1
|
Cai M, Qu Y, Ren Z, Xu X, Ye C, Lu H, Zhang Y, Pan W, Shen H, Li H. Nutritional supplements formulated to prevent cognitive impairment in animals. Curr Res Food Sci 2022; 5:2294-2308. [DOI: 10.1016/j.crfs.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/08/2022] [Accepted: 11/02/2022] [Indexed: 11/20/2022] Open
|
2
|
Gong L, Yin J, Zhang Y, Huang R, Lou Y, Jiang H, Sun L, Jia J, Zeng X. Neuroprotective Mechanisms of Ginsenoside Rb1 in Central Nervous System Diseases. Front Pharmacol 2022; 13:914352. [PMID: 35721176 PMCID: PMC9201244 DOI: 10.3389/fphar.2022.914352] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Panax ginseng and Panax notoginseng, two well-known herbs with enormous medical value in Asian countries, have a long usage history in China for the therapy of some diseases, such as stroke. Ginsenoside Rb1 is one of most important active ingredients in Panax ginseng and Panax notoginseng. In the last two decades, more attention has focused on ginsenoside Rb1 as an antioxidative, anti-apoptotic and anti-inflammatory agent that can protect the nervous system. In the review, we summarize the neuroprotective roles of ginsenoside Rb1 and its potential mechanisms in central nervous system diseases (CNSDs), including neurodegenerative diseases, cerebral ischemia injury, depression and spinal cord injury. In conclusion, ginsenoside Rb1 has a potential neuroprotection due to its inhibition of oxidative stress, apoptosis, neuroinflammation and autophagy in CNSDs and may be a promising candidate agent for clinical therapy of CNSDs in the future.
Collapse
Affiliation(s)
- Liang Gong
- Jiaxing University Medical College, Jiaxing, China
| | - Jiayi Yin
- Jiaxing University Medical College, Jiaxing, China
| | - Yu Zhang
- Jiaxing University Medical College, Jiaxing, China
| | - Ren Huang
- Jiaxing University Medical College, Jiaxing, China
| | - Yuxuan Lou
- Jiaxing University Medical College, Jiaxing, China
| | - Haojie Jiang
- Jiaxing University Medical College, Jiaxing, China
| | - Liyan Sun
- Department of Clinical Medicine, Jiaxing University Medical College, Jiaxing, China
| | - Jinjing Jia
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Xiansi Zeng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| |
Collapse
|
3
|
Zarneshan SN, Fakhri S, Khan H. Targeting Akt/CREB/BDNF signaling pathway by ginsenosides in neurodegenerative diseases: A mechanistic approach. Pharmacol Res 2022; 177:106099. [DOI: 10.1016/j.phrs.2022.106099] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/14/2022] [Accepted: 01/23/2022] [Indexed: 12/15/2022]
|
4
|
Ginsenoside Rb1 exerts antidepressant-like effects via suppression inflammation and activation of AKT pathway. Neurosci Lett 2020; 744:135561. [PMID: 33359924 DOI: 10.1016/j.neulet.2020.135561] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/11/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022]
Abstract
Depression-like behaviors caused by chronic stress are related to inflammation and microglia activation. Antidepressant therapy may contribute to inhibiting inflammation responses and microglia activation. Ginsenoside Rb1 (GRb1) is known to display antidepressant-like effect on chronic unpredictable mild stress-induced depressive rats. However, the antidepressant-like effects of GRb1 on chronic restraint stress (CRS) mice and the potential anti-inflammatory mechanisms are unclear. Here, we focused on the molecular mechanisms related to inhibition of inflammation response and the protection on microglia. Our results showed that GRb1 had an antidepressant effects via relieving the depression-like behaviors in CRS model. Furthermore, GRb1 increased the protein expressions of brain-derived neurotrophic factor and phospho- protein kinase B/ protein kinase B (p-AKT/AKT), and decreased the protein expressions of interleukin-1β (IL-1β), tumor necrosis factor α (TNF-α) and ionized calcium binding adapter molecule 1 in hippocampus, reduced the levels IL-1β and TNF-α in serum. Finally, GRb1 lowered the protein expressions of IL-1β and TNF-α in BV-2 microglia induced by lipopolysaccharides. Taken together, the results indicate that GRb1 prevents CRS-induced depression-like behaviors in mice, which may be related to anti-inflammatory effects in hippocampus, serum and microglia and activation of AKT pathway.
Collapse
|
5
|
Taurine and Ginsenoside Rf Induce BDNF Expression in SH-SY5Y Cells: A Potential Role of BDNF in Corticosterone-Triggered Cellular Damage. Molecules 2020; 25:molecules25122819. [PMID: 32570881 PMCID: PMC7356094 DOI: 10.3390/molecules25122819] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 11/17/2022] Open
Abstract
This study shows that taurine and ginsenoside Rf act synergistically to increase the expression of brain-derived neurotrophic factor (BDNF) in SH-SY5Y human neuroblastoma cells in a dose- and time-dependent manner. The increase of BDNF mRNA by taurine and ginsenoside Rf was markedly attenuated by inhibitors of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase. In addition, taurine and ginsenoside Rf protected cells from corticosterone-induced BDNF suppression and reduced cell viability and lactate dehydrogenase release. The results from this study showed that combined treatment with both taurine and ginsenoside Rf enhanced BDNF expression and protected cells against corticosterone-induced damage.
Collapse
|
6
|
Qu S, Meng X, Liu Y, Zhang X, Zhang Y. Ginsenoside Rb1 prevents MPTP-induced changes in hippocampal memory via regulation of the α-synuclein/PSD-95 pathway. Aging (Albany NY) 2020; 11:1934-1964. [PMID: 30958793 PMCID: PMC6503885 DOI: 10.18632/aging.101884] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
Memory deficiency is a common non-motor symptom of Parkinson’s disease (PD), and conventionally, α-synuclein is considered to be an important biomarker for both motor and cognitive characteristics attributed to PD. However, the role of physiological α-synuclein in cognitive impairment remains undetermined. Ginsenoside Rb1 has been shown to protect dopaminergic neurons (DA) from death and inhibit α-synuclein fibrillation and toxicity in vitro. Our recent study also revealed that ginsenoside Rb1 ameliorates motor deficits and prevents DA neuron death via upregulating glutamate transporter GLT-1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Whether Rb1 can improve memory deficiency and the underlying mechanism is still unknown. In this study, we found that Rb1 can prevent the spatial learning and memory deficits, increase long-term potentiation (LTP) and hippocampal glutamatergic transmission in the MPTP mouse model. The underlying neuroprotective mechanism of Rb1-improved synaptic plasticity involves Rb1 promoting hippocampal CA3 α-synuclein expression, restoring the glutamate in the CA3-schaffer collateral-CA1 pathway, and sequentially increasing postsynaptic density-95 (PSD-95) expression. Thus, we provide evidence that Rb1 modulates memory function, synaptic plasticity, and excitatory transmission via the trans-synaptic α-synuclein/PSD-95 pathway. Our findings suggest that Rb1 may serve as a functional drug in treating the memory deficiency in PD.
Collapse
Affiliation(s)
- Shaogang Qu
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Xingjun Meng
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, China
| | - Yan Liu
- Department of Traditional Chinese Medicine, Medical College, Xiamen University, Xiamen, China
| | - Xiuping Zhang
- Teaching Center of Experimental Medicine, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yunlong Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Shenzhen Research Institute of Xiamen University, Shenzhen, China
| |
Collapse
|
7
|
Korean Red Ginseng reduces chronic social defeat stress-induced mood disorders via N-methyl-D-aspartate receptor modulation in mice. J Ginseng Res 2019; 45:254-263. [PMID: 33841006 PMCID: PMC8020286 DOI: 10.1016/j.jgr.2019.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 09/11/2019] [Accepted: 11/01/2019] [Indexed: 01/17/2023] Open
Abstract
Background A chronic social defeat stress (CSDS) model has been proposed as relevant to stress-induced behavioral change in humans. In this study, we examined the effect of Korean Red Ginseng (KRG) on CSDS-induced mood disorders and protein expression in an animal model. Methods To evaluate the effect of KRG on social defeat stress, test mice were exposed in the resident aggressor's home cage compartment for 14 days beginning 1 h after KRG treatment (10, 20, and 40 mg/kg, per oral (p.o.)). After the exposure, behavioral tests to measure anxiety, social interaction, and depression-like behavior were performed. To investigate the underlying mechanism, N-methyl-D-aspartate receptor expression levels in CSDS-induced mice were evaluated using Western blot analysis. Results CSDS induced anxiety-like behaviors by decreasing central activity in the open-field test and open-arm approach in the elevated plus maze test and led to social avoidance behavior in the social interaction test. CSDS mice showed upregulated NR1, NR2A, and NR2B expression in the hippocampus. KRG 20 and 40 mg/kg ameliorated anxiety-like activities and KRG 20 mg/kg alleviated social avoidance by decreasing time in the corner zone. KRG treatment recovered CSDS-induced NR1, NR2A, and NR2B protein levels in the hippocampus. Conclusion These results indicate that KRG has a therapeutic effect on CSDS-induced mood disorder by alleviating N-methyl-D-aspartate receptor overexpression in the hippocampus.
Collapse
|
8
|
Gessi S, Merighi S, Bencivenni S, Battistello E, Vincenzi F, Setti S, Cadossi M, Borea PA, Cadossi R, Varani K. Pulsed electromagnetic field and relief of hypoxia-induced neuronal cell death: The signaling pathway. J Cell Physiol 2019; 234:15089-15097. [PMID: 30656694 DOI: 10.1002/jcp.28149] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/02/2019] [Indexed: 01/24/2023]
Abstract
Low-energy low-frequency pulsed electromagnetic fields (PEMFs) exert several protective effects, such as the regulation of kinases, transcription factors as well as cell viability in both central and peripheral biological systems. However, it is not clear on which bases they affect neuroprotection and the mechanism responsible is yet unknown. In this study, we have characterized in nerve growth factor-differentiated pheochromocytoma PC12 cells injured with hypoxia: (i) the effects of PEMF exposure on cell vitality; (ii) the protective pathways activated by PEMFs to relief neuronal cell death, including adenylyl cyclase, phospholipase C, protein kinase C epsilon and delta, p38, ERK1/2, JNK1/2 mitogen-activated protein kinases, Akt and caspase-3; (iii) the regulation by PEMFs of prosurvival heat-shock proteins of 70 (HSP70), cAMP response element-binding protein (CREB), brain-derived neurotrophic factor (BDNF), and Bcl-2 family proteins. The results obtained in this study show a protective effect of PEMFs that are able to reduce neuronal cell death induced by hypoxia by modulating p38, HSP70, CREB, BDNF, and Bcl-2 family proteins. Specifically, we found a rapid activation (30 min) of p38 kinase cascade, which in turns enrolles HSP70 survival chaperone molecule, resulting in a significant CREB phosphorylation increase (24 hr). In this cascade, later (48 hr), BDNF and the antiapoptotic pathway regulated by the Bcl-2 family of proteins are recruited by PEMFs to enhance neuronal survival. This study paves the way to elucidate the mechanisms triggered by PEMFs to act as a new neuroprotective approach to treat cerebral ischemia by reducing neuronal cell death.
Collapse
Affiliation(s)
- Stefania Gessi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Stefania Merighi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Serena Bencivenni
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Fabrizio Vincenzi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | | | - Pier Andrea Borea
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Katia Varani
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,University Center for Studies on Gender Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
9
|
Huang X, Li N, Pu Y, Zhang T, Wang B. Neuroprotective Effects of Ginseng Phytochemicals: Recent Perspectives. Molecules 2019; 24:E2939. [PMID: 31416121 PMCID: PMC6720911 DOI: 10.3390/molecules24162939] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
As our global population ages, the treatment of neurodegenerative diseases is critical to our society. In recent years, researchers have begun to study the role of biologically active chemicals from plants and herbs to gain new inspiration and develop new therapeutic drugs. Ginseng (Panax ginseng C.A. Mey.) is a famous Chinese herbal medicine with a variety of pharmacological activities. It has been used to treat various diseases since ancient times. Extensive research over the years has shown that ginseng has potential as a neuroprotective drug, and its neuroprotective effects can be used to treat and prevent neurological damage or pathologically related diseases (such as Alzheimer's disease, Parkinson's disease, Huntington's disease, depression symptoms, and strokes). Moreover, evidence for the medicinal and health benefits of ginsenoside, its main active ingredient, in the prevention of neurodegenerative diseases is increasing, and current clinical results have not reported any serious adverse reactions to ginseng. Therefore, we briefly review the recent research and development on the beneficial effects and mechanisms of ginseng and its main active ingredient, ginsenoside, in the prevention and treatment of neurodegenerative diseases, hoping to provide some ideas for the discovery and identification of ginseng neuroprotection.
Collapse
Affiliation(s)
- Xing Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ning Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Research Institute of KPC Pharmaceuticals, Inc., Kunming 650106, China
| | - Yiqiong Pu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
10
|
Husni A, Lailatussifa R, Isnansetyo A. Sargassum hystrix as a Source of Functional Food to Improve Blood Biochemistry Profiles of Rats under Stress. Prev Nutr Food Sci 2019; 24:150-158. [PMID: 31328119 PMCID: PMC6615359 DOI: 10.3746/pnf.2019.24.2.150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/22/2019] [Indexed: 02/05/2023] Open
Abstract
This study was conducted to determine the influence of Sargassum hystrix powder (SHP) as an alternative source of functional food for treating in vivo stress by measuring levels of glucose, triacylglycerol, total cholesterol, and cortisol, and liver histopathology. Wistar rats aged 3 months and weighing 150~200 g were divided into 7 groups: normal control, fasting control, negative control (stress without adaptogen), and 4 experimental conditions (stress+0.18 mg/kg diazepam, stress+450 mg/kg pellet, stress+mixture of pellet with SHP 450 mg/kg, and stress+450 mg/kg of SHP). Intake of liquids and and body weight were measured daily. Blood samples were collected on day 0 (baseline), day 5, and day 10 to analyze levels of glucose, triacylglycerol, cholesterol, and cortisol. On day 10, rats were euthanized and livers were collected to observe the severity of inflammation. The results indicated that rats receiving SHP 450 mg/kg and the mixture of pellet with SHP 450 mg/kg showed a similar ability as those receiving diazepam 0.18 mg/kg to cope with stress, indicated by an improvement in all blood biochemistry parameters. Supplementation with SHP 450 mg/kg can be used as an alternative source of functional food for overcoming oxidative stress, as indicated by its ability to improve levels of blood glucose, triacylglycerol, total cholesterol, and cortisol, and to improve liver histology by decreasing severity of liver inflammation.
Collapse
Affiliation(s)
- Amir Husni
- Department of Fisheries Faculty of Agriculture, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Radipta Lailatussifa
- Department of Fisheries Faculty of Agriculture, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia.,Sidoarjo Marine and Fisheries Polytechnic, Sidoarjo 61254, Indonesia
| | - Alim Isnansetyo
- Department of Fisheries Faculty of Agriculture, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
11
|
Cao C, Xiao J, Liu M, Ge Z, Huang R, Qi M, Zhu H, Zhu Y, Duan JA. Active components, derived from Kai-xin-san, a herbal formula, increase the expressions of neurotrophic factor NGF and BDNF on mouse astrocyte primary cultures via cAMP-dependent signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:554-562. [PMID: 29890314 DOI: 10.1016/j.jep.2018.06.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/23/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kai-Xin-San (KXS), an ancient formula composed of Ginseng Radix et Rhizoma, Polygalae Radix, Acori Tatarinowii Rhizoma and Poria, was frequently applied for Alzheimer's disease and major depression disorders for thousands of years. However, its active components and molecular mechanism have not clearly been investigated. AIM OF THE STUDY We aimed to reveal the active components of KXS on regulating neurotrophic factor NGF and BDNF expressions and its mechanisms on mouse astrocyte primary cultures. MATERIALS AND METHODS Extracts of KXS had been prepared by water reflux and chemical standardization was carried out by HPLC-MS/MS. Various ethanol elution components were prepared by eluting ethanol on macro pore resin column and compound identification was carried out by high-resolution mass spectrometry. KXS extract, elution components and identified chemicals were applied on mouse astrocytes and expressions of NGF and BDNF and related metabolic enzymes were analyzed by qPCR and western blotting analysis. RESULTS One compatible ratio of KXS named D-652 exerted the best effect on stimulation of NGF and BDNF expressions on mouse astrocytes. 70% ethanol elution fraction of D-652 exerted the highest increase tendency on expressions of NGF and BDNF by activating cAMP-dependent signaling pathway as well as stimulating enzymes accounting for neurotrophic factor synthesis. Combined with compound identification by high-resolution mass spectrometry, ginsenoside Rg1 and Rb1 might be the active compounds of this fraction on increasing NGF and BDNF expressions. CONCLUSIONS The active compounds of KXS on increasing NGF and BDNF expressions might be the ginsenosides via activating cAMP-dependent signaling pathway as well as stimulating enzymes accounting for neurotrophic factor synthesis, which partly reveal the target of this formulae supported the clinically usage of this decoction.
Collapse
Affiliation(s)
- Cheng Cao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Junyuan Xiao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mengqiu Liu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Zishuo Ge
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Renjie Huang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mingzhu Qi
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Hongliang Zhu
- Suzhou Guangji Hospital, Su Zhou, Jiangsu Province, China.
| | - Yue Zhu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Jin-Ao Duan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| |
Collapse
|
12
|
Dai Y, Zhang Y, Zhao X, Jeon Y, Zheng F, Ma L, Yue H. Identification and Evaluation of a Panel of Ginsenosides from Different Red Ginseng Extracts with Nootropic Effect. Chem Res Chin Univ 2018. [DOI: 10.1007/s40242-018-7422-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Sangiovanni E, Brivio P, Dell'Agli M, Calabrese F. Botanicals as Modulators of Neuroplasticity: Focus on BDNF. Neural Plast 2017; 2017:5965371. [PMID: 29464125 PMCID: PMC5804326 DOI: 10.1155/2017/5965371] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 11/09/2017] [Accepted: 12/02/2017] [Indexed: 12/19/2022] Open
Abstract
The involvement of brain-derived neurotrophic factor (BDNF) in different central nervous system (CNS) diseases suggests that this neurotrophin may represent an interesting and reliable therapeutic target. Accordingly, the search for new compounds, also from natural sources, able to modulate BDNF has been increasingly explored. The present review considers the literature on the effects of botanicals on BDNF. Botanicals considered were Bacopa monnieri (L.) Pennell, Coffea arabica L., Crocus sativus L., Eleutherococcus senticosus Maxim., Camellia sinensis (L.) Kuntze (green tea), Ginkgo biloba L., Hypericum perforatum L., Olea europaea L. (olive oil), Panax ginseng C.A. Meyer, Rhodiola rosea L., Salvia miltiorrhiza Bunge, Vitis vinifera L., Withania somnifera (L.) Dunal, and Perilla frutescens (L.) Britton. The effect of the active principles responsible for the efficacy of the extracts is reviewed and discussed as well. The high number of articles published (more than one hundred manuscripts for 14 botanicals) supports the growing interest in the use of natural products as BDNF modulators. The studies reported strengthen the hypothesis that botanicals may be considered useful modulators of BDNF in CNS diseases, without high side effects. Further clinical studies are mandatory to confirm botanicals as preventive agents or as useful adjuvant to the pharmacological treatment.
Collapse
Affiliation(s)
- Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Mario Dell'Agli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Moon M, Huh E, Lee W, Song EJ, Hwang DS, Lee TH, Oh MS. Coptidis Rhizoma Prevents Heat Stress-Induced Brain Damage and Cognitive Impairment in Mice. Nutrients 2017; 9:nu9101057. [PMID: 28946610 PMCID: PMC5691674 DOI: 10.3390/nu9101057] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/14/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022] Open
Abstract
Heat stress conditions lead to neuroinflammation, neuronal death, and memory loss in animals. Coptidis Rhizoma (CR) exhibits potent fever-reducing effects and has been used as an important traditional medicinal herb for treating fever. However, to date, the effects of antipyretic CR on heat-induced brain damages have not been investigated. In this study, CR significantly reduced the elevation of ear and rectal temperatures after exposure to heat in mice. Additionally, CR attenuated hyperthermia-induced stress responses, such as release of cortisol into the blood, and upregulation of heat shock protein and c-Fos in the hypothalamus and hippocampus of mice. The administration of CR inhibited gliosis and neuronal loss induced by thermal stress in the hippocampal CA3 region. Treatment with CR also reduced the heat stress-induced expression of nuclear factor kappa β, tumor necrosis factor-α, and interleukin-1β (IL-1β) in the hippocampus. Moreover, CR significantly decreased proinflammatory mediators such as IL-9 and IL-13 in the heat-stressed hypothalamus. Furthermore, CR attenuated cognitive dysfunction triggered by thermal stress. These results indicate that CR protects the brain against heat stress-mediated brain damage via amelioration of hyperthermia and neuroinflammation in mice, suggesting that fever-reducing CR can attenuate thermal stress-induced neuropathology.
Collapse
Affiliation(s)
- Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea.
| | - Eugene Huh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Department of Herbal Pharmacology, College of Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Wonil Lee
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Eun Ji Song
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea.
| | - Deok-Sang Hwang
- Department of Korean Gynecology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Tae Hee Lee
- Department of Formulae Pharmacology, School of Oriental Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam 13120, Korea.
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
15
|
Köse Çinar R, Sönmez MB, Görgülü Y. Peripheral blood mRNA expressions of stress biomarkers in manic episode and subsequent remission. Psychoneuroendocrinology 2016; 70:10-6. [PMID: 27138695 DOI: 10.1016/j.psyneuen.2016.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/19/2022]
Abstract
Theoretical models of the neuroprogressive nature of bipolar disorder (BD) are based on the hypothesis that it is an accelerated aging disease, with the allostatic load playing a major role. Glucocorticoids, oxidative stress markers, inflammatory cytokines and neurotrophins play important roles in BD. The messenger ribonucleic acid (mRNA) expressions of brain-derived neurotrophic factor (BDNF), tissue plasminogen activator (tPA), glucocorticoid receptor (GR), heat shock protein 70 (HSP70), tumour necrosis factor-alpha (TNF-α) were examined in the peripheral blood of 20 adult male, drug-free BD patients during manic and remission periods and in 20 adult male, healthy controls. mRNA expression was measured using the quantitative real-time polymerase chain reaction (qRT-PCR). Compared to the controls, the expressions of BDNF and tPA mRNA were down-regulated in mania. In remission, BNDF and tPA mRNA levels increased, but they were still lower than those of the controls. Between mania and remission periods, only the change in mRNA levels of BDNF reached statistical significance. The results suggest that BDNF and tPA may be biomarkers of BD and that proteolytic conversion of BDNF may be important in the pathophysiology of BD. The change in BDNF levels between mania and remission could be adaptive and used to follow the progression of BD.
Collapse
Affiliation(s)
- Rugül Köse Çinar
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne, Turkey.
| | | | - Yasemin Görgülü
- Department of Psychiatry, Trakya University Faculty of Medicine, Edirne, Turkey
| |
Collapse
|
16
|
Ahmed T, Raza SH, Maryam A, Setzer WN, Braidy N, Nabavi SF, de Oliveira MR, Nabavi SM. Ginsenoside Rb1 as a neuroprotective agent: A review. Brain Res Bull 2016; 125:30-43. [DOI: 10.1016/j.brainresbull.2016.04.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 03/21/2016] [Accepted: 04/05/2016] [Indexed: 12/30/2022]
|
17
|
Shan Y, Wang DD, Xu YX, Wang C, Cao L, Liu YS, Zhu CQ. Aging as a Precipitating Factor in Chronic Restraint Stress-Induced Tau Aggregation Pathology, and the Protective Effects of Rosmarinic Acid. J Alzheimers Dis 2016; 49:829-44. [PMID: 26577520 DOI: 10.3233/jad-150486] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stress is an important risk factor of Alzheimer's disease (AD). It has been evidenced that stress could induce tau phosphorylation and increase tau insolubility in brain; however, little is known about the interactional effect of stress with aging on tauopathy. Therefore, we explored the effects of aging on stress-induced tauopathy and the potential mechanism in mouse model of chronic restraint stress (CRS). Here we found that in general, the level of phosphorylated tau (P-tau) was higher in brain of middle-aged mice than that in adult mice under physiological conditions. CRS-induced tau phosphorylation and its insolubility were more prominent in middle-aged mice. The increase of AT8-labeled insoluble P-tau was dramatic in middle-aged mice, which was highly ubiquitinated but did not form PHF structures. The levels of chaperones were relatively lower in middle-aged mice brain; CRS further reduced the expression, especially for HDJ2/HSP40. CRS also suppressed the expression of Pin1, the peptidylprolyl cis/trans isomerase, in middle-aged mice but not in adult mice. Downregulation of HSP40 or Pin1 caused an increase of transfected extraneous tau in 293 cells. Rosmarinic acid (RA) could effectively suppress the elevation of P-tau and insoluble P-tau formation induced by CRS, and reversed the abnormal changes of chaperones and Pin1 particularly in middle-aged mice. Taken together, our findings provided evidence that aging could be a promoting factor in stress-induced tauopathy, which was relevant with malregulation of chaperones and Pin1, and RA might be a promising beneficial agent for stress-induced tauopathy.
Collapse
|