1
|
Liu C, Tang H, Liu C, Ma J, Liu G, Niu L, Li C, Li J. Transcutaneous auricular vagus nerve stimulation for post-stroke depression: A double-blind, randomized, placebo-controlled trial. J Affect Disord 2024; 354:82-88. [PMID: 38452937 DOI: 10.1016/j.jad.2024.03.005] [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: 12/20/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND It is a well-established fact that post-stroke depression (PSD) is a prevalent condition that affects a significant proportion of individuals who have suffered a stroke. Hence, our research endeavors to explore the safety, efficacy and the potential molecular mechanism of transcutaneous auricular vagus nerve stimulation (ta-VNS) for the treatment of depression in PSD patients by conducting a double-blind, sham-controlled, randomized trial. METHODS Patients who had experienced strokes and exhibited depressive symptoms, with a Hamilton Depression Scale (HAMD-17) score of ≥8 and met the DSM-IV criteria, were diagnosed with PSD. A volunteer sample of participants (N = 80) were randomly divided into either the ta-VNS group (which received ta-VNS in addition to conventional treatment) or the control group (which received conventional treatment only), in a 1:1 ratio. The effectiveness of the interventions was evaluated using the 17-item Hamilton Rating Scale for Depression (HAMD-17), Zung Self-Rating Depression Scale (SDS), and Barthel Index (BI) scores. Furthermore, Plasma BDNF, CREB1, and 5-HT levels were measured before and after treatment. RESULTS The concomitant application of ta-VNS demonstrated a remarkable reduction in HAMD-17 and SDS scores, leading to noteworthy enhancements in patients' daily functioning, as evidenced by improved activities of daily living, at all assessed time points, in contrast to the control group (p < 0.0001). Notably, the ta-VNS group exhibited superior effects in modulating the measured neurotrophic biomarkers when compared to the control group (p < 0.05). CONCLUSIONS The synergistic approach of combining ta-VNS with conventional treatment has demonstrated remarkable efficacy and tolerability in managing depression following a stroke.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingxi Ma
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China; Chongqing Key Laboratory of Neurodegenerative Disease, Chongqing, China
| | - Gang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingchuan Niu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changqing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiani Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Abdallah MS, Ramadan AN, Omara‐Reda H, Mansour NO, Elsokary MA, Elsawah HK, Zaki SA, Abo Mansour HE, Mosalam EM. Double-blind, randomized, placebo-controlled pilot study of the phosphodiesterase-3 inhibitor cilostazol as an adjunctive to antidepressants in patients with major depressive disorder. CNS Neurosci Ther 2021; 27:1540-1548. [PMID: 34545997 PMCID: PMC8611782 DOI: 10.1111/cns.13731] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Cilostazol (CLS) has shown antidepressant effect in cardiovascular patients, post-stroke depression, and animal models through its neurotrophic and antiinflammatory activities. Consequently, we aimed to investigate its safety and efficacy in patients with MDD by conducting double-blind, randomized, placebo-controlled pilot study. METHODS 80 participants with MDD (DSM-IV criteria) and Hamilton Depression Rating Scale (HDRS) score >20 were treated with CLS 50 mg or placebo twice daily plus escitalopram (ESC) 20 mg once daily for six weeks. Patients were evaluated by HDRS scores (weeks 0, 2, 4, and 6). Serum levels of CREB1, BDNF, 5-HT, TNF-α, NF- κB, and FAM19A5 were assessed pre- and post-treatment. RESULTS Co-administration of CLS had markedly decreased HDRS score at all-time points compared to the placebo group (p < 0.001). Early improvement, response, and remission rates after 6 weeks were significantly higher in the CLS group (90%, 90%, 80%, respectively) than in the placebo group (25%, 65%, 50% respectively) (p < 0.001). Moreover, the CLS group was superior to the placebo group in modulation of the measured neurotrophic and inflammatory biomarkers. CONCLUSION CLS is safe and effective short-term adjunctive therapy in patients with MDD with no other comorbid conditions. Trial registration ID:NCT04069819.
Collapse
Affiliation(s)
- Mahmoud S. Abdallah
- Department of Clinical PharmacyFaculty of PharmacyUniversity of Sadat CitySadat CityEgypt
| | - Ahmed N. Ramadan
- Department of NeuropsychiatryFaculty of MedicineMenoufia UniversityShebin El‐KomEgypt
| | - Hend Omara‐Reda
- Department of NeuropsychiatryFaculty of MedicineMenoufia UniversityShebin El‐KomEgypt
| | - Noha O. Mansour
- Clinical Pharmacy and Pharmacy Practice DepartmentFaculty of PharmacyMansoura UniversityMansouraEgypt
| | - Mohamed A. Elsokary
- Department of BiostatisticsHigh Institute of Public HealthAlexandria UniversityAlexandriaEgypt
| | - Hozaifa K. Elsawah
- Department of BiostatisticsHigh Institute of Public HealthAlexandria UniversityAlexandriaEgypt
| | - Shimaa Abdelsattar Zaki
- Department of Clinical Biochemistry and Molecular DiagnosticsNational Liver InstituteMenoufia UniversityShebin El‐KomEgypt
| | - Hend E. Abo Mansour
- Department of BiochemistryFaculty of PharmacyMenoufia UniversityShebin El‐KomEgypt
| | - Esraa M. Mosalam
- Department of BiochemistryFaculty of PharmacyMenoufia UniversityShebin El‐KomEgypt
| |
Collapse
|
3
|
Abbott LC, Nigussie F. Adult neurogenesis in the mammalian dentate gyrus. Anat Histol Embryol 2019; 49:3-16. [PMID: 31568602 DOI: 10.1111/ahe.12496] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/03/2019] [Accepted: 08/22/2019] [Indexed: 12/30/2022]
Abstract
Earlier observations in neuroscience suggested that no new neurons form in the mature central nervous system. Evidence now indicates that new neurons do form in the adult mammalian brain. Two regions of the mature mammalian brain generate new neurons: (a) the border of the lateral ventricles of the brain (subventricular zone) and (b) the subgranular zone (SGZ) of the dentate gyrus of the hippocampus. This review focuses only on new neuron formation in the dentate gyrus of the hippocampus. During normal prenatal and early postnatal development, neural stem cells (NSCs) give rise to differentiated neurons. NSCs persist in the dentate gyrus SGZ, undergoing cell division, with some daughter cells differentiating into functional neurons that participate in learning and memory and general cognition through integration into pre-existing neural networks. Axons, which emanate from neurons in the entorhinal cortex, synapse with dendrites of the granule cells (small neurons) of the dentate gyrus. Axons from granule cells synapse with pyramidal cells in the hippocampal CA3 region, which send axons to synapse with CA1 hippocampal pyramidal cells that send their axons out of the hippocampus proper. Adult neurogenesis includes proliferation, differentiation, migration, the death of some newly formed cells and final integration of surviving cells into neural networks. We summarise these processes in adult mammalian hippocampal neurogenesis and discuss the roles of major signalling molecules that influence neurogenesis, including neurotransmitters and some hormones. The recent controversy raised concerning whether or not adult neurogenesis occurs in humans also is discussed.
Collapse
Affiliation(s)
- Louise C Abbott
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Fikru Nigussie
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Propofol Affects Neurodegeneration and Neurogenesis by Regulation of Autophagy via Effects on Intracellular Calcium Homeostasis. Anesthesiology 2017; 127:490-501. [PMID: 28614084 DOI: 10.1097/aln.0000000000001730] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND In human cortical neural progenitor cells, we investigated the effects of propofol on calcium homeostasis in both the ryanodine and inositol 1,4,5-trisphosphate calcium release channels. We also studied propofol-mediated effects on autophagy, cell survival, and neuro- and gliogenesis. METHODS The dose-response relationship between propofol concentration and duration was studied in neural progenitor cells. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase release assays. The effects of propofol on cytosolic calcium concentration were evaluated using Fura-2, and autophagy activity was determined by LC3II expression levels with Western blot. Proliferation and differentiation were evaluated by bromodeoxyuridine incorporation and immunostaining with neuronal and glial markers. RESULTS Propofol dose- and time-dependently induced cell damage and elevated LC3II expression, most robustly at 200 µM for 24 h (67 ± 11% of control, n = 12 to 19) and 6 h (2.4 ± 0.5 compared with 0.6 ± 0.1 of control, n = 7), respectively. Treatment with 200 μM propofol also increased cytosolic calcium concentration (346 ± 71% of control, n = 22 to 34). Propofol at 10 µM stimulated neural progenitor cell proliferation and promoted neuronal cell fate, whereas propofol at 200 µM impaired neuronal proliferation and promoted glial cell fate (n = 12 to 20). Cotreatment with ryanodine and inositol 1,4,5-trisphosphate receptor antagonists and inhibitors, cytosolic Ca chelators, or autophagy inhibitors mostly mitigated the propofol-mediated effects on survival, proliferation, and differentiation. CONCLUSIONS These results suggest that propofol-mediated cell survival or neurogenesis is closely associated with propofol's effects on autophagy by activation of ryanodine and inositol 1,4,5-trisphosphate receptors.
Collapse
|
7
|
Stanslowsky N, Jahn K, Venneri A, Naujock M, Haase A, Martin U, Frieling H, Wegner F. Functional effects of cannabinoids during dopaminergic specification of human neural precursors derived from induced pluripotent stem cells. Addict Biol 2017; 22:1329-1342. [PMID: 27027565 DOI: 10.1111/adb.12394] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/22/2016] [Accepted: 02/22/2016] [Indexed: 01/28/2023]
Abstract
Among adolescents cannabis is one of the most widely used illicit drugs. In adolescence brain development continues, characterized by neuronal maturation and synaptic plasticity. The endocannabinoid system plays an important role during brain development by modulating neuronal function and neurogenesis. Changes in endocannabinoid signaling by Δ9 -tetrahydrocannabinol (THC), the psychoactive component of cannabis, might therefore lead to neurobiological changes influencing brain function and behavior. We investigated the functional maturation and dopaminergic specification of human cord blood-derived induced pluripotent stem cell (hCBiPSC)-derived small molecule neural precursor cells (smNPCs) after cultivation with the endogenous cannabinoid anandamide (AEA) and the exogenous THC, both potent agonists at the cannabinoid 1 receptor (CB1 R). Higher dosages of 10-μM AEA or THC significantly decreased functionality of neurons, indicated by reduced ion currents and synaptic activity. A lower concentration of 1-μM THC had no marked effect on neuronal and dopaminergic maturation, while 1-μM AEA significantly enhanced the frequency of synaptic activity. As there were no significant effects on DNA methylation in promotor regions of genes important for neuronal function, these cannabinoid actions seem to be mediated by another than this epigenetic mechanism. Our data suggest that there are concentration-dependent actions of cannabinoids on neuronal function in vitro indicating neurotoxic, dysfunctional effects of 10-μM AEA and THC during human neurogenesis.
Collapse
Affiliation(s)
| | - Kirsten Jahn
- Center for Addiction Research, Department of Psychiatry, Social Psychiatry and Psychotherapy; Hannover Medical School; Hannover Germany
| | - Anna Venneri
- Department of Neurology; Hannover Medical School; Hannover Germany
| | - Maximilian Naujock
- Department of Neurology; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiac, Thoracic, Transplantation and Vascular Surgery; Hannover Medical School; Hannover Germany
- REBIRTH-Cluster of Excellence; Hannover Germany
| | - Helge Frieling
- Center for Addiction Research, Department of Psychiatry, Social Psychiatry and Psychotherapy; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| | - Florian Wegner
- Department of Neurology; Hannover Medical School; Hannover Germany
- Center for Systems Neuroscience; Hannover Germany
| |
Collapse
|
8
|
Kim YR, Kim HN, Hong KW, Shin HK, Choi BT. Antidepressant Effects of Aripiprazole Augmentation for Cilostazol-Treated Mice Exposed to Chronic Mild Stress after Ischemic Stroke. Int J Mol Sci 2017; 18:ijms18020355. [PMID: 28208711 PMCID: PMC5343890 DOI: 10.3390/ijms18020355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/29/2017] [Accepted: 02/03/2017] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to determine the effects and underlying mechanism of aripiprazole (APZ) augmentation for cilostazol (CLS)-treated post-ischemic stroke mice that were exposed to chronic mild stress (CMS). Compared to treatment with either APZ or CLS alone, the combined treatment resulted in a greater reduction in depressive behaviors, including anhedonia, despair-like behaviors, and memory impairments. This treatment also significantly reduced atrophic changes in the striatum, cortex, and midbrain of CMS-treated ischemic mice, and inhibited neuronal cell apoptosis, particularly in the striatum and the dentate gyrus of the hippocampus. Greater proliferation of neuronal progenitor cells was also observed in the ipsilateral striatum of the mice receiving combined treatment compared to mice receiving either drug alone. Phosphorylation of the cyclic adenosine monophosphate response element binding protein (CREB) was increased in the striatum, hippocampus, and midbrain of mice receiving combined treatment compared to treatment with either drug alone, particularly in the neurons of the striatum and hippocampus, and dopaminergic neurons of the midbrain. Our results suggest that APZ may augment the antidepressant effects of CLS via co-regulation of the CREB signaling pathway, resulting in the synergistic enhancement of their neuroprotective effects.
Collapse
Affiliation(s)
- Yu Ri Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Korea.
| | - Ha Neui Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Korea.
| | - Ki Whan Hong
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea.
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Korea.
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Korea.
- Division of Meridian and Structural Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
9
|
Zhang T, Hong J, Di T, Chen L. MPTP Impairs Dopamine D1 Receptor-Mediated Survival of Newborn Neurons in Ventral Hippocampus to Cause Depressive-Like Behaviors in Adult Mice. Front Mol Neurosci 2016; 9:101. [PMID: 27790091 PMCID: PMC5062058 DOI: 10.3389/fnmol.2016.00101] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 09/28/2016] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by motor symptoms with depression. We evaluated the influence of dopaminergic depletion on hippocampal neurogenesis process to explore mechanisms of depression production. Five consecutive days of 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) injection in mice (MPTP-mice) reduced dopaminergic fibers in hippocampal dentate gyrus (DG). MPTP-mice exhibited depressive-like behaviors later for 2–3 weeks. BrdU was injected 4 h after last-injection of MPTP. BrdU-positive (BrdU+) cells in dorsal (d-DG) and ventral (v-DG) DG were examined on day 1 (D1), 7 (D7), 14 (D14) and 21 (D21) after BrdU injection. Fewer D7-, D14- and D21-BrdU+ cells or BrdU+/NeuN+ cells, but not D1-BrdU+ cells, were found in v-DG of MPTP-mice than in controls. However, the number of BrdU+ cells in d-DG did not differ between the both. Loss of doublecortin-positive (DCX+) cells was observed in v-DG of MPTP-mice. Protein kinase A (PKA) and Ca2+/cAMP-response element binding protein (CREB) phosphorylation were reduced in v-DG of MPTP-mice, which were reversed by D1-like receptor (D1R) agonist SKF38393, but not D2R agonist quinpirole. The treatment of MPTP-mice with SKF38393 on days 2–7 after BrdU-injection reduced the loss of D7- and D21-BrdU+ cells in v-DG and improved the depressive-like behaviors; these changes were sensitive to PKA inhibitor H89. Moreover, the v-DG injection of SKF38393 in MPTP-mice could reduce the loss of D21-BrdU+ cells and relieve the depressive-like behaviors. In control mice, the blockade of D1R by SCH23390 caused the reduction of D21-BrdU+ cells in v-DG and the depressive-like behaviors. Our results indicate that MPTP-reduced dopaminergic depletion impairs the D1R-mediated early survival of newborn neurons in v-DG, producing depressive-like behaviors.
Collapse
Affiliation(s)
- Tingting Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China; Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Juan Hong
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China; Department of Physiology, Nanjing Medical UniversityNanjing, China
| | - Tingting Di
- Department of Physiology, Nanjing Medical University Nanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical UniversityNanjing, China; Department of Physiology, Nanjing Medical UniversityNanjing, China
| |
Collapse
|
10
|
Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res 2016; 95:943-972. [PMID: 27350397 DOI: 10.1002/jnr.23777] [Citation(s) in RCA: 274] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 02/06/2023]
Abstract
Aging, hypertension, diabetes, hypoxia/obstructive sleep apnea (OSA), obesity, vitamin B12/folate deficiency, depression, and traumatic brain injury synergistically promote diverse pathological mechanisms including cerebral hypoperfusion and glucose hypometabolism. These risk factors trigger neuroinflammation and oxidative-nitrosative stress that in turn decrease nitric oxide and enhance endothelin, Amyloid-β deposition, cerebral amyloid angiopathy, and blood-brain barrier disruption. Proinflammatory cytokines, endothelin-1, and oxidative-nitrosative stress trigger several pathological feedforward and feedback loops. These upstream factors persist in the brain for decades, upregulating amyloid and tau, before the cognitive decline. These cascades lead to neuronal Ca2+ increase, neurodegeneration, cognitive/memory decline, and Alzheimer's disease (AD). However, strategies are available to attenuate cerebral hypoperfusion and glucose hypometabolism and ameliorate cognitive decline. AD is the leading cause of dementia among the elderly. There is significant evidence that pathways involving inflammation and oxidative-nitrosative stress (ONS) play a key pathophysiological role in promoting cognitive dysfunction. Aging and several comorbid conditions mentioned above promote diverse pathologies. These include inflammation, ONS, hypoperfusion, and hypometabolism in the brain. In AD, chronic cerebral hypoperfusion and glucose hypometabolism precede decades before the cognitive decline. These comorbid disease conditions may share and synergistically activate these pathophysiological pathways. Inflammation upregulates cerebrovascular pathology through proinflammatory cytokines, endothelin-1, and nitric oxide (NO). Inflammation-triggered ONS promotes long-term damage involving fatty acids, proteins, DNA, and mitochondria; these amplify and perpetuate several feedforward and feedback pathological loops. The latter includes dysfunctional energy metabolism (compromised mitochondrial ATP production), amyloid-β generation, endothelial dysfunction, and blood-brain-barrier disruption. These lead to decreased cerebral blood flow and chronic cerebral hypoperfusion- that would modulate metabolic dysfunction and neurodegeneration. In essence, hypoperfusion deprives the brain from its two paramount trophic substances, viz., oxygen and nutrients. Consequently, the brain suffers from synaptic dysfunction and neuronal degeneration/loss, leading to both gray and white matter atrophy, cognitive dysfunction, and AD. This Review underscores the importance of treating the above-mentioned comorbid disease conditions to attenuate inflammation and ONS and ameliorate decreased cerebral blood flow and hypometabolism. Additionally, several strategies are described here to control chronic hypoperfusion of the brain and enhance cognition. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mak Adam Daulatzai
- Sleep Disorders Group, EEE Dept/MSE, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
11
|
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
|
12
|
Wang Y, Fu W, Liu J. Neurodevelopment in children with intrauterine growth restriction: adverse effects and interventions. J Matern Fetal Neonatal Med 2016; 29:660-8. [PMID: 25758617 DOI: 10.3109/14767058.2015.1015417] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Intrauterine growth restriction (IUGR) is associated with higher rates of fetal, perinatal, and neonatal morbidity and mortality. The consequences of IUGR include short-term metabolic, hematological and thermal disturbances that lead to metabolic syndrome in children and adults. Additionally, IUGR severely affects short- and long-term fetal brain development and brain function (including motor, cognitive and executive function) and neurobehavior, especially neuropsychology. This review details the adverse effects of IUGR on fetal brain development and discusses intervention strategies.
Collapse
Affiliation(s)
- Yan Wang
- a Department of Neonatology and NICU , Bayi Children's Hospital, Beijing Military General Hospital , Beijing , China and
- b Department of Neonatology and NICU , Taian City Central Hospital of Shandong Province , Taian City , China
| | - Wei Fu
- a Department of Neonatology and NICU , Bayi Children's Hospital, Beijing Military General Hospital , Beijing , China and
| | - Jing Liu
- a Department of Neonatology and NICU , Bayi Children's Hospital, Beijing Military General Hospital , Beijing , China and
| |
Collapse
|
13
|
Liu XY, Gonzalez-Toledo ME, Fagan A, Duan WM, Liu Y, Zhang S, Li B, Piao CS, Nelson L, Zhao LR. Stem cell factor and granulocyte colony-stimulating factor exhibit therapeutic effects in a mouse model of CADASIL. Neurobiol Dis 2015; 73:189-203. [DOI: 10.1016/j.nbd.2014.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/27/2014] [Accepted: 09/12/2014] [Indexed: 11/28/2022] Open
|
14
|
Kwon KJ, Lee EJ, Kim MK, Kim SY, Kim JN, Kim JO, Kim HJ, Kim HY, Han JS, Shin CY, Han SH. Diabetes augments cognitive dysfunction in chronic cerebral hypoperfusion by increasing neuronal cell death: implication of cilostazol for diabetes mellitus-induced dementia. Neurobiol Dis 2014; 73:12-23. [PMID: 25281785 DOI: 10.1016/j.nbd.2014.08.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 02/06/2023] Open
Abstract
Many patients with diabetes are at increased risk of cognitive dysfunction and dementia. Diabetes mellitus is a vascular risk factor that may increase the risk of dementia through its associations with vascular dementia. We tested whether cognitive impairment could be exacerbated in combined injury using a rat model of chronic cerebral hypoperfusion with diabetes. We also determined whether a potent inhibitor of type III phosphodiesterase could prevent the cognitive decline caused by this combined injury. We used Otsuka Long-Evans Tokushima Fatty (OLETF) rats as a model of type II diabetes (T2DM) and Long-Evans Tokushima Otsuka (LETO) rats as a control. Chronic cerebral hypoperfusion was modeled by permanent bilateral common carotid artery occlusion (BCCAO). At 24weeks, the non-diabetic and T2DM rats were randomly assigned into groups for the following experiments: analysis I (1) sham non-diabetic rats (n=8); (2) hypoperfused non-diabetic rats (n=9); (3) sham T2DM rats (n=8); (4) hypoperfused T2DM rats (n=9); analysis II- (1) sham T2DM rats without treatment (n=8); (2) cilostazol-treated T2DM rats (n=8); (3) hypoperfused T2DM rats (n=9); and (4) hypoperfused T2DM rats and cilostazol treatment (n=9). The rats were orally administered cilostazol (50mg/kg) or vehicle once a day for 2weeks after 24weeks. Rats performed Morris water maze tasks, and neuronal cell death and neuroinflammation were investigated via Western blots and histological investigation. Spatial memory impairment was exacerbated synergistically in the hypoperfused T2DM group compared with the hypoperfused non-diabetic group and sham T2DBM group (P<0.05). Compared with the control group, neuronal cell death was increased in the hippocampus of the hypoperfused T2DM group. Cilostazol, a PDE-3 inhibitor, improved the memory impairments through inhibition of neuronal cell death, activation of CREB phosphorylation and BDNF expression in the hypoperfused T2DM group. Our experimental results support the hypothesis that there are deleterious interactions between chronic cerebral hypoperfusion and T2DM. That is, metabolic diseases such as diabetes may exacerbate cognitive impairment in a rat model of vascular dementia. We also suggest that surprisingly, the phosphodiesterase III inhibitor, cilostazol may be useful for the treatment of cognitive impairment in diabetes mellitus-induced dementia. In conclusion, diabetes can aggravate cognitive dysfunction in vascular dementia, and PDE-3 inhibitors, such as cilostazol, may form the basis of a novel therapeutic strategy for diabetes-associated cognitive impairment or vascular dementia.
Collapse
Affiliation(s)
- Kyoung Ja Kwon
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Eun Joo Lee
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Min Kyeong Kim
- Department of Pharmacology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Soo Young Kim
- Department of Pharmacology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Jung Nam Kim
- Department of Pharmacology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Jin Ok Kim
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea; Department of Neurology, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, 17 Heangdang-dong, Seongdong-gu, Seoul, Republic of Korea
| | - Hahn Young Kim
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea; Department of Neurology, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Jung-Soo Han
- Department of Biological Sciences, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Chan Young Shin
- Department of Pharmacology, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea
| | - Seol-Heui Han
- Department of Neuroscience, Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Republic of Korea; Department of Neurology, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
15
|
Fitzsimons CP, van Bodegraven E, Schouten M, Lardenoije R, Kompotis K, Kenis G, van den Hurk M, Boks MP, Biojone C, Joca S, Steinbusch HWM, Lunnon K, Mastroeni DF, Mill J, Lucassen PJ, Coleman PD, van den Hove DLA, Rutten BPF. Epigenetic regulation of adult neural stem cells: implications for Alzheimer's disease. Mol Neurodegener 2014; 9:25. [PMID: 24964731 PMCID: PMC4080757 DOI: 10.1186/1750-1326-9-25] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/06/2014] [Indexed: 01/27/2023] Open
Abstract
Experimental evidence has demonstrated that several aspects of adult neural stem cells (NSCs), including their quiescence, proliferation, fate specification and differentiation, are regulated by epigenetic mechanisms. These control the expression of specific sets of genes, often including those encoding for small non-coding RNAs, indicating a complex interplay between various epigenetic factors and cellular functions.Previous studies had indicated that in addition to the neuropathology in Alzheimer's disease (AD), plasticity-related changes are observed in brain areas with ongoing neurogenesis, like the hippocampus and subventricular zone. Given the role of stem cells e.g. in hippocampal functions like cognition, and given their potential for brain repair, we here review the epigenetic mechanisms relevant for NSCs and AD etiology. Understanding the molecular mechanisms involved in the epigenetic regulation of adult NSCs will advance our knowledge on the role of adult neurogenesis in degeneration and possibly regeneration in the AD brain.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Emma van Bodegraven
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Marijn Schouten
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Roy Lardenoije
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Konstantinos Kompotis
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Mark van den Hurk
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Marco P Boks
- Department Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Biojone
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Samia Joca
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Harry WM Steinbusch
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Katie Lunnon
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Diego F Mastroeni
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Jonathan Mill
- University of Exeter Medical School, RILD Level 4, Barrack Road, University of Exeter, Devon, UK
| | - Paul J Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Paul D Coleman
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands
| | - Daniel LA van den Hove
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
| | - Bart PF Rutten
- Department of Translational Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
16
|
Tomimoto H, Wakita H. Animal models of vascular dementia: translational potential at the present time and in 2050. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.71] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: Vascular dementia is a heterogeneous syndrome, and includes subcortical ischemic vascular dementia. For translational research, subcortical ischemic vascular dementia is an appropriate target since this is the most prevalent subtype and exhibits relatively uniform clinical and neuropathological changes. These changes consist of hypertensive arteriolar changes, lacunar infarctions, hypertensive hemorrhage and white matter lesions. Among various species, rodents are most frequently used, but their small volume of white matter may impede analysis of white matter lesions. Primate models have a larger volume, but the degree of white matter lesions is inconsistent. Animal models should accommodate the effect of aging and comorbidities, and in the case of primate models, low accessibility should be overcome by repeated and quantitative examinations using modern neuroimaging techniques and functional measures, especially for memory and motor function. There is no model that replicates all features of subcortical ischemic vascular dementia and, therefore, rodent and primate models should be selected appropriately for translational research.
Collapse
Affiliation(s)
- Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Edobashi 2-174, Tsu City 514-8507, Japan
| | - Hideaki Wakita
- Department of Internal Medicine, Nanakuri Sanatorium, Fujita Health University, Otoricho 424-1, Tsu City 514-12957, Japan
| |
Collapse
|
17
|
Liu XY, Gonzalez-Toledo ME, Fagan A, Duan WM, Liu Y, Zhang S, Li B, Piao CS, Nelson L, Zhao LR. Novel pathological features and potential therapeutic approaches for CADASIL: insights obtained from a mouse model of CADASIL. THERAPEUTIC TARGETS FOR NEUROLOGICAL DISEASES 2014; 1. [PMID: 30090853 DOI: 10.14800/ttnd.434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common condition of hereditary stroke and vascular dementia. CADASIL is caused by Notch3 mutation, leading to progressive degeneration of vascular smooth muscle cells (vSMCs) of the small arteries in the brain. However, the pathogenesis of CADASIL remains largely unknown, and treatment that can stop or delay the progression of CADASIL is not yet available. Using both wild type mice and transgenic mice carrying the human mutant Notch3 gene (CADASIL mice), we have recently characterized the pathological features of CADASIL and determined the therapeutic efficacy of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) in CADASIL. Our findings have revealed novel pathological changes in the endothelium of cerebral capillaries and in the neural stem cells (NSCs). We have also observed the impairment of cognitive function in CADASIL mice. Moreover, SCF+G-CSF treatment improves cognitive function, inhibits Notch3 mutation-induced vSMC degeneration, cerebral blood bed reduction, cerebral capillary damage, and NSC loss, and increases neurogenesis and angiogenesis. Here we compile an overview of our recently published studies, which provide new insights into understanding the pathogenesis of CADASIL and developing therapeutic strategies for this devastating neurological disease.
Collapse
Affiliation(s)
- Xiao-Yun Liu
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Maria E Gonzalez-Toledo
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Austin Fagan
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Wei-Ming Duan
- Department of Anatomy, Capital Medical University, Beijing 100069, China; Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Yanying Liu
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, New York, 13210, USA
| | - Siyuan Zhang
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Bin Li
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Chun-Shu Piao
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Lila Nelson
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA
| | - Li-Ru Zhao
- Departments of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA.,Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, Louisiana 71130, USA.,Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, New York, 13210, USA.,Department of Neurobiology, Capital Medical University, Beijing 100069, China, Beijing 100069, China
| |
Collapse
|
18
|
Ahmed Z. Cannabinoids: Do they have the potential to treat the symptoms of multiple sclerosis? World J Neurol 2013; 3:87-96. [DOI: 10.5316/wjn.v3.i4.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/16/2013] [Indexed: 02/06/2023] Open
Abstract
This article reviews the role of cannabinoids in inhibiting neurodegeneration in models of multiple sclerosis (MS). MS is a chronic, debilitating disease of the central nervous system (CNS), induced by autoimmunity-driven inflammation that leads to demyelination and thus disconnection of the normal transmission of nerve impulses. Despite the use of an array of immune modulating drugs that restore blood brain barrier function, disability continues in patients concomitant with the loss of axons in the spinal cord. MS patients therefore suffer neuropathic pain, spasticity and tremor. Anecdotal evidence suggests that MS patients using cannabis, though illegal, achieve symptomatic relief from neuropathic pain and spasticity associated with MS. The discovery of the endogenous cannabinoid (endocannabinoid) system that naturally exists in the body and which responds to cannabinoids to exert their effects has aided research into the therapeutic utility of cannabinoids. The endocannabinoid system consists of two G-protein coupled receptors cannabinoid receptor type-1 (CB1) and CB2. CB1 is mainly expressed in the CNS and CB2 is predominantly found in leukocytes, while an increasing number of potential ligands and endocannabinoid degradation molecules are being isolated. Several studies have highlighted the involvement of this system in regulating neurotransmission and its ability to prevent excessive neurotransmitter release, consistent with a capacity to provide symptomatic relief. In summary, antagonism of the CB1 receptor pathway contributes to neuronal damage in chronic relapsing experimental allergic encephalomyelitis (EAE) and suppresses tremor and spasticity. The addition of exogenous CB1 agonists derived from cannabis also afforded significant neuroprotection from the consequences of inflammatory CNS disease in EAE and experimental allergic uveitis models. Although clear neuroprotective benefits of cannabinoids have been demonstrated, the unwanted psychotropic effects need to be addressed. However, manipulating the endogenous cannabinoid system may be one way of eliciting beneficial effects without some or all of the unwanted side effects.
Collapse
|
19
|
Liu J, Liu Y, Wang XF, Chen H, Yang N. Antenatal taurine supplementation improves cerebral neurogenesis in fetal rats with intrauterine growth restriction through the PKA-CREB signal pathway. Nutr Neurosci 2013; 16:282-7. [PMID: 23433181 DOI: 10.1179/1476830513y.0000000057] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study seeks to explore whether antenatal supplement of taurine may improve the brain development of fetal rats with intrauterine growth restriction (IUGR) via the protein kinase A-cyclic adenosine monophosphate (cAMP) response element protein (PKA-CREB) pathway. METHODS Fifteen pregnant rats were randomly divided into control group, IUGR model, and IUGR with antenatal taurine supplement group. Brain tissues were obtained immediately after rats were born. PKA-CREB signal pathway activity and glial cell line-derived neurotrophic factor (GDNF) mRNA and protein levels were measured by reverse transcription polymerase chain reaction and immunohistochemistry stains, whereas immunoreactive cells of neuron-specific enolase (NSE) and proliferating cell nuclear antigen (PCNA) were detected by immunohistochemistry stains. RESULTS The results showed that: (1) In the IUGR group, a greater number of PCNA and NSE immunoreactive cells were found in brain tissues compared with controls, and prenatal taurine supplementation led to a further increase. (2) Expression of PKA, CREB, and GDNF were increased in mRNA and protein levels due to taurine supplementation. DISCUSSION Antenatal taurine supplementation shows effects of promotion of cell proliferation and activation of neurotrophic factors on fetal rat brain in a model of IUGR by activating the PKA-CREB signal pathway, increasing expression of neurotrophic factors, and promoting cell proliferation to counteract neuron loss caused by IUGR.
Collapse
Affiliation(s)
- Jing Liu
- General Hospital of Beijing Military Command, Beijing 100700, China
| | | | | | | | | |
Collapse
|
20
|
Abe A, Nishiyama Y, Hagiwara H, Okubo S, Ueda M, Katsura KI, Katayama Y. Administration of cilostazol, an antiplatelet, to patients with acute-stage cerebral infarction and its effects on plasma substance P level and latent time of swallowing reflex. J NIPPON MED SCH 2013; 80:50-6. [PMID: 23470806 DOI: 10.1272/jnms.80.50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND OBJECTIVE It has been reported that medical treatment with cilostazol (cilo) as an antiplatelet may increase a substance P level in the striatum to shorten the latent time of swallowing reflex (LTSR). We undertook a pilot study to confirm whether cilo administration to patients with cerebral infarction is effective in increasing their plasma substance P level and then in ameliorating the status of LTSR. METHODS AND SUBJECTS Eligible subjects were recruited, after informed consents, from 20 hospitalized patients with acute-phase cerebral infarction within 72 hours from the onset. At the start of treatment, the subjects were assigned at random to those given aspirin alone (non-cilo group) and those given aspirin plus cilo (cilo group). Plasma substance P levels and LTSR values were measured at the starting point (baseline), 28 days after, and 180 days after. RESULTS AND DISCUSSION No significant time-dependent change in plasma substance P level was found probably because of large individual differences but, 28 days after the start of treatment, this value tended to become higher in cilo group than in non-cilo group (P<0.10). Whereas, in terms of fold changes of LTSR in cilo group, there was a significant between-term difference at P<0.05, indicating that this medication is effective in ameliorating the swallowing function is improved in the long run. CONCLUSION The LTSR values was significantly shortened within 180 days after the start of cilo treatment, but the result was not well explained by substance P levels as far as these were measured using the plasma, probably because this substance had diluted during blood circulation. However, it will become clinically usable as a single swallowing index, if in the future some ingeneus method of its measurement is developed. A larger-scale study would also be needed to confirm our conclusion from this pilot study.
Collapse
Affiliation(s)
- Arata Abe
- Divisions of Neurology, Nephrology, and Rheumatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
21
|
Culver JC, Vadakkan TJ, Dickinson ME. A specialized microvascular domain in the mouse neural stem cell niche. PLoS One 2013; 8:e53546. [PMID: 23308251 PMCID: PMC3538546 DOI: 10.1371/journal.pone.0053546] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 12/03/2012] [Indexed: 12/27/2022] Open
Abstract
The microenvironment of the subependymal zone (SEZ) neural stem cell niche is necessary for regulating adult neurogenesis. In particular, signaling from the microvasculature is essential for adult neural stem cell maintenance, but microvascular structure and blood flow dynamics in the SEZ are not well understood. In this work, we show that the mouse SEZ constitutes a specialized microvascular domain defined by unique vessel architecture and reduced rates of blood flow. Additionally, we demonstrate that hypoxic conditions are detectable in the ependymal layer that lines the ventricle, and in a subpopulation of neurons throughout the SEZ and striatum. Together, these data highlight previously unidentified features of the SEZ neural stem cell niche, and further demonstrate the extent of microvascular specialization in the SEZ microenvironment.
Collapse
Affiliation(s)
- James C. Culver
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tegy J. Vadakkan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mary E. Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
22
|
Pimentel-Coelho PM, Rivest S. The early contribution of cerebrovascular factors to the pathogenesis of Alzheimer’s disease. Eur J Neurosci 2012; 35:1917-37. [DOI: 10.1111/j.1460-9568.2012.08126.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
23
|
Gowran A, Noonan J, Campbell VA. The multiplicity of action of cannabinoids: implications for treating neurodegeneration. CNS Neurosci Ther 2011; 17:637-44. [PMID: 20875047 PMCID: PMC6493861 DOI: 10.1111/j.1755-5949.2010.00195.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cannabinoid (CB) system is widespread in the central nervous system and is crucial for controlling a range of neurophysiological processes such as pain, appetite, and cognition. The endogenous CB molecules, anandamide, and 2-arachidonoyl glycerol, interact with the G-protein coupled CB receptors, CB(1) and CB(2). These receptors are also targets for the phytocannabinoids isolated from the cannabis plant and synthetic CB receptor ligands. The CB system is emerging as a key regulator of neuronal cell fate and is capable of conferring neuroprotection by the direct engagement of prosurvival pathways and the control of neurogenesis. Many neurological conditions feature a neurodegenerative component that is associated with excitotoxicity, oxidative stress, and neuroinflammation, and certain CB molecules have been demonstrated to inhibit these events to halt the progression of neurodegeneration. Such properties are attractive in the development of new strategies to treat neurodegenerative conditions of diverse etiology, such as Alzheimer's disease, multiple sclerosis, and cerebral ischemia. This article will discuss the experimental and clinical evidence supporting a potential role for CB-based therapies in the treatment of certain neurological diseases that feature a neurodegenerative component.
Collapse
MESH Headings
- Aged
- Alzheimer Disease/drug therapy
- Alzheimer Disease/metabolism
- Alzheimer Disease/psychology
- Brain Ischemia/drug therapy
- Cannabinoids/pharmacology
- Cannabinoids/therapeutic use
- Humans
- Huntington Disease/drug therapy
- Multiple Sclerosis/drug therapy
- Neurodegenerative Diseases/drug therapy
- Parkinson Disease/drug therapy
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/physiology
- Receptor, Cannabinoid, CB2/drug effects
- Receptor, Cannabinoid, CB2/physiology
- Receptors, Cannabinoid/drug effects
- Receptors, Cannabinoid/physiology
Collapse
Affiliation(s)
- Aoife Gowran
- Department of Physiology, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
24
|
Hiramatsu M, Takiguchi O, Nishiyama A, Mori H. Cilostazol prevents amyloid β peptide(25-35)-induced memory impairment and oxidative stress in mice. Br J Pharmacol 2011; 161:1899-912. [PMID: 20825411 DOI: 10.1111/j.1476-5381.2010.01014.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Cilostazol may be effective in dementia associated with a cerebral ischaemia. In this study, we examined whether it exerts beneficial effects on learning and/or memory impairment induced by Aβ(25-35) in mice, and compared its effects with those of aspirin. EXPERIMENTAL APPROACH Aβ(25-35) (9 nmol) was administered to mice i.c.v. Learning and memory behaviour were evaluated by measuring spontaneous alternation in a Y-maze and a step-down type passive avoidance test, on the 5th and 8th days after injection respectively. Levels of lipid peroxidation (malondialdehyde) and cytokines in the frontal cortex and hippocampus were measured 2, 3, 5 and 7 days after the Aβ(25-35) injection. The effects of repeated administration of cilostazol and aspirin (both at 30 and 100 mg·kg(-1), p.o.) on any changes induced by Aβ(25-35) were evaluated. KEY RESULTS Repeated administration of cilostazol significantly attenuated the impairment of spontaneous alternation and the shortened step-down latency induced by Aβ(25-35) . Aspirin did not show any beneficial effect. A significant increase in the levels of malondialdehyde (MDA) and IL-1β (only measured in hippocampus) was observed 2, 3 and 5 days after the Aβ(25-35) injection in the frontal cortex and hippocampus. Repeated administration of cilostazol (100 mg·kg(-1)) completely prevented the increase in MDA levels but failed to antagonize the increase in the expression of IL-1β induced by Aβ(25-35). CONCLUSIONS AND IMPLICATIONS These results suggest that the protective effect of cilostazol on Aβ(25-35)-induced memory impairment may be related to oxidative stress in the frontal cortex and the hippocampus.
Collapse
Affiliation(s)
- Masayuki Hiramatsu
- Laboratory of Neuropsychopharmacology, Graduate School of Environmental and Human Sciences, Meijo University, Nagoya, Japan.
| | | | | | | |
Collapse
|
25
|
Gampe K, Brill MS, Momma S, Götz M, Zimmermann H. EGF induces CREB and ERK activation at the wall of the mouse lateral ventricles. Brain Res 2011; 1376:31-41. [PMID: 21081118 DOI: 10.1016/j.brainres.2010.11.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/20/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
Abstract
The subependymal zone at the lateral ventricular wall represents a major neurogenic niche of the adult mammalian brain and continuously provides new neurons for the olfactory bulb. A mosaic of stem and progenitor cells in this niche has the potential to respond to multiple signals including growth factors such as EGF. Recent studies using long-term ventricular infusion of EGF demonstrate intense cell proliferation around the ventricular wall, implicating the presence of EGF-reactive cells also outside the classical neurogenic lateral niche. Here we show that intraventricular injection of EGF induces within minutes CREB and ERK phosphorylation in astrocyte-like progenitor cells (type B cells) and EGF receptor-expressing transit-amplifying progenitor cells-both in the striatal and septal ventricular walls. EGF infusion for 6 days induced continued CREB and ERK activation in nestin+ cells paralleled by intense periventricular cell proliferation. In addition, the ependyma became EGF receptor-immunoreactive, revealed intense CREB phosphorylation and underwent partial de-differentiation. Our results demonstrate that intraventricular application of EGF induces CREB and ERK phosphorylation along the entire ventricular walls and thus permits a direct identification of EGF-responsive cell types. They further support the notion that not only the striatal ventricular wall where the SEZ is located but also the septal ventricular wall carries latent potential for the formation of neurons and glial cells.
Collapse
Affiliation(s)
- Kristine Gampe
- Institute of Cell Biology and Neuroscience, Biocenter, Goethe-University, 60438 Frankfurt, Germany
| | | | | | | | | |
Collapse
|
26
|
Tanaka Y, Tanaka R, Liu M, Hattori N, Urabe T. Cilostazol attenuates ischemic brain injury and enhances neurogenesis in the subventricular zone of adult mice after transient focal cerebral ischemia. Neuroscience 2010; 171:1367-76. [PMID: 20933581 DOI: 10.1016/j.neuroscience.2010.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/29/2010] [Accepted: 10/02/2010] [Indexed: 11/30/2022]
Abstract
Evidence suggests that neurogenesis occurs in the adult mammalian brain, and that various stimuli, for example, ischemia/hypoxia, enhance the generation of neural progenitor cells in the subventricular zone (SVZ) and their migration into the olfactory bulb. In a mouse stroke model, focal ischemia results in activation of neural progenitor cells followed by their migration into the ischemic lesion. The present study assessed the in vivo effects of cilostazol, a type 3 phosphodiesterase inhibitor known to activate the cAMP-responsive element binding protein (CREB) signaling, on neurogenesis in the ipsilateral SVZ and peri-infarct area in a mouse model of transient middle cerebral artery occlusion. Mice were divided into sham operated (n=12), vehicle- (n=18) and cilostazol-treated (n=18) groups. Sections stained for 5-bromodeoxyuridine (BrdU) and several neuronal and a glial markers were analyzed at post-ischemia days 1, 3 and 7. Cilostazol reduced brain ischemic volume (P<0.05) and induced earlier recovery of neurologic deficit (P<0.05). Cilostazol significantly increased the density of BrdU-positive newly-formed cells in the SVZ compared with the vehicle group without ischemia. Increased density of doublecortin (DCX)-positive and BrdU/DCX-double positive neural progenitor cells was noted in the ipsilateral SVZ and peri-infarct area at 3 and 7 days after focal ischemia compared with the vehicle group (P<0.05). Cilostazol increased DCX-positive phosphorylated CREB (pCREB)-expressing neural progenitor cells, and increased brain derived neurotrophic factor (BDNF)-expressing astrocytes in the ipsilateral SVZ and peri-infarct area. The results indicated that cilostazol enhanced neural progenitor cell generation in both ipsilateral SVZ and peri-infarct area through CREB-mediated signaling pathway after focal ischemia.
Collapse
Affiliation(s)
- Y Tanaka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
27
|
Culver JC, Dickinson ME. The effects of hemodynamic force on embryonic development. Microcirculation 2010; 17:164-78. [PMID: 20374481 DOI: 10.1111/j.1549-8719.2010.00025.x] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Blood vessels have long been known to respond to hemodynamic force, and several mechanotransduction pathways have been identified. However, only recently have we begun to understand the effects of hemodynamic force on embryonic development. In this review, we will discuss specific examples illustrating the role of hemodynamic force during the development of the embryo, with particular focus on the development of the vascular system and the morphogenesis of the heart. We will also discuss the important functions served by mechanotransduction and hemodynamic force during placentation, as well as in regulating the maintenance and division of embryonic, hematopoietic, neural, and mesenchymal stem cells. Pathological misregulation of mechanosensitive pathways during pregnancy and embryonic development may contribute to the occurrence of cardiovascular birth defects, as well as to a variety of other diseases, including preeclampsia. Thus, there is a need for future studies focusing on better understanding the physiological effects of hemodynamic force during embryonic development and their role in the pathogenesis of disease.
Collapse
Affiliation(s)
- James C Culver
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|