1
|
Nelissen E, Schepers M, Ponsaerts L, Foulquier S, Bronckaers A, Vanmierlo T, Sandner P, Prickaerts J. Soluble guanylyl cyclase: A novel target for the treatment of vascular cognitive impairment? Pharmacol Res 2023; 197:106970. [PMID: 37884069 DOI: 10.1016/j.phrs.2023.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Vascular cognitive impairment (VCI) describes neurodegenerative disorders characterized by a vascular component. Pathologically, it involves decreased cerebral blood flow (CBF), white matter lesions, endothelial dysfunction, and blood-brain barrier (BBB) impairments. Molecularly, oxidative stress and inflammation are two of the major underlying mechanisms. Nitric oxide (NO) physiologically stimulates soluble guanylate cyclase (sGC) to induce cGMP production. However, under pathological conditions, NO seems to be at the basis of oxidative stress and inflammation, leading to a decrease in sGC activity and expression. The native form of sGC needs a ferrous heme group bound in order to be sensitive to NO (Fe(II)sGC). Oxidation of sGC leads to the conversion of ferrous to ferric heme (Fe(III)sGC) and even heme-loss (apo-sGC). Both Fe(III)sGC and apo-sGC are insensitive to NO, and the enzyme is therefore inactive. sGC activity can be enhanced either by targeting the NO-sensitive native sGC (Fe(II)sGC), or the inactive, oxidized sGC (Fe(III)sGC) and the heme-free apo-sGC. For this purpose, sGC stimulators acting on Fe(II)sGC and sGC activators acting on Fe(III)sGC/apo-sGC have been developed. These sGC agonists have shown their efficacy in cardiovascular diseases by restoring the physiological and protective functions of the NO-sGC-cGMP pathway, including the reduction of oxidative stress and inflammation, and improvement of vascular functioning. Yet, only very little research has been performed within the cerebrovascular system and VCI pathology when focusing on sGC modulation and its potential protective mechanisms on vascular and neural function. Therefore, within this review, the potential of sGC as a target for treating VCI is highlighted.
Collapse
Affiliation(s)
- Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Laura Ponsaerts
- Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium; Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience (MHeNS), School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Annelies Bronckaers
- Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; Hannover Medical School, 30625 Hannover, Germany
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
2
|
Yanai S, Tago T, Toyohara J, Arasaki T, Endo S. Reversal of spatial memory impairment by phosphodiesterase 3 inhibitor cilostazol is associated with reduced neuroinflammation and increased cerebral glucose uptake in aged male mice. Front Pharmacol 2022; 13:1031637. [PMID: 36618932 PMCID: PMC9810637 DOI: 10.3389/fphar.2022.1031637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The nucleotide second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP) mediate fundamental functions of the brain, including learning and memory. Phosphodiesterase 3 (PDE3) can hydrolyze both cAMP and cGMP and appears to be involved in the regulation of their contents in cells. We previously demonstrated that long-term administration of cilostazol, a PDE3 inhibitor, maintained good memory performance in aging mice. Here, we report on studies aimed at determining whether cilostazol also reverses already-impaired memory in aged male mice. One month of oral 1.5% cilostazol administration in 22-month-old mice reversed age-related declines in hippocampus-dependent memory tasks, including the object recognition and the Morris water maze. Furthermore, cilostazol reduced neuroinflammation, as evidenced by immunohistochemical staining, and increased glucose uptake in the brain, as evidence by positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). These results suggest that already-expressed memory impairment in aged male mice that depend on cyclic nucleotide signaling can be reversed by inhibition of PDE3. The reversal of age-related memory impairments may occur in the central nervous system, either through cilostazol-enhanced recall or strengthening of weak memories that otherwise may be resistant to recall.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan,*Correspondence: Shogo Endo,
| |
Collapse
|
3
|
Gomaa AA, Farghaly HS, Ahmed AM, El-Mokhtar MA, Hemida FK. Advancing combination treatment with cilostazol and caffeine for Alzheimer's disease in high fat-high fructose-STZ induced model of amnesia. Eur J Pharmacol 2022; 921:174873. [DOI: 10.1016/j.ejphar.2022.174873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022]
|
4
|
de Havenon A, Sheth KN, Madsen TE, Johnston KC, Turan T, Toyoda K, Elm JJ, Wardlaw JM, Johnston SC, Williams OA, Shoamanesh A, Lansberg MG. Cilostazol for Secondary Stroke Prevention: History, Evidence, Limitations, and Possibilities. Stroke 2021; 52:e635-e645. [PMID: 34517768 PMCID: PMC8478840 DOI: 10.1161/strokeaha.121.035002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cilostazol is a PDE3 (phosphodiesterase III) inhibitor with a long track record of safety that is Food and Drug Administration and European Medicines Agency approved for the treatment of claudication in patients with peripheral arterial disease. In addition, cilostazol has been approved for secondary stroke prevention in several Asian countries based on trials that have demonstrated a reduction in stroke recurrence among patients with noncardioembolic stroke. The onset of benefit appears after 60 to 90 days of treatment, which is consistent with cilostazol's pleiotropic effects on platelet aggregation, vascular remodeling, blood flow, and plasma lipids. Cilostazol appears safe and does not increase the risk of major bleeding when given alone or in combination with aspirin or clopidogrel. Adverse effects such as headache, gastrointestinal symptoms, and palpitations, however, contributed to a 6% increase in drug discontinuation among patients randomized to cilostazol in a large secondary stroke prevention trial (CSPS.com [Cilostazol Stroke Prevention Study for Antiplatelet Combination]). Due to limitations of prior trials, such as open-label design, premature trial termination, large loss to follow-up, lack of functional or cognitive outcome data, and exclusive enrollment in Asia, the existing trials have not led to a change in clinical practice or guidelines in Western countries. These limitations could be addressed by a double-blind placebo-controlled randomized trial conducted in a broader population. If positive, it would increase the evidence in support of long-term treatment with cilostazol for secondary prevention in the millions of patients worldwide who have experienced a noncardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Adam de Havenon
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kevin N. Sheth
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tracy E. Madsen
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Karen C. Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tanya Turan
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kazunori Toyoda
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Jordan J. Elm
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Joanna M. Wardlaw
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - S. Claiborne Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Olajide A. Williams
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Ashkan Shoamanesh
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Maarten G. Lansberg
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| |
Collapse
|
5
|
Jankowska A, Wesołowska A, Pawłowski M, Chłoń-Rzepa G. Multifunctional Ligands Targeting Phosphodiesterase as the Future Strategy for the Symptomatic and Disease-Modifying Treatment of Alzheimer’s Disease. Curr Med Chem 2020; 27:5351-5373. [DOI: 10.2174/0929867326666190620095623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Alzheimer’s Disease (AD) is a chronic neurodegenerative disorder characterized by cognitive
impairments such as memory loss, decline in language skills, and disorientation that affects
over 46 million people worldwide. Patients with AD also suffer from behavioral and psychological
symptoms of dementia that deteriorate their quality of life and lead to premature death. Currently
available drugs provide modest symptomatic relief but do not reduce pathological hallmarks (senile
plaques and neurofibrillary tangles) and neuroinflammation, both of which are integral parts of dementia.
A large body of evidence indicates that impaired signaling pathways of cyclic-3′,5′-
Adenosine Monophosphate (cAMP) and cyclic-3′,5′-guanosine Monophosphate (cGMP) may contribute
to the development and progression of AD. In addition, Phosphodiesterase (PDE) inhibitors,
commonly known as cAMP and/or cGMP modulators, were found to be involved in the phosphorylation
of tau; aggregation of amyloid beta; neuroinflammation; and regulation of cognition, mood,
and emotion processing. The purpose of this review was to update the most recent reports on the
development of novel multifunctional ligands targeting PDE as potential drugs for both symptomatic
and disease-modifying therapy of AD. This review collected the chemical structures of representative
multifunctional ligands, results of experimental in vitro and in vivo pharmacological studies,
and current opinions regarding the potential utility of these compounds for the comprehensive
therapy of AD. Finally, the multiparameter predictions of drugability of the representative compounds
were calculated and discussed.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Maciej Pawłowski
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Grażyna Chłoń-Rzepa
- Department of Medicinal Chemistry, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| |
Collapse
|
6
|
PDE3 Inhibitors Repurposed as Treatments for Age-Related Cognitive Impairment. Mol Neurobiol 2018; 56:4306-4316. [PMID: 30311144 DOI: 10.1007/s12035-018-1374-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/27/2018] [Indexed: 12/21/2022]
Abstract
As the population of older individuals grows worldwide, researchers have increasingly focused their attention on identifying key molecular targets of age-related cognitive impairments, with the aim of developing possible therapeutic interventions. Two such molecules are the intracellular cyclic nucleotides, cAMP and cGMP. These second messengers mediate fundamental aspects of brain function relevant to memory, learning, and cognitive function. Consequently, phosphodiesterases (PDEs), which hydrolyze cAMP and cGMP, are promising targets for the development of cognition-enhancing drugs. Inhibitors that target PDEs work by elevating intracellular cAMP. In this review, we provide an overview of different PDE inhibitors, and then we focus on pharmacological and physiological effects of PDE3 inhibitors in the CNS and peripheral tissues. Finally, we discuss findings from experimental and preliminary clinical studies and the potential beneficial effects of the PDE3 inhibitor cilostazol on age-related cognitive impairments. In the innovation pipeline of pharmaceutical development, the antiplatelet agent cilostazol has come into the spotlight as a novel treatment for mild cognitive impairment. Overall, the repurposing of cilostazol may represent a potentially promising way to treat mild cognitive impairment, Alzheimer's disease, and vascular dementia. In this review, we present a brief summary of cAMP signaling and different PDE inhibitors, followed by a discussion of the pharmacological and physiological role of PDE3 inhibitors. In this context, we discuss the repurposing of a PDE3 inhibitor, cilostazol, as a potential treatment for age-related cognitive impairment based on recent research.
Collapse
|
7
|
Santiago A, Soares LM, Schepers M, Milani H, Vanmierlo T, Prickaerts J, Weffort de Oliveira RM. Roflumilast promotes memory recovery and attenuates white matter injury in aged rats subjected to chronic cerebral hypoperfusion. Neuropharmacology 2018; 138:360-370. [DOI: 10.1016/j.neuropharm.2018.06.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 04/24/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
|
8
|
Mohamed MZ, Hafez HM, Zenhom NM, Mohammed HH. Cilostazol alleviates streptozotocin-induced testicular injury in rats via PI3K/Akt pathway. Life Sci 2018; 198:136-142. [DOI: 10.1016/j.lfs.2018.02.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/22/2018] [Accepted: 02/24/2018] [Indexed: 12/19/2022]
|
9
|
Nunes Santiago A, Dias Fiuza Ferreira E, Weffort de Oliveira RM, Milani H. Cognitive, neurohistological and mortality outcomes following the four-vessel occlusion/internal carotid artery model of chronic cerebral hypoperfusion: The impact of diabetes and aging. Behav Brain Res 2018; 339:169-178. [DOI: 10.1016/j.bbr.2017.11.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 10/18/2022]
|
10
|
Yanai S, Ito H, Endo S. Long-term cilostazol administration prevents age-related decline of hippocampus-dependent memory in mice. Neuropharmacology 2017; 129:57-68. [PMID: 29122629 DOI: 10.1016/j.neuropharm.2017.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/30/2017] [Accepted: 11/04/2017] [Indexed: 12/17/2022]
Abstract
Phosphodiesterases (PDEs) are enzymes that hydrolyze and inactivate 3', 5'-cyclic adenosine monophosphate (cAMP) and/or 3', 5'-cyclic guanosine monophosphate (cGMP). The regulation of intracellular signaling pathways mediated by cyclic nucleotides is imperative to synaptic plasticity and memory in animals. Because PDEs play an important role in this regulation, PDE inhibitors are considered as candidate compounds for treating cognitive and memory disorders. In the present study, we tested whether cilostazol, a selective PDE3 inhibitor, prevents the cognitive deterioration that occurs during the course of normal aging in mice. Ten months of cilostazol administration (1.5%) in 13-month-old mice improved spatial memory when tested at 23 months of age. First, it prevented the decline in the ability of these aged mice to recognize a change in an object's location in the object recognition task. Second, spatial memory of these cilostazol-treated aged mice in the Morris water maze was comparable to that of untreated middle-aged mice (13 months old). Cilostazol administration had no effect on the emotional states and physical ability of aged mice. Thus, long-term cilostazol administration prevented hippocampus-dependent memory decline in aged mice, allowing them to achieve a level of cognitive performance similar to middle-aged mice and without negative behavioral side effects. Considering its well-established safety in other medical contexts, cilostazol may be a potential therapeutic candidate drug for staving off cognitive decline in the aging human population.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Hideki Ito
- Department of CNS Research, Otsuka Pharmaceutical Co., Ltd., Tokushima, 771-0192, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan.
| |
Collapse
|
11
|
Ölmestig JNE, Marlet IR, Hainsworth AH, Kruuse C. Phosphodiesterase 5 inhibition as a therapeutic target for ischemic stroke: A systematic review of preclinical studies. Cell Signal 2017; 38:39-48. [PMID: 28648945 DOI: 10.1016/j.cellsig.2017.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/10/2017] [Accepted: 06/20/2017] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase 5 inhibitors (PDE5i), such as sildenafil (Viagra®) are widely used for erectile dysfunction and pulmonary hypertension. Preclinical studies suggest that PDE5i may improve functional outcome following ischemic stroke. In this systematic review we aimed to evaluate the effects of selective PDE5i in animal models of brain ischaemia. A systematic search in Medline, Embase, and The Cochrane Library was performed including studies in English assessing the effects of selective PDE5i. 32 publications were included describing outcome in 3646 animals. Neuroprotective effects of PDE5i were dependent on the NO-cGMP-PKG-pathway. These included reduced neuronal apoptosis (n=3 studies), oxidative stress (n=5), and neuroinflammation (n=2). PDE5i increased angiogenesis and elevated regional cerebral blood flow in the ischemic penumbra, and improved functional recovery. Some studies found that PDE5i treatment reduced lesion volume (n=9), others found no effect (n=9). Treatment was effective when administered within 24h post-ischemia, though treatment delayed to seven days improved outcome in one study. This review demonstrates both neuroprotective and neurorestorative effects of PDE5i in animal models of stroke, though the specific underlying signaling pathways relating to PDE5 inhibition and cGMP may remain serendipitous in some studies. There is currently limited evidence on the effects of selective PDE5i in human stroke patients, hence translation of preclinical results into clinical trials may be warranted.
Collapse
Affiliation(s)
- Joakim N E Ölmestig
- Neurovascular Research Unit, Department of Neurology, Herlev Gentofte Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Ida R Marlet
- Neurovascular Research Unit, Department of Neurology, Herlev Gentofte Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Atticus H Hainsworth
- Clinical Neuroscience, Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Christina Kruuse
- Neurovascular Research Unit, Department of Neurology, Herlev Gentofte Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| |
Collapse
|
12
|
de Oliveira JN, Reis LO, Ferreira EDF, Godinho J, Bacarin CC, Soares LM, de Oliveira RMW, Milani H. Postischemic fish oil treatment confers task-dependent memory recovery. Physiol Behav 2017; 177:196-207. [DOI: 10.1016/j.physbeh.2017.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/27/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022]
|
13
|
Yao XL, Yao ZH, Li L, Nie L, Zhang SF. Oxiracetam can improve cognitive impairment after chronic cerebral hypoperfusion in rats. Psychiatry Res 2016; 246:284-292. [PMID: 27741481 DOI: 10.1016/j.psychres.2016.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/10/2016] [Accepted: 10/04/2016] [Indexed: 10/20/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) induces cognitive deficits. Although CCH can be improved, cognitive impairment is not improved accordingly. To date, many studies have focused on investigating the pathophysiological mechanisms of CCH; however, the treatment of the induced cognitive impairment remains ineffective. Thus, the mechanisms underlying cognitive impairment after CCH and potential agents for treating this impairment need to be explored further. Oxiracetam is a nootropic drug that improves clinical outcomes for some central nervous system (CNS) disorders. Whether it can improve cognitive impairment after CCH is unknown. In this study, we used behavioural methods, electrophysiology, biochemistry, histopathological staining and transmission electron microscope to investigate rat's cognitive impairment by CCH, and found that Oxiracetam could improve CCH-induced cognitive impairment and prevent deficits of neural plasticity, white matter lesions, and synaptic ultrastructure. These results suggest that Oxiracetam may be effective as a potential agent against CCH-induced cognitive impairment.
Collapse
Affiliation(s)
- Xiao-Li Yao
- Department of Neurology, Central hospital of Zhengzhou, #195 Tongbo Road, Zhengzhou, China; Department of Neurology, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China
| | - Zhao-Hui Yao
- Department of Geriatrics, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China; Department of Neurology, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China.
| | - Li Li
- Department of Geriatrics, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China; Department of Neurology, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China
| | - Li Nie
- Department of Geriatrics, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China; Department of Neurology, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China
| | - Shao-Feng Zhang
- Department of Geriatrics, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China; Department of Neurology, Renmin hospital of Wuhan University, #238 Jiefang Road, Wuhan, China
| |
Collapse
|
14
|
Yanai S, Toyohara J, Ishiwata K, Ito H, Endo S. Long-term cilostazol administration ameliorates memory decline in senescence-accelerated mouse prone 8 (SAMP8) through a dual effect on cAMP and blood-brain barrier. Neuropharmacology 2016; 116:247-259. [PMID: 27979612 DOI: 10.1016/j.neuropharm.2016.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/18/2016] [Accepted: 12/09/2016] [Indexed: 12/12/2022]
Abstract
Phosphodiesterases (PDEs), which hydrolyze and inactivate 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP), play an important role in synaptic plasticity that underlies memory. Recently, several PDE inhibitors were assessed for their possible therapeutic efficacy in treating cognitive disorders. Here, we examined how cilostazol, a selective PDE3 inhibitor, affects brain functions in senescence-accelerated mouse prone 8 (SAMP8), an animal model of age-related cognitive impairment. Long-term administration of cilostazol restored the impaired context-dependent conditioned fear memory of SAMP8 to match that in normal aging control substrain SAMR1. Cilostazol also increased the number of cells containing phosphorylated cAMP-responsive element binding protein (CREB), a downstream component of the cAMP pathway. Finally, cilostazol improves blood-brain barrier (BBB) integrity, demonstrated by reduced extravasation of 2-deoxy-2-18F-fluoro-d-glucose and Evans Blue dye in the brains of SAMP8. This improvement in BBB integrity was associated with an increased amount of zona occludens protein 1 (ZO-1) and occludin proteins, components of tight junctions integral to the BBB. The results suggest that long-term administration of cilostazol exerts its beneficial effects on age-related cognitive impairment through a dual mechanism: by enhancing the cAMP system in the brain and by maintaining or improving BBB integrity.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan; Institute of Cyclotron and Drug Discovery Research, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Fukushima 963-8052, Japan; Department of Biofunctional Imaging, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hideki Ito
- Department of CNS Research, Otsuka Pharmaceutical Co., Ltd., Tokushima 771-0192, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan.
| |
Collapse
|
15
|
Qi DS, Tao JH, Zhang LQ, Li M, Wang M, Qu R, Zhang SC, Liu P, Liu F, Miu JC, Ma JY, Mei XY, Zhang F. Neuroprotection of Cilostazol against ischemia/reperfusion-induced cognitive deficits through inhibiting JNK3/caspase-3 by enhancing Akt1. Brain Res 2016; 1653:67-74. [PMID: 27769787 DOI: 10.1016/j.brainres.2016.10.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/04/2016] [Accepted: 10/17/2016] [Indexed: 12/28/2022]
Abstract
Cilostazol(CTL) is a phosphodiesterase inhibitor, which has been widely used as anti-platelet agent. It also has preventive effects on various central nervous system (CNS) diseases, including ischemic stroke, Parkinson's disease and Alzheimer disease. However, the molecular mechanism underlying the protective effects of CTL is still unclear, and whether CTL can prevent I/R induced cognitive deficit has not been reported. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The open field tasks and Morris water maze were used to assess the effect of CTL on anxiety-like behavioral and cognitive impairment after I/R. Western blotting were performed to examine the expression of related proteins, and HE-staining was used to detect the percentage of neuronal death in the hippocampal CA1 region. Here we found that CTL significantly improved cognitive deficits and the behavior of rats in Morris water maze and open field tasks (P<0.05). HE staining results showed that CTL could significantly protect CA1 neurons against cerebral I/R (P<0.05). Additionally, Akt1 phosphorylation levels were evidently up-regulated (P<0.05), while the activation of JNK3, which is an important contributor to I/R-induced neuron apoptosis, was reduced by CTL after I/R (P<0.05), and caspase-3 levels were also decreased by CTL treatment. Furthermore, all of CTL's protective effects were reversed by LY294002, which is a PI3K/Akt1 inhibitor. Taken together, our results suggest that CTL could protect hippocampal neurons and ameliorate the impairment of learning/memory abilities and locomotor/ exploratory activities in ischemic stroke via a PI3K-Akt1/JNK3/caspase-3 dependent mechanism.
Collapse
Affiliation(s)
- Da-Shi Qi
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, People's Republic of China; Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China.
| | - Jin-Hao Tao
- Pediatric Emergency and Critical Care Center, Children' Hospital of Fudan University, Shanghai, People's Republic of China
| | - Lian-Qin Zhang
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, People's Republic of China; Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Man Li
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, People's Republic of China; Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Mei Wang
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Rui Qu
- Xuzhou Medical College affiliated Hospital, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Shi-Chun Zhang
- Xuzhou Mine Hosptial, Xuzhou, Jiangsu, People's Republic of China
| | - Pei Liu
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Fuming Liu
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, People's Republic of China
| | - Jian-Cheng Miu
- Sino-British SIPPR/B&K Lab Animal Ltd., People's Republic of China
| | - Jing-Yi Ma
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Xin-Yu Mei
- Interdisciplinary Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry and Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, People's Republic of China
| | - Fayong Zhang
- Department of Neurosurgery, Huashan Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
16
|
Abstract
Antidementive drugs have made Alzheimer's disease a symptomatically treatable condition but have essentially bypassed vascular dementia. The complexity of its pathology, the variable pathognomonic manifestations, the absence of validated biomarkers and the continuum with Alzheimer's disease that makes vascular dementia a comparatively small market are the major contributing factors. This report discusses how drug repurposing can be harnessed to identify new therapeutic opportunities, where such efforts are already yielding promising results, and which ones must be considered failures. Most investigations address obvious aspects cerebral small vessel disease, but some early-stage developments attempt changes in gene expression or modulation of complex biological pathways. A stronger focus on the nature and dynamics of white matter lesions should yield additional molecular targets.
Collapse
Affiliation(s)
- Hermann AM Mucke
- HM Pharma Consultancy, Enenkelstrasse 28/32, A-1160 Vienna, Austria
| |
Collapse
|
17
|
Zaghi GGD, Godinho J, Ferreira EDF, Ribeiro MHDM, Previdelli IS, de Oliveira RMW, Milani H. Robust and enduring atorvastatin-mediated memory recovery following the 4-vessel occlusion/internal carotid artery model of chronic cerebral hypoperfusion in middle-aged rats. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:179-87. [PMID: 26485403 DOI: 10.1016/j.pnpbp.2015.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/12/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) is a common condition associated with the development and/or worsening of age-related dementia.We previously reported persistent memory loss and neurodegeneration after CCH in middle-aged rats. Statin-mediated neuroprotection has been reported after acute cerebral ischemia. Unknown, however, is whether statins can alleviate the outcome of CCH. The present study investigated whether atorvastatin attenuates the cognitive and neurohistological outcome of CCH. Rats (12–15 months old) were trained in a non-food-rewarded radial maze, and then subjected to CCH. Atorvastatin (10 mg/kg, p.o.) was administered for 42 days or 15 days, beginning 5 h after the first occlusion stage. Retrograde memory performance was assessed at 7, 14, 21, 28, and 35 days of CCH, and expressed by “latency,” “number of reference memory errors” and “number of working memory errors.” Neurodegeneration was then examined at the hippocampus and cerebral cortex. Compared to sham, CCH caused profound and persistent memory loss in the vehicle-treated groups, as indicated by increased latency (91.2% to 107.3%) and number of errors (123.5% to 2508.2%), effects from which the animals did not spontaneously recover across time. This CCH-induced retrograde amnesia was completely prevented by atorvastatin (latency: −4.3% to 3.3%; reference/working errors: −2.5% to 45.7%), regardless of the treatment duration. This effect was sustained during the entire behavioral testing period (5 weeks), even after discontinuing treatment. This robust and sustained memory-protective effect of atorvastatin occurred in the absence of neuronal rescue (39.58% to 56.45% cell loss). We suggest that atorvastatin may be promising for the treatment of cognitive sequelae associated with CCH.
Collapse
Affiliation(s)
| | - Jacqueline Godinho
- Department of Pharmacology and Therapeutics, Health Science Center, Brazil
| | | | - Matheus Henrique Dal Molin Ribeiro
- Department of Statistics, Exact Science Center, State University of Maringá, Maringá, Brazil; Federal Institute of Parana, Palmas, Paraná, Brazil
| | | | | | - Humberto Milani
- Department of Pharmacology and Therapeutics, Health Science Center, Brazil.
| |
Collapse
|