Y. The neuroprotective and antidepressant-like effects of Hcyb1, a novel selective PDE2 inhibitor. CNS Neurosci Ther 2018; 24:652-660. [PMID: 29704309 DOI: 10.1111/cns.12863] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/28/2018] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
AIMS Depression is currently the most common mood disorder. Regulation of intracellular cyclic adenosine monophosphate (cAMP) and/or cyclic guanosine monophosphate (cGMP) signaling by phosphodiesterase (PDE) inhibition has been paid much attention for treatment of depression. This study aimed to investigate the neuroprotective effects of Hcyb1, a novel PDE2 inhibitor, in HT-22 cells and antidepressant-like effects in mouse models of depression. METHODS Hcyb1 was synthesized and its selectivity upon PDE2 was tested. Moreover, HT-22 hippocampal cells were used to determine the effects of Hcyb1 on cell viability, cyclic nucleotide levels, and the downstream molecules related to cAMP/cGMP signaling by neurochemical, enzyme-linked immunosorbent, and immunoblot assays in vitro. The antidepressant-like effects of Hcyb1 were also determined in the forced swimming and tail suspension tests in mice. RESULTS Hcyb1 had a highly selective inhibition of PDE2A (IC50 = 0.57 ± 0.03 μmol/L) and over 250-fold selectivity against other recombinant PDE family members. Hcyb1 at concentrations of 10-10 and 10-9 mol/L significantly increased cell viability after treatment for 24 hours. At concentrations of 10-9 ~10-7 mol/L, Hcyb1 also increased cGMP levels by 1.7~2.3 folds after 10-minute treatment. Furthermore, Hcyb1 at the concentrations of 10-9 mol/L increased both cGMP and cAMP levels 24 hours after treatment. The levels of phosphorylation of CREB and BDNF were also increased by Hcyb1 treatment in HT-22 cells for 24 hours. Finally, in the in vivo tests, Hcyb1 (0.5, 1, and 2 mg/kg, i.g.) decreased the immobility time in both forced swimming and tail suspension tests, without altering locomotor activity. CONCLUSION These results suggest that the novel PDE2 inhibitor Hcyb1 produced neuroprotective and antidepressant-like effects most likely mediated by cAMP/cGMP-CREB-BDNF signaling.
Collapse
Affiliation(s)
- Li Liu
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Jing Zheng
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Xian-Feng Huang
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Xia Zhu
- Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Shu-Ming Ding
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Heng-Ming Ke
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Han-Ting Zhang
- Department of Behavioral Medicine & Psychiatry and Physiology, Pharmacology & Neuroscience, Rockefeller Neurosciences Institute, West Virginia University Health Science Center, Morgantown, WV, USA
| | - Guo-Qiang Song
- School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
64
|
Nakashima M, Imada H, Shiraishi E, Ito Y, Suzuki N, Miyamoto M, Taniguchi T, Iwashita H. Phosphodiesterase 2A Inhibitor TAK-915 Ameliorates Cognitive Impairments and Social Withdrawal in N-Methyl-d-Aspartate Receptor Antagonist-Induced Rat Models of Schizophrenia. J Pharmacol Exp Ther 2018; 365:179-188. [PMID: 29440309 DOI: 10.1124/jpet.117.245506] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 02/05/2018] [Indexed: 11/22/2022] Open
Abstract
The pathophysiology of schizophrenia has been associated with glutamatergic dysfunction. Modulation of the glutamatergic signaling pathway, including N-methyl-d-aspartate (NMDA) receptors, can provide a new therapeutic target for schizophrenia. Phosphodiesterase 2A (PDE2A) is highly expressed in the forebrain, and is a dual substrate enzyme that hydrolyzes both cAMP and cGMP, which play pivotal roles as intracellular second messengers downstream of NMDA receptors. Here we characterize the in vivo pharmacological profile of a selective and 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) as a novel treatment of schizophrenia. Oral administration of TAK-915 at 3 and 10 mg/kg significantly increased cGMP levels in the frontal cortex, hippocampus, and striatum of rats. TAK-915 at 10 mg/kg significantly upregulated the phosphorylation of α-amino-3-hydroxy-5-methylisoxazole-4-proprionic acid receptor subunit GluR1 in the rat hippocampus. TAK-915 at 3 and 10 mg/kg significantly attenuated episodic memory deficits induced by the NMDA receptor antagonist (+)-MK-801 hydrogen maleate (MK-801) in the rat passive avoidance test. TAK-915 at 10 mg/kg significantly attenuated working memory deficits induced by MK-801 in the rat radial arm maze test. Additionally, TAK-915 at 10 mg/kg prevented subchronic phencyclidine-induced social withdrawal in social interaction in rats. In contrast, TAK-915 did not produce antipsychotic-like activity; TAK-915 had little effect on MK-801- or methamphetamine-induced hyperlocomotion in rats. These results suggest that TAK-915 has a potential to ameliorate cognitive impairments and social withdrawal in schizophrenia.
Collapse
Affiliation(s)
- Masato Nakashima
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruka Imada
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Eri Shiraishi
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuki Ito
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Noriko Suzuki
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Maki Miyamoto
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takahiko Taniguchi
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroki Iwashita
- Neuroscience Drug Discovery Unit (M.N., H.Im., E.S., Y.I., N.S., T.T., H.Iw.) and Drug Metabolism and Pharmacokinetics Research Laboratories (M.M.), Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
65
|
Acute administration of roflumilast enhances immediate recall of verbal word memory in healthy young adults. Neuropharmacology 2018; 131:31-38. [DOI: 10.1016/j.neuropharm.2017.12.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 12/31/2022]
|
66
|
cAMP, cGMP and Amyloid β: Three Ideal Partners for Memory Formation. Trends Neurosci 2018; 41:255-266. [PMID: 29501262 DOI: 10.1016/j.tins.2018.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 02/03/2023]
Abstract
cAMP and cGMP are well established second messengers required for long-term potentiation (LTP) and memory formation/consolidation. By contrast, amyloid β (Aβ), mostly known as one of the main culprits for Alzheimer's disease (AD), has received relatively little attention in the context of plasticity and memory. Of note, however, low physiological concentrations of Aβ seem necessary for LTP induction and for memory formation. This should come as no surprise, since hormesis emerged as a central dogma in biology. Additionally, recent evidence indicates that Aβ is one of the downstream effectors for cAMP and cGMP to trigger synaptic plasticity and memory. We argue that these emerging findings depict a new scenario that should change the general view on the amyloidogenic pathway, and that could have significant implications for the understanding of AD and its pharmacological treatment in the future.
Collapse
|
67
|
Zuccarini M, Giuliani P, Frinchi M, Mudò G, Serio RM, Belluardo N, Buccella S, Carluccio M, Condorelli DF, Caciagli F, Ciccarelli R, Di Iorio P. Uncovering the Signaling Pathway behind Extracellular Guanine-Induced Activation of NO System: New Perspectives in Memory-Related Disorders. Front Pharmacol 2018; 9:110. [PMID: 29515443 PMCID: PMC5826394 DOI: 10.3389/fphar.2018.00110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/31/2022] Open
Abstract
Mounting evidence suggests that the guanine-based purines stand out as key player in cell metabolism and in several models of neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases. Guanosine (GUO) and guanine (GUA) are extracellular signaling molecules derived from the breakdown of the correspondent nucleotide, GTP, and their intracellular and extracellular levels are regulated by the fine-tuned activity of two major enzymes, purine nucleoside phosphorylase (PNP) and guanine deaminase (GDA). Noteworthy, GUO and GUA, seem to play opposite roles in the modulation of cognitive functions, such as learning and memory. Indeed GUO, despite exerting neuroprotective, anti-apoptotic and neurotrophic effects, causes a decay of cognitive activities, whereas GUA administration in rats results in working memory improvement (prevented by L-NAME pre-treatment). This study was designed to investigate, in a model of SH-SY5Y neuroblastoma cell line, the signal transduction pathway activated by extracellular GUA. Altogether, our results showed that: (i) in addition to an enhanced phosphorylation of ASK1, p38 and JNK, likely linked to a non-massive and transient ROS production, the PKB/NO/sGC/cGMP/PKG/ERK cascade seems to be the main signaling pathway elicited by extracellular GUA; (ii) the activation of this pathway occurs in a pertussis-toxin sensitive manner, thus suggesting the involvement of a putative G protein coupled receptor; (iii) the GUA-induced NO production, strongly reduced by cell pre-treatment with L-NAME, is negatively modulated by the EPAC-cAMP-CaMKII pathway, which causes the over-expression of GDA that, in turn, reduces the levels of GUA. These molecular mechanisms activated by GUA may be useful to support our previous observation showing that GUA improves learning and memory functions through the stimulation of NO signaling pathway, and underscore the therapeutic potential of oral administration of guanine for treating memory-related disorders.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patricia Giuliani
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Monica Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Rosa Maria Serio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Silvana Buccella
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Marzia Carluccio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | | | - Francesco Caciagli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
- Aging Research Center, Ce.S.I., “G. d’Annunzio” University Foundation, Chieti, Italy
| |
Collapse
|
68
|
Heckman PRA, Blokland A, Bollen EPP, Prickaerts J. Phosphodiesterase inhibition and modulation of corticostriatal and hippocampal circuits: Clinical overview and translational considerations. Neurosci Biobehav Rev 2018; 87:233-254. [PMID: 29454746 DOI: 10.1016/j.neubiorev.2018.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/20/2022]
Abstract
The corticostriatal and hippocampal circuits contribute to the neurobiological underpinnings of several neuropsychiatric disorders, including Alzheimer's disease, Parkinson's disease and schizophrenia. Based on biological function, these circuits can be clustered into motor circuits, associative/cognitive circuits and limbic circuits. Together, dysfunctions in these circuits produce the wide range of symptoms observed in related neuropsychiatric disorders. Intracellular signaling in these circuits is largely mediated through the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway with an additional role for the cyclic guanosine monophosphate (cGMP)/ protein kinase G (PKG) pathway, both of which can be regulated by phosphodiesterase inhibitors (PDE inhibitors). Through their effects on cAMP response element-binding protein (CREB) and Dopamine- and cAMP-Regulated PhosphoProtein MR 32 kDa (DARPP-32), cyclic nucleotide pathways are involved in synaptic transmission, neuron excitability, neuroplasticity and neuroprotection. In this clinical review, we provide an overview of the current clinical status, discuss the general mechanism of action of PDE inhibitors in relation to the corticostriatal and hippocampal circuits and consider several translational challenges.
Collapse
Affiliation(s)
- P R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands.
| | - A Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands
| | - E P P Bollen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - J Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
69
|
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
|
70
|
Qiu Y, Wang Y, Wang X, Wang C, Xia ZY. Role of the hippocampal 5-HT1A receptor-mediated cAMP/PKA signalling pathway in sevoflurane-induced cognitivedysfunction in aged rats. J Int Med Res 2018; 46:1073-1085. [PMID: 29332488 PMCID: PMC5972259 DOI: 10.1177/0300060517744037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective This study aimed to evaluate the role of the hippocampal 5-hydroxytryptamine-1A (5-HT1A)-mediated cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signalling pathway in sevoflurane-induced cognitive dysfunction in aged rats. Methods Sixty 18-month-old Sprague–Dawley rats were divided into the control (n = 30) and experimental (Sev, n = 30) groups. The experimental group inhaled 50% air/oxygen mixture (2 L/min) and 2% sevoflurane for 4 hours. The control group inhaled 50% air/oxygen mixture (2 L/min) for 4 hours. The Morris water maze test was performed The mRNA expression of 5-HT1A receptor, and cAMP PKA, cAMP response element-binding protein (CREB), and phosphorylated CREB (p-CREB) protein expression were determined. Results The escape latency and swimming distance were greater, and the number of crossings of the platform location and time spent in the platform quadrant were less in the Sev group compared with the control group. cAMP, PKA, CREB, and p-CREB protein expression was downregulated in the Sev group 1 day after anaesthesia compared with the control group. Hippocampal 5-HT1A receptor mRNA expression was higher 7 days after anaesthesia compared with the control group. Conclusion Sevoflurane-induced cognitive dysfunction in aged rats may be related to inhibited expression of the hippocampal 5-HT1A receptor-mediated cAMP/PKA signalling pathway.
Collapse
Affiliation(s)
- Yi Qiu
- 1 Department of Anesthesiology, Renmin Hospitai of Wuhan University, Wuhan, Hubei Province, China
| | - Ying Wang
- 2 Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia, China
| | - Xiaodong Wang
- 2 Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia, China
| | - Caixia Wang
- 2 Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia, China
| | - Zhong-Yuan Xia
- 1 Department of Anesthesiology, Renmin Hospitai of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
71
|
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: 128] [Impact Index Per Article: 16.0] [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
|
72
|
Calcagno E, Caudano F, Passalacqua M, Pronzato MA, Fedele E, Ricciarelli R. Investigating the amyloid-beta enhancing effect of cGMP in neuro2a cells. Mech Ageing Dev 2017; 166:1-5. [PMID: 28789837 DOI: 10.1016/j.mad.2017.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 10/24/2022]
Abstract
Long-term potentiation (LTP) and the process of memory formation require activation of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) pathways. Notably, recent evidence indicated that both cyclic nucleotides boost the production of amyloid-beta (Aβ) peptides. In particular, cAMP was shown to favor hippocampal LTP by stimulating the synthesis of the amyloid precursor protein APP, whereas cGMP was found to enhance LTP and to improve memory by increasing Aβ levels without affecting the expression of APP. The results of the present study substantiate that cGMP has a role in the endocytic pathway of APP and suggest a scenario where the cyclic nucleotide enhances the production of Aβ by favoring the trafficking of APP from the cell cortex to the endolysosomal compartment.
Collapse
Affiliation(s)
- Elisa Calcagno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Francesca Caudano
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Maria A Pronzato
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Ernesto Fedele
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | |
Collapse
|
73
|
Abstract
High levels of amyloid-β peptide (Aβ) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aβ are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aβ levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aβ due to a change in the approximation of amyloid precursor protein (APP) and the β-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aβ function, by using anti-murine Aβ antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aβ levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-β (Aβ) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aβ, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aβ levels and function during LTP. We show that cGMP enhances Aβ production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aβ, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aβ.
Collapse
|
74
|
de Matos AM, de Macedo MP, Rauter AP. Bridging Type 2 Diabetes and Alzheimer's Disease: Assembling the Puzzle Pieces in the Quest for the Molecules With Therapeutic and Preventive Potential. Med Res Rev 2017; 38:261-324. [PMID: 28422298 DOI: 10.1002/med.21440] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/18/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes (T2D) and Alzheimer's disease (AD) are two age-related amyloid diseases that affect millions of people worldwide. Broadly supported by epidemiological data, the higher incidence of AD among type 2 diabetic patients led to the recognition of T2D as a tangible risk factor for the development of AD. Indeed, there is now growing evidence on brain structural and functional abnormalities arising from brain insulin resistance and deficiency, ultimately highlighting the need for new approaches capable of preventing the development of AD in type 2 diabetic patients. This review provides an update on overlapping pathophysiological mechanisms and pathways in T2D and AD, such as amyloidogenic events, oxidative stress, endothelial dysfunction, aberrant enzymatic activity, and even shared genetic background. These events will be presented as puzzle pieces put together, thus establishing potential therapeutic targets for drug discovery and development against T2D and diabetes-induced cognitive decline-a heavyweight contributor to the increasing incidence of dementia in developed countries. Hoping to pave the way in this direction, we will present some of the most promising and well-studied drug leads with potential against both pathologies, including their respective bioactivity reports, mechanisms of action, and structure-activity relationships.
Collapse
Affiliation(s)
- Ana Marta de Matos
- Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016, Lisbon, Portugal.,CEDOC Chronic Diseases, Nova Medical School, Rua Câmara Pestana n 6, 6-A, Ed. CEDOC II, 1150-082, Lisbon, Portugal
| | - Maria Paula de Macedo
- CEDOC Chronic Diseases, Nova Medical School, Rua Câmara Pestana n 6, 6-A, Ed. CEDOC II, 1150-082, Lisbon, Portugal
| | - Amélia Pilar Rauter
- Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016, Lisbon, Portugal
| |
Collapse
|
75
|
Memory-enhancing effects of GEBR-32a, a new PDE4D inhibitor holding promise for the treatment of Alzheimer's disease. Sci Rep 2017; 7:46320. [PMID: 28402318 PMCID: PMC5389348 DOI: 10.1038/srep46320] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/13/2017] [Indexed: 11/20/2022] Open
Abstract
Memory loss characterizes several neurodegenerative disorders, including Alzheimer’s disease (AD). Inhibition of type 4 phosphodiesterase (PDE4) and elevation of cyclic adenosine monophosphate (cAMP) has emerged as a promising therapeutic approach to treat cognitive deficits. However, PDE4 exists in several isoforms and pan inhibitors cannot be used in humans due to severe emesis. Here, we present GEBR-32a, a new PDE4D full inhibitor that has been characterized both in vitro and in vivo using biochemical, electrophysiological and behavioural analyses. GEBR-32a efficiently enhances cAMP in neuronal cultures and hippocampal slices. In vivo pharmacokinetic analysis shows that GEBR-32a is rapidly distributed within the central nervous system with a very favourable brain/blood ratio. Specific behavioural tests (object location and Y-maze continuous alternation tasks) demonstrate that this PDE4D inhibitor is able to enhance memory in AD transgenic mice and concomitantly rescues their hippocampal long-term potentiation deficit. Of great relevance, our preliminary toxicological analysis indicates that GEBR-32a is not cytotoxic and genotoxic, and does not seem to possess emetic-like side effects. In conclusion, GEBR-32a could represent a very promising cognitive-enhancing drug with a great potential for the treatment of Alzheimer’s disease.
Collapse
|
76
|
Selective phosphodiesterase-2A inhibitor alleviates radicular inflammation and mechanical allodynia in non-compressive lumbar disc herniation rats. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2017; 26:1961-1968. [PMID: 28283839 DOI: 10.1007/s00586-017-5023-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/18/2017] [Accepted: 02/28/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE Phosphodiesterase inhibitors possess anti-inflammatory properties. In addition, some studies report that phosphodiesterase 2A (PDE2A) are highly expressed in the dorsal horn of the spinal cord. The present study aimed to investigate whether intrathecal administration of Bay 60-7550, a specific PDE2A inhibitor, could alleviate mechanical allodynia in non-compressive lumbar disc herniation (NCLDH) rats. METHODS Rat NCLDH models by autologous nucleus pulposus implantation to dorsal root ganglion were established. Vehicle or Bay 60-7550 (0.1, 1.0 mg/kg) was injected by intrathecal catheter at day 1 post-operation. The ipsilateral mechanical withdrawal thresholds were analyzed from the day before surgery to day 7 after surgery. At day 7 post-operation, the ipsilateral lumbar (L4-L6) segments of the spinal dorsal horns were removed, and tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), cyclic adenosine monophosphate (cAMP), and cyclic guanosine monophosphate (cGMP) expressions were measured by ELISA. Furthermore, PDE2A mRNA and protein expressions in spinal cord were measured by Real-Time PCR and Western blot. RESULTS Intrathecal administration of the PDE2A inhibitor Bay 60-7550, significantly attenuated mechanical allodynia, down-regulated spinal TNF-α, IL-1β and IL-6 over-expressions, increased the expression of spinal cAMP, as well as cGMP in a more remarkable manner, and decreased the spinal PDE2A expression in NCLDH rats in a dose-dependent manner. CONCLUSIONS Bay 60-7550 alleviated mechanical allodynia and inflammation in NCLDH rats, which might be associated with increased cAMP and especially cGMP increase. Thus, spinal PDE2A inhibition might represent a potential analgesic strategy for radiculopathy treatment in non-compressive lumbar disc herniation.
Collapse
|
77
|
Gomez L, Massari ME, Vickers T, Freestone G, Vernier W, Ly K, Xu R, McCarrick M, Marrone T, Metz M, Yan YG, Yoder ZW, Lemus R, Broadbent NJ, Barido R, Warren N, Schmelzer K, Neul D, Lee D, Andersen CB, Sebring K, Aertgeerts K, Zhou X, Tabatabaei A, Peters M, Breitenbucher JG. Design and Synthesis of Novel and Selective Phosphodiesterase 2 (PDE2a) Inhibitors for the Treatment of Memory Disorders. J Med Chem 2017; 60:2037-2051. [DOI: 10.1021/acs.jmedchem.6b01793] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Laurent Gomez
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Mark Eben Massari
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Troy Vickers
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Graeme Freestone
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - William Vernier
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Kiev Ly
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Rui Xu
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Margaret McCarrick
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Tami Marrone
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Markus Metz
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Yingzhou G. Yan
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Zachary W. Yoder
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Robert Lemus
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Nicola J. Broadbent
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Richard Barido
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Noelle Warren
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Kara Schmelzer
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - David Neul
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Dong Lee
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Carsten B. Andersen
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Kristen Sebring
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Kathleen Aertgeerts
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Xianbo Zhou
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Ali Tabatabaei
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - Marco Peters
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| | - J. Guy Breitenbucher
- Dart Neuroscience LLC, 12278 Scripps Summit Drive, San Diego, California 92131, United States
| |
Collapse
|
78
|
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: 20] [Impact Index Per Article: 2.5] [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
|
79
|
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
|
80
|
Li J, Liu CN, Wei N, Li XD, Liu YY, Yang R, Jia YJ. Protective effects of BAY 73-6691, a selective inhibitor of phosphodiesterase 9, on amyloid-β peptides-induced oxidative stress in in-vivo and in-vitro models of Alzheimer's disease. Brain Res 2016; 1642:327-335. [DOI: 10.1016/j.brainres.2016.04.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/06/2016] [Accepted: 04/05/2016] [Indexed: 12/11/2022]
|
81
|
Vanmierlo T, Creemers P, Akkerman S, van Duinen M, Sambeth A, De Vry J, Uz T, Blokland A, Prickaerts J. The PDE4 inhibitor roflumilast improves memory in rodents at non-emetic doses. Behav Brain Res 2016; 303:26-33. [DOI: 10.1016/j.bbr.2016.01.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/10/2016] [Accepted: 01/13/2016] [Indexed: 11/29/2022]
|
82
|
Lueptow LM, Zhan CG, O'Donnell JM. Cyclic GMP-mediated memory enhancement in the object recognition test by inhibitors of phosphodiesterase-2 in mice. Psychopharmacology (Berl) 2016; 233:447-56. [PMID: 26525565 DOI: 10.1007/s00213-015-4129-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/14/2015] [Indexed: 12/24/2022]
Abstract
RATIONALE AND OBJECTIVES Cyclic nucleotide phosphodiesterase-2 (PDE2) is a potential therapeutic target for the treatment of cognitive dysfunction. Using the object recognition test (ORT), this study assessed the effects of two PDE2 inhibitors, Bay 60-7550 and ND7001, on learning and memory, and examined underlying mechanisms. METHODS To assess the role of PDE2 inhibition on phases of memory, Bay 60-7550 (3 mg/kg) was administered: 30 min prior to training; 0, 1, or 3 h after training; or 30 min prior to recall testing. To assess cyclic nucleotide involvement in PDE2 inhibitor-enhanced memory consolidation, either the nitric oxide synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME; 20 mg/kg; intraperitoneal (IP)), soluble guanylyl cyclase inhibitor 1H-[-1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ; 20 mg/kg; IP), protein kinase G inhibitor KT5823 (2.5 μg; intracerebroventricular (ICV)), or protein kinase A inhibitor H89 (1 μg; ICV) was administered 30 min prior to the PDE2 inhibitor Bay 60-7550 (3 mg/kg) or ND7001 (3 mg/kg). Changes in the phosphorylation of 3'5'-cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) at Ser-133 and vasodilator-stimulated phosphoprotein (VASP) at Ser-239 were determined to confirm activation of cAMP and 3'5'-cyclic guanosine monophosphate (cGMP) signaling. RESULTS Bay 60-7550 (3 mg/kg) enhanced memory of mice in the ORT when given 30 min prior to training, immediately after training, or 30 min prior to recall. Inhibitors of the cGMP pathway blocked the memory-enhancing effects of both Bay 60-7550 (3 mg/kg) and ND7001 (3 mg/kg) on early consolidation processes. Bay 60-7550 (3 mg/kg) enhanced phosphorylation of CREB and VASP, both targets of cGMP-dependent protein kinase (PKG). CONCLUSIONS These results confirm a potential of PDE2, or components of its signaling pathway, as a therapeutic target for drug discovery focused on restoring memory function.
Collapse
Affiliation(s)
- Lindsay M Lueptow
- Neuroscience Graduate Program, West Virginia University Health Sciences Center, Morgantown, WV, USA.
- West Virginia University, 1 Medical Center Drive, PO Box 9128, Morgantown, WV, USA.
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center and Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA
| | - James M O'Donnell
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
83
|
Snyder GL, Prickaerts J, Wadenberg ML, Zhang L, Zheng H, Yao W, Akkerman S, Zhu H, Hendrick JP, Vanover KE, Davis R, Li P, Mates S, Wennogle LP. Preclinical profile of ITI-214, an inhibitor of phosphodiesterase 1, for enhancement of memory performance in rats. Psychopharmacology (Berl) 2016; 233:3113-24. [PMID: 27342643 PMCID: PMC4980415 DOI: 10.1007/s00213-016-4346-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/07/2016] [Indexed: 01/07/2023]
Abstract
RATIONALE Therapeutic agents for memory enhancement in psychiatric disorders, such as schizophrenia, are urgently needed. OBJECTIVE The aim of this study is to characterize the preclinical profile of ITI-214, a potent inhibitor of phosphodiesterase 1 (PDE1). METHODS ITI-214 was assayed for inhibition of PDE1 versus other PDE enzyme families using recombinant human PDE enzymes and for off-target binding to 70 substrates (General SEP II diversity panel; Caliper Life Sciences). Effects of ITI-214 (0.1-10 mg/kg, po) on memory performance were assayed in rats using the novel object recognition (NOR) paradigm, with drug given at specified time points prior to or following exposure to objects in an open field. ITI-214 was evaluated for potential drug-drug interaction with risperidone in rats using conditioned avoidance response (CAR) and pharmacokinetic assessments. RESULTS ITI-214 inhibited PDE1A (K i = 33 pmol) with >1000-fold selectivity for the nearest other PDE family (PDE4D) and displayed minimal off-target binding interactions in a 70-substrate selectivity profile. By using specific timing of oral ITI-214 administration, it was demonstrated in the NOR that ITI-214 is able to enhance acquisition, consolidation, and retrieval memory processes. All memory effects were in the absence of effects on exploratory behavior. ITI-214 did not disrupt the risperidone pharmacokinetic profile or effects in CAR. CONCLUSIONS ITI-214 improved the memory processes of acquisition, consolidation, and retrieval across a broad dose range (0.1-10 mg/kg, po) without disrupting the antipsychotic-like activity of a clinical antipsychotic medication, specifically risperidone. Clinical development of ITI-214 is currently in progress.
Collapse
Affiliation(s)
- Gretchen L. Snyder
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, NL-6200 Maastricht, MD The Netherlands
| | | | - Lei Zhang
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Hailin Zheng
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Wei Yao
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Sven Akkerman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, NL-6200 Maastricht, MD The Netherlands
| | - Hongwen Zhu
- Tianjin Hospital, Tianjin, 300211 People’s Republic of China
| | - Joseph P. Hendrick
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Kimberly E. Vanover
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Robert Davis
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Peng Li
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Sharon Mates
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| | - Lawrence P. Wennogle
- Intra-Cellular Therapies Inc., 430 East 29th Street, Suite 900, New York, NY 10016 USA
| |
Collapse
|
84
|
Akkerman S, Blokland A, Prickaerts J. Possible overlapping time frames of acquisition and consolidation phases in object memory processes: a pharmacological approach. ACTA ACUST UNITED AC 2015; 23:29-37. [PMID: 26670184 PMCID: PMC4749836 DOI: 10.1101/lm.040162.115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/03/2015] [Indexed: 02/03/2023]
Abstract
In previous studies, we have shown that acetylcholinesterase inhibitors and phosphodiesterase inhibitors (PDE-Is) are able to improve object memory by enhancing acquisition processes. On the other hand, only PDE-Is improve consolidation processes. Here we show that the cholinesterase inhibitor donepezil also improves memory performance when administered within 2 min after the acquisition trial. Likewise, both PDE5-I and PDE4-I reversed the scopolamine deficit model when administered within 2 min after the learning trial. PDE5-I was effective up to 45 min after the acquisition trial and PDE4-I was effective when administered between 3 and 5.5 h after the acquisition trial. Taken together, our study suggests that acetylcholine, cGMP, and cAMP are all involved in acquisition processes and that cGMP and cAMP are also involved in early and late consolidation processes, respectively. Most important, these pharmacological studies suggest that acquisition processes continue for some time after the learning trial where they share a short common time frame with early consolidation processes. Additional brain concentration measurements of the drugs suggest that these acquisition processes can continue up to 4-6 min after learning.
Collapse
Affiliation(s)
- Sven Akkerman
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, European Graduate School of Neuroscience
| | - Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, European Graduate School of Neuroscience
| |
Collapse
|
85
|
Abstract
Cyclic adenosine monophosphate (cAMP) is one of the second messengers critically involved in the molecular mechanisms underlying memory formation. In the CNS, the availability of cAMP is tightly controlled by phosphodiesterase 4 (PDE4), a family of enzymes that degrades the cyclic nucleotide to inactive AMP. Among the different PDE4 isoforms, in the last few years PDE4D has been hogging the limelight due to accumulating evidence for its crucial role in cognitive processes, which makes this enzyme a promising target for therapeutic interventions in a variety of pathological conditions characterized by memory impairment, such as Alzheimer's disease. In this article, we review the role of the cAMP signal transduction pathway in memory formation with a particular focus on the recent progress in PDE4D research.
Collapse
Affiliation(s)
- Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, University of Genoa, Genoa, Italy
| | - Ernesto Fedele
- Department of Pharmacy, Section of Pharmacology and Toxicology, Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
86
|
A key role for TGF-β1 in hippocampal synaptic plasticity and memory. Sci Rep 2015; 5:11252. [PMID: 26059637 PMCID: PMC4462026 DOI: 10.1038/srep11252] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/08/2015] [Indexed: 01/06/2023] Open
Abstract
Transforming Growth Factor β1 (TGF-β1) is a well-known neuroprotective and neurotrophic factor demonstrated to play a role in synaptic transmission. However, its involvement in physiological mechanisms underlying synaptic plasticity and memory at hippocampal level has not been thoroughly investigated. Here, we examine the role of TGF-β1 in hippocampal long-term potentiation (LTP) and memory in adult wild type mice. Our data provide evidence that administration of exogenous TGF-β1 is able to convert early-phase-LTP into late-phase-LTP. Furthermore, we show that the block of the endogenous TGF-β1 signaling pathway by the specific TGF-β1 inhibitor SB431542, impairs LTP and object recognition memory. The latter impairment was rescued by administration of exogenous TGF-β1, suggesting that endogenously produced TGF-β1 plays a role in physiological mechanisms underlying LTP and memory. Finally, TGF-β1 functional effect correlates with an increased expression of the phosphorylated transcription factor cAMP-Responsive Element Binding protein.
Collapse
|
87
|
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
|
88
|
Teich AF, Nicholls RE, Puzzo D, Fiorito J, Purgatorio R, Fa’ M, Arancio O. Synaptic therapy in Alzheimer's disease: a CREB-centric approach. Neurotherapeutics 2015; 12:29-41. [PMID: 25575647 PMCID: PMC4322064 DOI: 10.1007/s13311-014-0327-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).
Collapse
Affiliation(s)
- Andrew F. Teich
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Russell E. Nicholls
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Daniela Puzzo
- />Department of Bio-Medical Sciences, Section of Physiology, University of Catania, Catania, 95125 Italy
| | - Jole Fiorito
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Rosa Purgatorio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Mauro Fa’
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Ottavio Arancio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| |
Collapse
|
89
|
Heckman PRA, Blokland A, Ramaekers J, Prickaerts J. PDE and cognitive processing: beyond the memory domain. Neurobiol Learn Mem 2014; 119:108-22. [PMID: 25464010 DOI: 10.1016/j.nlm.2014.10.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 01/01/2023]
Abstract
Phosphodiesterase inhibitors (PDE-Is) enhance cAMP and/or cGMP signaling via reducing the degradation of these cyclic nucleotides. Both cAMP and cGMP signaling are essential for a variety of cellular functions and exert their effects both pre- and post-synaptically. Either of these second messengers relays and amplifies incoming signals at receptors on the cell surface making them important elements in signal transduction cascades and essential in cellular signaling in a variety of cell functions including neurotransmitter release and neuroprotection. Consequently, these processes can be influenced by PDE-Is as they increase cAMP and/or cGMP concentrations. PDE-Is have been considered as possible therapeutic agents to treat impaired memory function linked to several brain disorders, including depression, schizophrenia and Alzheimer's disease (AD). This review will, however, focus on the possible role of phosphodiesterases (PDEs) in cognitive decline beyond the memory domain. Here we will discuss the involvement of PDEs on three related domains: attention, information filtering (sensory- and sensorimotor gating) and response inhibition (drug-induced hyperlocomotion). Currently, these are emerging cognitive domains in the field of PDE research. Here we discuss experimental studies and the potential beneficial effects of PDE-I drugs on these cognitive domains, as effects of PDE-Is on these domains could potentially influence effects on memory performance. Overall, PDE4 seems to be the most promising target for all domains discussed in this review.
Collapse
Affiliation(s)
- P R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands; Department of Neuropsychology and Psychopharmacology, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - A Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - J Ramaekers
- Department of Neuropsychology and Psychopharmacology, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - J Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
90
|
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: 73] [Impact Index Per Article: 6.6] [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
|