1
|
Sadeghi MA, Hemmati S, Yousefi-Manesh H, Foroutani L, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Dehpour AR, Chamanara M. Cilostazol pretreatment prevents PTSD-related anxiety behavior through reduction of hippocampal neuroinflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:133-144. [PMID: 37382600 DOI: 10.1007/s00210-023-02578-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Current pharmacological treatments against post-traumatic stress disorder (PTSD) lack adequate efficacy. As a result, intense research has focused on identifying other molecular pathways mediating the pathogenesis of this condition. One such pathway is neuroinflammation, which has demonstrated a role in PTSD pathogenesis by causing synaptic dysfunction, neuronal death, and functional impairment in the hippocampus. Phosphodiesterase (PDE) inhibitors (PDEIs) have emerged as promising therapeutic agents against neuroinflammation in other neurological conditions. Furthermore, PDEIs have shown some promise in animal models of PTSD. However, the current model of PTSD pathogenesis, which is based on dysregulated fear learning, implies that PDE inhibition in neurons should enhance the acquisition of fear memory from the traumatic event. As a result, we hypothesized that PDEIs may improve PTSD symptoms through inhibiting neuroinflammation rather than long-term potentiation-related mechanisms. To this end, we tested the therapeutic efficacy of cilostazol, a selective inhibitor of PDE3, on PTSD-related anxiety symptoms in the underwater trauma model of PTSD. PDE3 is expressed much more richly in microglia and astrocytes compared to neurons in the murine brain. Furthermore, we used hippocampal indolamine 2,3-dioxygenase 1 (IDO) expression and interleukin 1 beta (IL-1β) concentration as indicators of neuroinflammation. We observed that cilostazol pretreatment prevented the development of anxiety symptoms and the increase in hippocampal IDO and IL-1β following PTSD induction. As a result, PDE3 inhibition ameliorated the neuroinflammatory processes involved in the development of PTSD symptoms. Therefore, cilostazol and other PDEIs may be promising candidates for further investigation as pharmacological therapies against PTSD.
Collapse
Affiliation(s)
- Mohammad Amin Sadeghi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hasan Yousefi-Manesh
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Laleh Foroutani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba Yousefi Zoshk
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pediatrics, AJA University of Medical Sciences, Tehran, Iran
| | - Yasaman Hosseini
- Cognitive Neuroscience Center, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Abbasian
- Management and Health Economics Department, AJA University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Cilostazol as an adjunctive treatment in major depressive disorder: a pilot randomized, double-blind, and placebo-controlled clinical trial. Psychopharmacology (Berl) 2022; 239:551-559. [PMID: 35072758 DOI: 10.1007/s00213-021-06041-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/30/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cilostazol, a phosphodiesterase-3 inhibitor, has been reported to improve depressive-like behavior in experimental studies of depression. We investigated the safety and efficacy of cilostazol combination therapy with sertraline in treating patients with major depressive disorder (MDD) in a 6-week, parallel, randomized controlled trial. METHOD Among patients referred to the outpatient clinic of a tertiary hospital, those with a diagnosis of MDD with moderate to severe severity (a score of >19 on the Hamilton depression rating scale (HAM-D)) were enrolled. A total of 54 MDD patients aged 18-65 years were randomly assigned to either the cilostazol (100 mg daily) or the placebo group. Both groups received sertraline 100 mg per day similarly. Changes in HAM-D at weeks 2, 4, and 6 were the primary outcome. Participants and outcome assessors were blinded. RESULTS At week 6, patients in the cilostazol group had significantly lower HAM-D score (p value= 0.015). General linear model repeated-measure analysis showed significant effect for treatment in improving MDD severity (p value <0.001). The remission rate at the study endpoint and number of responders at week 4 were significantly higher in the cilostazol group (p value= 0.047, p value= 0.032, respectively). The cilostazol group demonstrated a significantly shorter time to response. No significant difference was observed in treatment response at the study endpoint, and there were no serious adverse effects. CONCLUSION Our study supports safety and efficacy of cilostazol in treating MDD patients. TRIAL REGISTRATION This trial was registered at the Iranian registry of clinical trials (IRCT: www.irct.ir ; registration number: IRCT20090117001556N130).
Collapse
|
3
|
Transdermal System Based on Solid Cilostazol Nanoparticles Attenuates Ischemia/Reperfusion-Induced Brain Injury in Mice. NANOMATERIALS 2021; 11:nano11041009. [PMID: 33920878 PMCID: PMC8071240 DOI: 10.3390/nano11041009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/04/2021] [Accepted: 04/12/2021] [Indexed: 11/17/2022]
Abstract
Cilostazol (CIL) exerted a protective effect by promoting blood-brain barrier integrity as well as improving the status of neurological dysfunctions following cerebral ischemia/reperfusion (I/R) injury. We attempted to design a 0.5% CIL carbopol gel using solid nanoparticles (CIL-Ngel), and then investigated the relationships between energy-dependent endocytosis and the skin penetration of CIL-Ngel in this study. In addition, we evaluated whether the CIL-Ngel attenuated I/R-induced brain injury in a middle cerebral artery occlusion (MCAO)/reperfusion model mouse. The particle size of CIL was decreased using a bead mill, and the CIL particles (14.9 × 1014 particles/0.3 g) in the CIL-Ngel were approximately 50-180 nm. The release of CIL in the CIL-Ngel was higher than that in gel containing CIL powder (CIL-Mgel), and the CIL particles were released from the CIL-Ngel as nanoparticles. In addition, the percutaneous absorption of CIL from the CIL-Ngel was higher in comparison with that from CIL-Mgel, and clathrin-dependent endocytosis and caveolae-dependent endocytosis were related to the enhanced skin penetration of CIL-NPs. In the traditional (oral administration of CIL powder, 3 mg/kg) and transdermal administration (CIL-Ngel, 0.3 g) for 3 days (once a day), the area under the plasma CIL concentration-time curves (AUC) was similar, although the CIL supplied to the blood by the CIL-Ngel was more sustained than that via oral administration of CIL powder. Furthermore, the CIL-Ngel attenuated the ischemic stroke. In conclusion, we designed a gel using solid CIL-NPs, and we showed that the sustained release of CIL by CIL-Ngel provided an effective treatment for ischemic stroke in MCAO/reperfusion model mice. These findings induce the possibilities of developing novel applications of CIL solid nanoparticles.
Collapse
|
4
|
Rombaut B, Kessels S, Schepers M, Tiane A, Paes D, Solomina Y, Piccart E, Hove DVD, Brône B, Prickaerts J, Vanmierlo T. PDE inhibition in distinct cell types to reclaim the balance of synaptic plasticity. Theranostics 2021; 11:2080-2097. [PMID: 33500712 PMCID: PMC7797685 DOI: 10.7150/thno.50701] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Synapses are the functional units of the brain. They form specific contact points that drive neuronal communication and are highly plastic in their strength, density, and shape. A carefully orchestrated balance between synaptogenesis and synaptic pruning, i.e., the elimination of weak or redundant synapses, ensures adequate synaptic density. An imbalance between these two processes lies at the basis of multiple neuropathologies. Recent evidence has highlighted the importance of glia-neuron interactions in the synaptic unit, emphasized by glial phagocytosis of synapses and local excretion of inflammatory mediators. These findings warrant a closer look into the molecular basis of cell-signaling pathways in the different brain cells that are related to synaptic plasticity. In neurons, intracellular second messengers, such as cyclic guanosine or adenosine monophosphate (cGMP and cAMP, respectively), are known mediators of synaptic homeostasis and plasticity. Increased levels of these second messengers in glial cells slow down inflammation and neurodegenerative processes. These multi-faceted effects provide the opportunity to counteract excessive synapse loss by targeting cGMP and cAMP pathways in multiple cell types. Phosphodiesterases (PDEs) are specialized degraders of these second messengers, rendering them attractive targets to combat the detrimental effects of neurological disorders. Cellular and subcellular compartmentalization of the specific isoforms of PDEs leads to divergent downstream effects for these enzymes in the various central nervous system resident cell types. This review provides a detailed overview on the role of PDEs and their inhibition in the context of glia-neuron interactions in different neuropathologies characterized by synapse loss. In doing so, it provides a framework to support future research towards finding combinational therapy for specific neuropathologies.
Collapse
|
5
|
Bacopa monnieri (L.) Wettst. Extract Improves Memory Performance via Promotion of Neurogenesis in the Hippocampal Dentate Gyrus of Adolescent Mice. Int J Mol Sci 2020; 21:ijms21093365. [PMID: 32397562 PMCID: PMC7247711 DOI: 10.3390/ijms21093365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/29/2022] Open
Abstract
Bacopa monnieri L. Wettst. (BM) is a botanical component of Ayurvedic medicines and of dietary supplements used worldwide for cognitive health and function. We previously reported that administration of BM alcoholic extract (BME) prevents trimethyltin (TMT)-induced cognitive deficits and hippocampal cell damage and promotes TMT-induced hippocampal neurogenesis. In this study, we demonstrate that administration of BME improves spatial working memory in adolescent (5-week- old) healthy mice but not adult (8-week-old) mice. Moreover, improved spatial working memory was retained even at 4 weeks after terminating 1-week treatment of adolescent mice. One-week BME treatment of adolescent mice significantly enhanced hippocampal BrdU incorporation and expression of genes involved in neurogenesis determined by RNAseq analysis. Cell death, as detected by histochemistry, appeared not to be significant. A significant increase in neurogenesis was observed in the dentate gyrus region 4 weeks after terminating 1-week treatment of adolescent mice with BME. Bacopaside I, an active component of BME, promoted the proliferation of neural progenitor cells in vitro in a concentration-dependent manner via the facilitation of the Akt and ERK1/2 signaling. These results suggest that BME enhances spatial working memory in healthy adolescent mice by promoting hippocampal neurogenesis and that the effects of BME are due, in significant amounts, to bacopaside I.
Collapse
|
6
|
Pham HTN, Phan SV, Tran HN, Phi XT, Le XT, Nguyen KM, Fujiwara H, Yoneyama M, Ogita K, Yamaguchi T, Matsumoto K. Bacopa monnieri (L.) Ameliorates Cognitive Deficits Caused in a Trimethyltin-Induced Neurotoxicity Model Mice. Biol Pharm Bull 2019; 42:1384-1393. [DOI: 10.1248/bpb.b19-00288] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | | | | | | | | | - Hironori Fujiwara
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama
| | - Masanori Yoneyama
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Kiyokazu Ogita
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Taro Yamaguchi
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Kinzo Matsumoto
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama
| |
Collapse
|
7
|
Schepers M, Tiane A, Paes D, Sanchez S, Rombaut B, Piccart E, Rutten BPF, Brône B, Hellings N, Prickaerts J, Vanmierlo T. Targeting Phosphodiesterases-Towards a Tailor-Made Approach in Multiple Sclerosis Treatment. Front Immunol 2019; 10:1727. [PMID: 31396231 PMCID: PMC6667646 DOI: 10.3389/fimmu.2019.01727] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) characterized by heterogeneous clinical symptoms including gradual muscle weakness, fatigue, and cognitive impairment. The disease course of MS can be classified into a relapsing-remitting (RR) phase defined by periods of neurological disabilities, and a progressive phase where neurological decline is persistent. Pathologically, MS is defined by a destructive immunological and neuro-degenerative interplay. Current treatments largely target the inflammatory processes and slow disease progression at best. Therefore, there is an urgent need to develop next-generation therapeutic strategies that target both neuroinflammatory and degenerative processes. It has been shown that elevating second messengers (cAMP and cGMP) is important for controlling inflammatory damage and inducing CNS repair. Phosphodiesterases (PDEs) have been studied extensively in a wide range of disorders as they breakdown these second messengers, rendering them crucial regulators. In this review, we provide an overview of the role of PDE inhibition in limiting pathological inflammation and stimulating regenerative processes in MS.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Assia Tiane
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Dean Paes
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Selien Sanchez
- Department of Morphology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Ben Rombaut
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Elisabeth Piccart
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bart P F Rutten
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Bert Brône
- Department of Physiology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Niels Hellings
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Tim Vanmierlo
- Department of Neuroimmunology, European Graduate School of Neuroscience, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Department Psychiatry and Neuropsychology, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
8
|
Park YS, Jin Z, Jeong EA, Yi CO, Lee JY, Park IS, Roh GS. Cilostazol attenuates kainic acid-induced hippocampal cell death. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 22:63-70. [PMID: 29302213 PMCID: PMC5746513 DOI: 10.4196/kjpp.2018.22.1.63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 01/27/2023]
Abstract
Cilostazol is a selective inhibitor of type 3 phosphodiesterase (PDE3) and has been widely used as an antiplatelet agent. Cilostazol mediates this activity through effects on the cyclic adenosine monophosphate (cAMP) signaling cascade. Recently, it has attracted attention as a neuroprotective agent. However, little is known about cilostazol's effect on excitotoxicity induced neuronal cell death. Therefore, this study evaluated the neuroprotective effect of cilostazol treatment against hippocampal neuronal damage in a mouse model of kainic acid (KA)-induced neuronal loss. Cilostazol pretreatment reduced KA-induced seizure scores and hippocampal neuron death. In addition, cilostazol pretreatment increased cAMP response element-binding protein (CREB) phosphorylation and decreased neuroinflammation. These observations suggest that cilostazol may have beneficial therapeutic effects on seizure activity and other neurological diseases associated with excitotoxicity.
Collapse
Affiliation(s)
- Young-Seop Park
- Department of Neurosurgery, Institute of Health Sciences, Gyeongsang National University Changwon Hospital, Changwon 51472, Korea.,Department of Neurosurgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Zhen Jin
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Eun Ae Jeong
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Chin-Ok Yi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - In Sung Park
- Department of Neurosurgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 52727, Korea
| |
Collapse
|
9
|
Zhu J, Chen Z, Tian J, Meng Z, Ju M, Wu G, Tian Z. miR-34b attenuates trauma-induced anxiety-like behavior by targeting CRHR1. Int J Mol Med 2017; 40:90-100. [PMID: 28498394 PMCID: PMC5466391 DOI: 10.3892/ijmm.2017.2981] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/25/2017] [Indexed: 02/04/2023] Open
Abstract
Exposure to trauma is a potential contributor to anxiety; however, the molecular mechanisms responsible for trauma-induced anxiety require further clarification. In this study, in an aim to explore these mechanisms, we observed the changes in the hypothalamic pituitary adrenal (HPA) axis using a radioimmunoassay and the changes in anxiety-like behavior using the open field test and elevated plus maze test in a rat model following intervention with NBI-27914, a specific corticotropin-releasing hormone receptor 1 (CRHR1) antagonist. CRHR1 was found to be involved in trauma-induced anxiety. We then applied bioinformatic analysis to screen microRNAs (miRNAs or miRs) that target CRHR1, and miR-34b was determined to negatively regulate CRHR1 mRNA in primary hypothalamic neurons. The overexpression of miR-34b in the paraventricular nucleus (PVN) by a miRNA agomir using a drug delivery system decreased the hyperactivity of the HPA axis and anxiety-like behavior. Overall, the involvement of the HPA axis in trauma-induced anxiety was demonstrated, and trauma-induced anxiety was attenuated by decreasing the hyperactivity of the HPA axis via miR-34b by targeting CRHR1.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Zhejun Chen
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jinxing Tian
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Zehui Meng
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Mingda Ju
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Gencheng Wu
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Zhanzhuang Tian
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Institute of Acupuncture Research, WHO Collaborating Centre for Traditional Medicine, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
10
|
Knott EP, Assi M, Rao SNR, Ghosh M, Pearse DD. Phosphodiesterase Inhibitors as a Therapeutic Approach to Neuroprotection and Repair. Int J Mol Sci 2017; 18:E696. [PMID: 28338622 PMCID: PMC5412282 DOI: 10.3390/ijms18040696] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
A wide diversity of perturbations of the central nervous system (CNS) result in structural damage to the neuroarchitecture and cellular defects, which in turn are accompanied by neurological dysfunction and abortive endogenous neurorepair. Altering intracellular signaling pathways involved in inflammation and immune regulation, neural cell death, axon plasticity and remyelination has shown therapeutic benefit in experimental models of neurological disease and trauma. The second messengers, cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP), are two such intracellular signaling targets, the elevation of which has produced beneficial cellular effects within a range of CNS pathologies. The only known negative regulators of cyclic nucleotides are a family of enzymes called phosphodiesterases (PDEs) that hydrolyze cyclic nucleotides into adenosine monophosphate (AMP) or guanylate monophosphate (GMP). Herein, we discuss the structure and physiological function as well as the roles PDEs play in pathological processes of the diseased or injured CNS. Further we review the approaches that have been employed therapeutically in experimental paradigms to block PDE expression or activity and in turn elevate cyclic nucleotide levels to mediate neuroprotection or neurorepair as well as discuss both the translational pathway and current limitations in moving new PDE-targeted therapies to the clinic.
Collapse
Affiliation(s)
- Eric P Knott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Sudheendra N R Rao
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Neuroscience Program, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- Bruce Wayne Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
11
|
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
|
12
|
Zhang F, Fu Y, Zhou X, Pan W, Shi Y, Wang M, Zhang X, Qi D, Li L, Ma K, Tang R, Zheng K, Song Y. Depression-like behaviors and heme oxygenase-1 are regulated by Lycopene in lipopolysaccharide-induced neuroinflammation. J Neuroimmunol 2016; 298:1-8. [DOI: 10.1016/j.jneuroim.2016.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/18/2016] [Accepted: 06/04/2016] [Indexed: 12/26/2022]
|
13
|
Gamil NM, Maklad YA, Ahmed MA, Nofal S, Ahmed AA. Modulatory effect of cilostazol on tramadol-induced behavioral and neurochemical alterations in rats challenged across the forced swim despair test. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2016. [DOI: 10.1016/j.bjbas.2016.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
14
|
Kim YR, Kim HN, Hong KW, Shin HK, Choi BT. Anti-depressant effects of phosphodiesterase 3 inhibitor cilostazol in chronic mild stress-treated mice after ischemic stroke. Psychopharmacology (Berl) 2016; 233:1055-66. [PMID: 26686236 DOI: 10.1007/s00213-015-4185-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/07/2015] [Indexed: 01/07/2023]
Abstract
RATIONALE Phosphodiesterase 3 (PDE3) inhibitor cilostazol ameliorates negative effects of cerebral hypoperfusion against cerebral ischemic injury through the phosphodiesterase 3-cyclic adenosine monophosphate (cAMP) signaling cascade. OBJECTIVES We investigated the question of whether cilostazol would have an anti-depressant effect on chronic mild stress (CMS)-treated mice after ischemic stroke. METHODS An animal model of post-stroke depression was developed by additional CMS procedures in middle cerebral artery occlusion (MCAO). We performed behavioral, histological, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), immunohistochemical, Western blot and enzyme linked immunosorbent assays (ELISA). RESULTS In the open field, sucrose preference, forced swim and Morris water maze test, treatment with cilostazol resulted in reduction of all depressive behaviors examined, particularly in the Morris water maze test. Treatment with cilostazol reduced prominent atrophic changes in the ipsilateral striatum and hippocampus of CMS-treated ischemic mice through inhibition of neuronal cell death and microglial activation. In addition, treatment of the CMS-treated ischemic mice with cilostazol resulted in significantly increased phosphorylation of cAMP response element-binding protein (CREB) and expression of mature brain-derived neurotrophic factor (BDNF) with its receptor tropomyosin receptor kinase B (TrkB) in the ipsilateral striatum and hippocampus. Phosphorylation of CREB was also demonstrated in the dopaminergic neurons of the midbrain. Treatment with cilostazol also resulted in an increased number of newly formed cells and enhanced differentiation into neurons in the ipsilateral striatum and hippocampus. CONCLUSIONS Our results suggest that phosphodiesterase 3 inhibitor cilostazol may have anti-depressant effects on post-stroke depression through inhibition of neurodegeneration in the primary lesion and secondary extrafocal sites and promotion of neurogenesis. These beneficial effects on post-stroke depression may be involved in activation of CREB/BDNF signaling.
Collapse
Affiliation(s)
- Yu Ri Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea
| | - Ha Neui Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea.,Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Ki Whan Hong
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 626-870, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea.,Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, 626-870, Republic of Korea.,Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea. .,Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, 626-870, Republic of Korea. .,Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Gyeongnam, Yangsan, 626-870, Republic of Korea.
| |
Collapse
|