1
|
Alipanah-Moghadam R, Aghamohammadi V, Seifi S, Esmaeili H, Noroozzadeh S, Jeddi F, Salimnejad R, Nemati A. Protective effect of Panax ginseng extract on cisplatin-induced AKI via downregulating cell death associated genes. Sci Rep 2025; 15:3233. [PMID: 39863673 PMCID: PMC11763254 DOI: 10.1038/s41598-025-87447-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
This study is designed to assess the effect of root extract of P. ginseng on kidney tissue injury attributed to cisplatin and its molecular mechanism involved in this process in the AKI rat model. Twenty-four male Wistar rats were randomly allocated into 4 experimental groups including: the control group, the cisplatin group, the extract 100 mg/kg group, and the extract 200 mg/kg group. The duration of the investigation was 7 days, and all rats except the control group received a single dose of 10 mg/kg cisplatin on the 4th day. Our findings exhibited a significant reduction in blood concentration of creatinine in extract groups compared to the cisplatin group. In the cisplatin group, severe renal histopathological alterations were observed compared to the control group. In extract groups, significantly less tissue damage was observed than in the cisplatin group. Ginseng extract 200 showed minimal tissue damage as compared to extract 100. The expression of p21, p27, p53, TIMP2, IGFBP7, and NF-κB decreased significantly in extract groups compared to the cisplatin group. Our findings displayed amelioration of cisplatin-induced AKI and dose-dependent decrease of the NF-κB gene expression and cell death-inducing genes by administration of P. ginseng extract.
Collapse
Affiliation(s)
- Reza Alipanah-Moghadam
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Sina Seifi
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hedieh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Somaieh Noroozzadeh
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ramin Salimnejad
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Nemati
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
2
|
Zhao A, Liu N, Yao M, Zhang Y, Yao Z, Feng Y, Liu J, Zhou G. A Review of Neuroprotective Effects and Mechanisms of Ginsenosides From Panax Ginseng in Treating Ischemic Stroke. Front Pharmacol 2022; 13:946752. [PMID: 35873557 PMCID: PMC9302711 DOI: 10.3389/fphar.2022.946752] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/14/2022] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke has been considered one of the leading causes of mortality and disability worldwide, associated with a series of complex pathophysiological processes. However, effective therapeutic methods for ischemic stroke are still limited. Panax ginseng, a valuable traditional Chinese medicine, has been long used in eastern countries for various diseases. Ginsenosides, the main active ingredient of Panax ginseng, has demonstrated neuroprotective effects on ischemic stroke injury during the last decade. In this article, we summarized the pathophysiology of ischemic stroke and reviewed the literature on ginsenosides studies in preclinical and clinical ischemic stroke. Available findings showed that both major ginsenosides and minor ginsenosides (such as Rg3, Rg5, and Rh2) has a potential neuroprotective effect, mainly through attenuating the excitotoxicity, Ca2+ overload, mitochondria dysfunction, blood-brain barrier (BBB) permeability, anti-inflammation, anti-oxidative, anti-apoptosis, anti-pyroptosis, anti-autophagy, improving angiogenesis, and neurogenesis. Therefore, this review brings a current understanding of the mechanisms of ginsenosides in the treatment of ischemic stroke. Further studies, especially in clinical trials, will be important to confirm the clinical value of ginseng and ginsenosides.
Collapse
Affiliation(s)
- Aimei Zhao
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Nan Liu
- Beijing Increasepharm Safety and Efficacy Co., Ltd., Beijing, China
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Zengyu Yao
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yujing Feng
- Department of Anesthesiology, Punan Hospital, Shanghai, China
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jianxun Liu, ; Guoping Zhou,
| | - Guoping Zhou
- Department of Acupuncture and Moxibustion, Neuroscience Centre, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Jianxun Liu, ; Guoping Zhou,
| |
Collapse
|
3
|
Bilia AR, Bergonzi MC. The G115 standardized ginseng extract: an example for safety, efficacy, and quality of an herbal medicine. J Ginseng Res 2019; 44:179-193. [PMID: 32148399 PMCID: PMC7031746 DOI: 10.1016/j.jgr.2019.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/23/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022] Open
Abstract
Ginseng products on the market show high variability in their composition and overall quality. This becomes a challenge for both consumers and health-care professionals who are in search of high-quality, reliable ginseng products that have a proven safety and efficacy profile. The botanical extract standardization is of crucial importance in this context as it determines the reproducibility of the quality of the product that is essential for the evaluation of effectiveness and safety. This review focuses on the well-characterized and standardized ginseng extract, G115, which represents an excellent example of an herbal drug preparation with constant safety and efficacy within the herbal medicinal products. Over the many decades, extensive preclinical and clinical research has been conducted to evaluate the efficacy and safety of G115. In vitro and in vivo studies of G115 have shown pharmacological effects on physical performance, cognitive function, metabolism, and the immune system. Furthermore, a significant number of G115 clinical studies, most of them double-blind placebo-controlled, have reinforced the findings of preclinical evidence and proved the efficacy of this extract on blood glucose and lipid regulation, chronic obstructive pulmonary disease, energy, physical performance, and immune and cognitive functions. Clinical trials and 50 years of presence on the market are proof of a good safety profile of G115.
Collapse
Key Words
- 3′,5′-AMP, adenosine 3′5′ monophosphate
- AMPK, 5′ AMP-activated protein kinase
- ATP, adenosine triphosphate
- Blood glucose and lipid regulation
- CDR, cognitive drug research
- CDRI, cognitive drug research index
- CO, crossover
- COPD, chronic obstructive pulmonary disease
- Chronic obstructive pulmonary disease
- DB, double-blind
- DER, drug extract ratio
- Energy and physical performance
- FBG, fasting blood glucose
- FEF50, forced expiratory flow50
- FEF75, forced expiratory flow75
- FER, forced expiratory ratio
- FEV1, forced expiration volume in one second
- FEV1/FVC, ratio of FEV1/FVC
- FVC, forced vital capacity
- G115 standardized ginseng extract
- G115, standardized root extract of P. ginseng Meyer
- GACPs, good agricultural and collection practices
- GMPs, good manufacturing practices
- HDL-c, high-density lipoprotein
- HMPs, herbal medicinal products
- HbAlc, glycated hemoglobin
- Immune and cognitive functions
- LA, lipoic acid
- LDLc, low-density lipoprotein
- MVV, maximum ventilation volume
- PC, placebo-controlled
- PEF, peak expiration flow
- PEFR, peak expiration flow rate
- PFTs, pulmonary function tests
- PG, parallel group
- PGC-1α, proliferator-activated receptor gamma coactivator-1α
- PS, pilot study
- PaO2, blood oxygen pressure
- R, randomized
- RVIP, rapid visual information processing
- S-SIgA, SIgA secretion rate
- SB, single-blind
- SFR, saliva flow rate
- SIRT1, sirtuin 1
- SIgA, secretory immunoglobulin A
- TC, total cholesterol
- TG, triglyceride
- VLDL, very-low-density lipoprotein
- VO2 max, maximal oxygen consumption
- WHO, World Health Organization
- pO2, partial oxygen pressure
Collapse
Affiliation(s)
- Anna R Bilia
- Department of Chemistry, School of Human Health Sciences, University of Florence, Florence, Italy
| | - Maria C Bergonzi
- Department of Chemistry, School of Human Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Liu L, Anderson GA, Fernandez TG, Doré S. Efficacy and Mechanism of Panax Ginseng in Experimental Stroke. Front Neurosci 2019; 13:294. [PMID: 31068769 PMCID: PMC6491687 DOI: 10.3389/fnins.2019.00294] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/13/2019] [Indexed: 12/30/2022] Open
Abstract
Stroke is one of the leading causes of death and long-term disability worldwide. However, effective therapeutic approaches are still limited. The disruption of blood supply triggers complicated temporal and spatial events involving hemodynamic, biochemical, and neurophysiologic changes, eventually leading to pathological disturbance and diverse clinical symptoms. Ginseng (Panax ginseng), a popular herb distributed in East Asia, has been extensively used as medicinal and nutritional supplements for a variety of disorders worldwide. In recent years, ginseng has displayed attractive beneficial effects in distinct neurological disorders including stroke, involving multiple protective mechanisms. In this article, we reviewed the literature on ginseng studies in the experimental stroke field, particularly focusing on the in vivo evidence on the preventive or therapeutic efficacy and mechanisms of ginseng and ginsenosides in various stroke models of mice and rats. We also summarized the efficacy and underlying mechanisms of ginseng and ginsenosides on short- and long-term stroke outcomes.
Collapse
Affiliation(s)
- Lei Liu
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Gigi A Anderson
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Tyler G Fernandez
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Departments of Neurology, Psychiatry, Pharmaceutics, and Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Hwang SN, Kim JC, Bhuiyan MIH, Kim JY, Yang JS, Yoon SH, Yoon KD, Kim SY. Black Rice ( Oryza sativa L., Poaceae) Extract Reduces Hippocampal Neuronal Cell Death Induced by Transient Global Cerebral Ischemia in Mice. Exp Neurobiol 2018; 27:129-138. [PMID: 29731679 PMCID: PMC5934544 DOI: 10.5607/en.2018.27.2.129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022] Open
Abstract
Rice is the most commonly consumed grain in the world. Black rice has been suggested to contain various bioactive compounds including anthocyanin antioxidants. There is currently little information about the nutritional benefits of black rice on brain pathology. Here, we investigated the effects of black rice (Oryza sativa L., Poaceae) extract (BRE) on the hippocampal neuronal damage induced by ischemic insult. BRE (300 mg/kg) was orally administered to adult male C57BL/6 mice once a day for 21 days. Bilateral common carotid artery occlusion (BCCAO) was performed for 23 min on the 8th day of BRE or vehicle administration. Histological analyses conducted on the 22nd day of BRE or vehicle administration revealed that administering BRE profoundly attenuated neuronal cell death, inhibited reactive astrogliosis, and prevented loss of glutathione peroxidase expression in the hippocampus when compared to vehicle treatment. In addition, BRE considerably ameliorated BCCAO-induced memory impairment on the Morris water maze test from the 15th day to the 22nd day of BRE or vehicle administration. These results indicate that chronic administration of BRE is potentially beneficial in cerebral ischemia.
Collapse
Affiliation(s)
- Sun-Nyoung Hwang
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jae-Cheon Kim
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mohammad Iqbal Hossain Bhuiyan
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Joo Youn Kim
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ji Seon Yang
- Department of Physiology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Shin Hee Yoon
- Department of Physiology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Kee Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea
| | - Seong Yun Kim
- Department of Pharmacology, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
6
|
Di Meo S, Reed TT, Venditti P, Victor VM. Role of ROS and RNS Sources in Physiological and Pathological Conditions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1245049. [PMID: 27478531 PMCID: PMC4960346 DOI: 10.1155/2016/1245049] [Citation(s) in RCA: 859] [Impact Index Per Article: 95.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/04/2016] [Accepted: 05/23/2016] [Indexed: 12/19/2022]
Abstract
There is significant evidence that, in living systems, free radicals and other reactive oxygen and nitrogen species play a double role, because they can cause oxidative damage and tissue dysfunction and serve as molecular signals activating stress responses that are beneficial to the organism. Mitochondria have been thought to both play a major role in tissue oxidative damage and dysfunction and provide protection against excessive tissue dysfunction through several mechanisms, including stimulation of opening of permeability transition pores. Until recently, the functional significance of ROS sources different from mitochondria has received lesser attention. However, the most recent data, besides confirming the mitochondrial role in tissue oxidative stress and protection, show interplay between mitochondria and other ROS cellular sources, so that activation of one can lead to activation of other sources. Thus, it is currently accepted that in various conditions all cellular sources of ROS provide significant contribution to processes that oxidatively damage tissues and assure their survival, through mechanisms such as autophagy and apoptosis.
Collapse
Affiliation(s)
- Sergio Di Meo
- Dipartimento di Biologia, Università di Napoli “Federico II”, 80126 Napoli, Italy
| | - Tanea T. Reed
- Department of Chemistry, Eastern Kentucky University, Richmond, KY 40475, USA
| | - Paola Venditti
- Dipartimento di Biologia, Università di Napoli “Federico II”, 80126 Napoli, Italy
| | - Victor Manuel Victor
- Service of Endocrinology, University Hospital Dr. Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46010 Valencia, Spain
| |
Collapse
|
7
|
Sun ZG, Chen LP, Wang FW, Xu CY, Geng M. Protective effects of ginsenoside Rg1 against hydrogen peroxide-induced injury in human neuroblastoma cells. Neural Regen Res 2016; 11:1159-64. [PMID: 27630703 PMCID: PMC4994462 DOI: 10.4103/1673-5374.187057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2016] [Indexed: 01/02/2023] Open
Abstract
The active ingredient of ginseng, ginsenosides Rg1, has been shown to scavenge free radicals and improve antioxidant capacity. This study hypothesized that ginsenosides Rg1 has a protective role in human neuroblastoma cells injured by H2O2. Ginsenosides Rg1 at different concentrations (50 and 100 μM) was used to treat H2O2 (150 μM)-injured SH-SY5Y cells. Results demonstrated that ginsenoside Rg1 elevated the survival rate of SH-SY5Y cells injured by H2O2, diminished the amount of leaked lactate dehydrogenase, and increased superoxide dismutase activity. Ginsenoside Rg1 effectively suppressed caspase-3 immunoreactivity, and contributed to heat shock protein 70 gene expression, in a dose-dependent manner. These results indicate that ginsenoside Rg1 has protective effects on SH-SY5Y cells injured by H2O2 and that its mechanism of action is associated with anti-oxidation and the inhibition of apoptosis.
Collapse
Affiliation(s)
- Zhi-gao Sun
- Department of Traditional Chinese Medicine, Hainan Branch Hospital of Chinese PLA General Hospital, Sanya, Hainan Province, China
| | - Li-ping Chen
- Department of Traditional Chinese Medicine, Chinese PLA General Hospital, Beijing, China
| | - Fa-wei Wang
- Department of Traditional Chinese Medicine, Chinese PLA General Hospital, Beijing, China
| | - Cheng-yong Xu
- Department of Traditional Chinese Medicine, Hainan Branch Hospital of Chinese PLA General Hospital, Sanya, Hainan Province, China
| | - Miao Geng
- Institute of Gerontology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Song X, Zhu W, An R, Li Y, Du Z. Protective effect of Daming capsule against chronic cerebral ischemia. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:149. [PMID: 25966684 PMCID: PMC4456789 DOI: 10.1186/s12906-015-0668-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 05/07/2015] [Indexed: 12/20/2022]
Abstract
Background Accumulating evidence has shown that chronic cerebral ischemia (CCI) is one of the major causes of vascular dementia (VD) characterized by dysregulated cholesterol homeostasis and lipoprotein disturbances. Positive value of lipid-lowering agents has been widely evaluated for the treatment of VD. In the present study, we investigated whether Daming capsule (DMC) protected against CCI-induced VD and its possible mechanisms of action. DMC is a multi-herbal formula composed of Rheum palmatum L., Cassia obtusifolia L., Salvia miltiorrhiza, and Panax ginseng C.A., which has been used to treat hyperlipidemia for years in China. Methods A network pharmacology method was established to reveal whether DMC contained any chemical constituent targeting CCI-related proteins. Furthermore, the potential anti-CCI effects of DMC (100 mg/kg or 200 mg/kg) administered for 30 days were investigated in vivo on rats that were subjected to permanent bilateral occlusion of the carotid arteries (2-VO). Spatial learning and memory abilities were evaluated using a Morris water maze (MWM) and morphological changes of cerebral cortex and hippocampus were assessed using hematoxylin and eosin staining. Moreover, the lipid peroxidation levels and antioxidative capabilities were measured using biochemical analysis. Results Our network pharmacology analysis revealed the existence of multiple CCI-related chemical-target interactions in DMC, suggesting a potential protective effect. An in vivo experiment verified that 200 mg/kg DMC improved cognitive deficits of 2-VO rats in the MWM test and attenuated pathological alterations in both the cerebral cortex and the hippocampus. Biochemical assays indicated that DMC decreased malondialdehyde levels and CCI-elevated superoxide dismutase activities, but increased the activities of glutathione peroxidase and catalase. Conclusions Our findings suggested that DMC protected against cognitive dysfunction and nerve injuries caused by CCI, which is most likely related to its antioxidant actions.
Collapse
|
9
|
Huang W, Li Y, Lin Y, Ye X, Zang D. Effects of leukemia inhibitory factor and basic fibroblast growth factor on free radicals and endogenous stem cell proliferation in a mouse model of cerebral infarction. Neural Regen Res 2015; 7:1469-74. [PMID: 25657681 PMCID: PMC4308777 DOI: 10.3969/j.issn.1673-5374.2012.19.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/23/2012] [Indexed: 11/24/2022] Open
Abstract
The present study established a mouse model of cerebral infarction by middle cerebral artery occlusion, and monitored the effect of 25 μg/kg leukemia inhibitory factor and (or) basic fibroblast growth factor administration 2 hours after model establishment. Results showed that following administration, the number of endogenous neural stem cells in the infarct area significantly increased, malondialdehyde content in brain tissue homogenates significantly decreased, nitric oxide content, glutathione peroxidase and superoxide dismutase activity significantly elevated, and mouse motor function significantly improved as confirmed by the rotarod and bar grab tests. In particular, the effect of leukemia inhibitory factor in combination with basic fibroblast growth factor was the most significant. Results indicate that leukemia inhibitory factor and basic fibroblast growth factor can improve the microenvironment after cerebral infarction by altering free radical levels, improving the quantity of endogenous neural stem cells, and promoting neurological function of mice with cerebral infarction.
Collapse
Affiliation(s)
- Weihui Huang
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Yadan Li
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Xue Ye
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| | - Dawei Zang
- Department of Neurology, Tianjin First Center Hospital, Tianjin Medical University, Tianjin 300192, China
| |
Collapse
|
10
|
Rastogi V, Santiago-Moreno J, Doré S. Ginseng: a promising neuroprotective strategy in stroke. Front Cell Neurosci 2015; 8:457. [PMID: 25653588 PMCID: PMC4299449 DOI: 10.3389/fncel.2014.00457] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 12/16/2014] [Indexed: 12/30/2022] Open
Abstract
Ginseng is one of the most widely used herbal medicines in the world. It has been used in the treatment of various ailments and to boost immunity for centuries; especially in Asian countries. The most common ginseng variant in traditional herbal medicine is ginseng, which is made from the peeled and dried root of Panax Ginseng. Ginseng has been suggested as an effective treatment for a vast array of neurological disorders, including stroke and other acute and chronic neurodegenerative disorders. Ginseng’s neuroprotective effects are focused on the maintenance of homeostasis. This review involves a comprehensive literature search that highlights aspects of ginseng’s putative neuroprotective effectiveness, focusing on stroke. Attenuation of inflammation through inhibition of various proinflammatory mediators, along with suppression of oxidative stress by various mechanisms, including activation of the cytoprotective transcriptional factor Nrf2, which results in decrease in reactive oxygen species, could account for its neuroprotective efficacy. It can also prevent neuronal death as a result of stroke, thus decreasing anatomical and functional stroke damage. Although there are diverse studies that have investigated the mechanisms involved in the efficacy of ginseng in treating disorders, there is still much that needs to be clarified. Both in vitro and in vivo studies including randomized controlled clinical trials are necessary to develop in-depth knowledge of ginseng and its practical applications.
Collapse
Affiliation(s)
- Vaibhav Rastogi
- Departments of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA ; Departments of Neurology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Juan Santiago-Moreno
- Departments of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| | - Sylvain Doré
- Departments of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA ; Departments of Neurology, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA ; Departments of Psychiatry, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA ; Departments of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine Gainesville, FL, USA
| |
Collapse
|
11
|
Influence of Panax ginseng on Alpha-Adrenergic Receptor of Benign Prostatic Hyperplasia. Int Neurourol J 2014; 18:179-86. [PMID: 25558416 PMCID: PMC4280437 DOI: 10.5213/inj.2014.18.4.179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 11/15/2014] [Indexed: 12/13/2022] Open
Abstract
Purpose Benign prostatic hyperplasia (BPH) is the most common prostate problem in older men. The present study aimed to investigate the inhibitory effect of Panax ginseng C.A. Meyer (P. ginseng) on a rat model of testosterone-induced BPH. Methods The rats were divided into 3 groups (each group, n=10): control, testosterone-induced BPH (20 mg/kg, subcutaneous injection), and P. ginseng (200 mg/kg, orally) groups. After 4 weeks, all animals were sacrificed to examine the blood biochemical profiles, prostate volume, weight, histopathological changes, alpha-1D adrenergic receptor (Adra1d) mRNA expression, and epidermal growth factor receptor (EGFR) and B-cell CLL/lymphoma 2 (BCL2) protein expression. Results The group treated with P. ginseng showed significantly lesser prostate size and weight than the testosterone-induced BPH group. In addition, P. ginseng decreased the mRNA expression of Adra1d as well as the expression of EGFR and BCL2 in prostate tissue. Conclusions These results suggest that P. ginseng may inhibit the alpha-1-adrenergic receptor to suppress the development of BPH.
Collapse
|
12
|
Rinwa P, Kumar A. Panax quinquefolium involves nitric oxide pathway in olfactory bulbectomy rat model. Physiol Behav 2014; 129:142-51. [DOI: 10.1016/j.physbeh.2014.02.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 02/06/2014] [Accepted: 02/15/2014] [Indexed: 12/14/2022]
|
13
|
Smith I, Williamson EM, Putnam S, Farrimond J, Whalley BJ. Effects and mechanisms of ginseng and ginsenosides on cognition. Nutr Rev 2014; 72:319-33. [DOI: 10.1111/nure.12099] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Imogen Smith
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | - Elizabeth M Williamson
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| | | | | | - Benjamin J Whalley
- School of Chemistry; Food and Nutritional Sciences and Pharmacy; University of Reading; Reading Berkshire UK
| |
Collapse
|
14
|
Shin IS, Jeon WY, Shin HK, Cha SW, Lee MY. Banhabaekchulchunma-tang, a traditional herbal formula attenuates absolute ethanol-induced gastric injury by enhancing the antioxidant status. Altern Ther Health Med 2013; 13:170. [PMID: 23844748 PMCID: PMC3711929 DOI: 10.1186/1472-6882-13-170] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/08/2013] [Indexed: 01/20/2023]
Abstract
Background Banhabaekchulchunma-tang (hange-byakujutsu-tenma-to in Japanese and banxia-baizhu-tianma-tang in Chinese) is a mixture of fourteen herbs. It is used traditionally for the treatment of anemia, anorexia, general weakness, and female infertility in China, Japan, and Korea. In this study, we investigated the protective effects of a Banhabaekchulchunma-tang water extract (BCT) against ethanol-induced acute gastric injury in rats. Methods Gastric injury was induced by intragastric administration of 5 mL/kg body weight of absolute ethanol to each rat. The positive control group and the BCT group were given oral doses of omeprazole (50 mg/kg) or BCT (400 mg/kg), respectively, 2 h prior to the administration of absolute ethanol. The stomach of each animal was excised and examined for gastric mucosal lesions. To confirm the protective effects of BCT, we evaluated the degree of lipid peroxidation, the level of reduced glutathione (GSH), and the activities of the antioxidant enzymes catalase, glutathione-S-transferase, glutathione peroxidase, and glutathione reductase in the stomach. In addition, we conducted an acute toxicity study to evaluate the safety of BCT according to OECD guideline. Results BCT reduced ethanol-induced hemorrhage, hyperemia, and loss of epithelial cell in the gastric mucosa. BCT reduced the increased lipid peroxidation associated with ethanol-induced acute gastric lesions, and increased the mucosal GSH content and the activities of antioxidant enzymes. In addition, BCT did not cause any adverse effects at up to 5000 mg/kg. Conclusions These results indicate that BCT protects the gastric mucosa against ethanol-induced gastric injury by increasing the antioxidant status. We suggest that BCT could be developed as an effective drug for the treatment of gastric injury caused by alcohol intake.
Collapse
|
15
|
Kim HJ, Kim P, Shin CY. A comprehensive review of the therapeutic and pharmacological effects of ginseng and ginsenosides in central nervous system. J Ginseng Res 2013; 37:8-29. [PMID: 23717153 PMCID: PMC3659622 DOI: 10.5142/jgr.2013.37.8] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/30/2012] [Accepted: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
Ginseng is one of the most widely used herbal medicines in human. Central nervous system (CNS) diseases are most widely investigated diseases among all others in respect to the ginseng’s therapeutic effects. These include Alzheimer’s disease, Parkinson’s disease, cerebral ischemia, depression, and many other neurological disorders including neurodevelopmental disorders. Not only the various types of diseases but also the diverse array of target pathways or molecules ginseng exerts its effect on. These range, for example, from neuroprotection to the regulation of synaptic plasticity and from regulation of neuroinflammatory processes to the regulation of neurotransmitter release, too many to mention. In general, ginseng and even a single compound of ginsenoside produce its effects on multiple sites of action, which make it an ideal candidate to develop multi-target drugs. This is most important in CNS diseases where multiple of etiological and pathological targets working together to regulate the final pathophysiology of diseases. In this review, we tried to provide comprehensive information on the pharmacological and therapeutic effects of ginseng and ginsenosides on neurodegenerative and other neurological diseases. Side by side comparison of the therapeutic effects in various neurological disorders may widen our understanding of the therapeutic potential of ginseng in CNS diseases and the possibility to develop not only symptomatic drugs but also disease modifying reagents based on ginseng.
Collapse
Affiliation(s)
- Hee Jin Kim
- Department of Pharmacology, School of Medicine and Advanced Institute of Biomedical Science and Technology, Konkuk University, Seoul 143-701, Korea
| | | | | |
Collapse
|
16
|
Lee MY, Shin IS, Jeon WY, Seo CS, Ha H, Huh JI, Shin HK. Protective effect of Bojungikki-tang, a traditional herbal formula, against alcohol-induced gastric injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2012; 142:346-353. [PMID: 22580157 DOI: 10.1016/j.jep.2012.04.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/13/2012] [Accepted: 04/25/2012] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL EVIDENCE Oxidative stress plays an important role in the pathogenesis of ethanol-induced acute gastric mucosal injury. Bojungikki-tang (Hochuekkito in Japanese, Bu-zhong-yi-qi-tang in Chinese) is a traditional herbal formula used in Korea, Japan, and China to treat allergic diseases and gastrointestinal disorders. However, the mechanism responsible for its actions has not been investigated experimentally. AIM OF THE STUDY The aims of this study were to investigate whether Bojungikki-tang water extract (BJITE) has protective effects against ethanol-induced acute gastric injury in rats and to perform an acute toxicity study to evaluate its safety. MATERIALS AND METHODS In this rat model, gastric mucosal injury was imposed by oral administration of 5 mL/kg body weight of absolute ethanol. BJITE at one of two doses (200 or 400 mg/kg body weight) was administered by gavage 2 h before ethanol administration. Gastric tissues were collected and analyzed to assess the gastric injury index, and content or activity of catalase, superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), glutathione-S-transferase (GST), glutathione reductase (GR), and glutathione peroxidase (GPx). RESULTS Acute administration of ethanol significantly increased the gastric injury index concomitantly with an increase in MDA and GSH content, and a decrease in the activities of catalase, GST, GR, GPx, and SOD. Pretreatment with 200 or 400 mg/kg BJITE attenuated ethanol-induced gastric mucosal injury; this was accompanied by an increase in the content or activity of PGE(2), catalase, GSH, GST, GR, GPx, and SOD, and a decrease in MDA content. In the acute toxicity study, no adverse effects of BJITE were observed at doses up to 2000 mg/kg body weight. CONCLUSION These results indicate that BJITE can partly protect the gastric mucosa from ethanol-induced acute gastric injury and suggest that these protective effects might be induced by increasing the antioxidant status. We suggest that BJITE can be developed as an effective drug for the treatment of acute gastric injury.
Collapse
Affiliation(s)
- Mee-Young Lee
- Basic Herbal Medicine Research Group, Korea Institute of Oriental Medicine, 483 Expo-ro, Yusung-gu, Daejeon 305-811, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
17
|
Ban JY, Kang SW, Lee JS, Chung JH, Ko YG, Choi HS. Korean red ginseng protects against neuronal damage induced by transient focal ischemia in rats. Exp Ther Med 2012; 3:693-698. [PMID: 22969953 DOI: 10.3892/etm.2012.449] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 12/23/2011] [Indexed: 11/05/2022] Open
Abstract
In the present study, we investigated the neuroprotective effect of Korean red ginseng (KRG) following focal brain ischemia/reperfusion injury, in relation to its antioxidant activities. The middle cerebral artery occlusion/reperfusion (MCAO/R) model in rats was employed. The KRG extract (100 mg/kg, perorally) was administered once daily for 7 days following MCAO/R. The elevated levels of lipid peroxidation in the MCAO/R group were attenuated significantly in the KRG-administered group. The significantly depleted activity of the antioxidant enzymes glutathione peroxidase, superoxide dismutase and catalase was prevented in the KRG-administered group. In the neurobehavioral evaluation expressed as the modified neurological severity score and corner-turn test, the daily intake of KRG showed consistent and significant improvement in the neurological deficits for 7 days following MCAO/R injury. These results indicate that KRG has a neuroprotective effect against ischemia/reperfusion brain injury by reducing the level of lipid peroxidation and increasing the endogenous antioxidant enzymatic activity.
Collapse
Affiliation(s)
- Ju Yeon Ban
- Department of Pharmacology, College of Dentistry, Dankook University, Cheonan
| | | | | | | | | | | |
Collapse
|
18
|
Kang A, Hao H, Zheng X, Liang Y, Xie Y, Xie T, Dai C, Zhao Q, Wu X, Xie L, Wang G. Peripheral anti-inflammatory effects explain the ginsenosides paradox between poor brain distribution and anti-depression efficacy. J Neuroinflammation 2011; 8:100. [PMID: 21843370 PMCID: PMC3169467 DOI: 10.1186/1742-2094-8-100] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Accepted: 08/16/2011] [Indexed: 11/17/2022] Open
Abstract
Background The effectiveness of ginseng in preventing and treating various central nervous system (CNS) diseases has been widely confirmed. However, ginsenosides, the principal components of ginseng, are characterized by poor accessibility to the brain, and this pharmacokinetic-pharmacological paradox remains poorly explained. Anti-inflammatory approaches are becoming promising therapeutic strategies for depression and other CNS diseases; however, previous studies have focused largely on anti-inflammatory therapies directed at the central nervous system. It is thus of interest to determine whether ginsenosides, characterized by poor brain distribution, are also effective in treating lipopolysaccharide- (LPS) induced depression-like behavior and neuroinflammation. Methods In an LPS-induced depression-like behavior model, the antidepressant effects of ginseng total saponins (GTS) were assessed using a forced swimming test, a tail suspension test, and a sucrose preference test. The anti-inflammatory efficacies of GTS in brain, plasma, and LPS-challenged RAW264.7 cells were validated using ELISA and quantitative real-time PCR. Moreover, indoleamine 2,3-dioxygenase (IDO) activity in the periphery and brain were also determined by measuring levels of kynurenine/tryptophan. Results GTS significantly attenuated LPS-induced depression-like behavior. Moreover, LPS-induced increases in 5-HT and tryptophane turnover in the brain were significantly reduced by GTS. IDO activities in brain and periphery were also suppressed after pretreatment with GTS. Furthermore, GTS-associated recovery from LPS-induced depression-like behavior was paralleled with reduced mRNA levels for IL-1β, IL-6, TNF-α, and IDO in hippocampus. Poor brain distribution of ginsenosides was confirmed in LPS-challenged mice. GTS treatment significantly decreased production of various proinflammatory cytokines in both LPS-challenged mice and RAW264.7 cells. Conclusion This study suggests that the anti-depression efficacy of GTS may be largely attributable to its peripheral anti-inflammatory activity. Our study also strengthens an important notion that peripheral anti-inflammation strategies may be useful in the therapy of inflammation-related depression and possibly other CNS diseases.
Collapse
Affiliation(s)
- An Kang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yoo KY, Lee CH, Li H, Park JH, Choi JH, Hwang IK, Kang IJ, Won MH. Ethyl acetate extracts of raw and steamed Codonopsis lanceolata protects against ischemic damage potentially by maintaining SOD1 and BDNF levels. Int J Neurosci 2011; 121:503-9. [PMID: 21671837 DOI: 10.3109/00207454.2011.580867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We observed the neuroprotective effects of ECLs treatment on ischemic damage in the gerbil hippocampal CA1 region four days after an ischemic insult. Among the 10 ECLs, EERCL and EESCL showed significant neuroprotection: the percentage of neurons remaining after treatment with EERCL and EESCL was 72.7% and 68.4% of that seen in the sham-ischemia group, respectively. The administration of EERCL and EESCL significantly decreased the reactive gliosis of microglia compared with that seen in the vehicle-treated ischemia group. In addition, SOD1 and BDNF immunoreactivity in the EERCL- and EESCL-ischemia groups were markedly increased compared with that in the vehicle-treated ischemia group. These results suggest that the administration of EERCL and EESCL can reduce ischemic neuronal loss potentially by maintaining SOD1 and BDNF immunoreactivity in the ischemic hippocampal CA1 region.
Collapse
Affiliation(s)
- Ki-Yeon Yoo
- Department of Oral Anatomy, College of Dentistry, Gangneung-Wonju National University, Gangneung, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Effect of combination therapy with sodium ozagrel and panax ginseng on transient cerebral ischemia model in rats. J Biomed Biotechnol 2011; 2010:893401. [PMID: 21274269 PMCID: PMC3022223 DOI: 10.1155/2010/893401] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 12/08/2010] [Indexed: 12/24/2022] Open
Abstract
Sodium ozagrel (SO) prevents platelet aggregation and vasoconstriction in the cerebral ischemia. It plays an important role in the prevention of brain damage induced by cerebral ischemia/reperfusion. Recently, many animal studies have suggested that the Panax ginseng (PG) has neuroprotective effects in the ischemic brain. In this study, we assessed the neuroprotective effects that come from a combination therapy of SO and PG in rat models with middle cerebral artery occlusion (MCAO). Animals with MCAO were assigned randomly to one of the following four groups: (1) control (Con) group, (2) SO group (3 mg/kg, intravenously), (3) PG group (200 mg/kg, oral feeding), and (4) SO + PG group. The rats were subjected to a neurobehavior test including adhesive removal test and rotarod test at 1, 3, 7, 10, and 15 days after MCAO. The cerebral ischemic volume was quantified by Metamorph imaging software after 2-3-5-triphenyltetrazolium (TTC) staining. The neuronal cell survival and astrocytes expansion were assessed by immunohistofluorescence staining. In the adhesive removal test, the rats of PG or SO + PG group showed significantly better performance than those of the control group (Con: 88.1 ± 24.8, PG: 43.6 ± 11, SO + PG: 11.8 ± 7, P < .05). Notably, the combination therapy group (SO + PG) showed better performance than the SO group alone (SO: 56 ± 12, SO + PG: 11.8 ± 7, P < .05). In TTC staining for infarct volume, cerebral ischemic areas were also significantly reduced in the PG group and SO + PG group (Con: 219 ± 32, PG: 117 ± 8, SO + PG: 99 ± 11, P < .05). Immunohistofluorescence staining results showed that the group which received SO + PG group therapy had neuron cells in the normal range. They also had a low number of astrocytes and apoptotic cells compared with the control or SO group in the peri-infarction area. During astrocytes staining, compared to the SO + PG group, the PG group showed only minor differences in the number of NeuN-positive cells and quantitative analysis of infarct volume. In conclusion, these studies showed that in MCAO rat models, the combination therapy with SO and PG may provide better neuroprotective effects such as higher neuronal cell survival and inhibition of astrocytes expansion than monotherapy with SO alone.
Collapse
|
21
|
Xu C, Teng J, Chen W, Ge Q, Yang Z, Yu C, Yang Z, Jia W. 20(S)-protopanaxadiol, an active ginseng metabolite, exhibits strong antidepressant-like effects in animal tests. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:1402-11. [PMID: 20647027 DOI: 10.1016/j.pnpbp.2010.07.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 07/10/2010] [Accepted: 07/12/2010] [Indexed: 12/21/2022]
Abstract
Ginseng has been used for mood adjustment in traditional Chinese medicine for thousands of years. Our previous study has shown that, total ginsenosides, the major pharmacologically functional ingredients of ginseng, possess antidepressant activity. In the present study, we hypothesized that an intestinal metabolite of ginseng, 20(S)-protopanaxadiol (code name S111), as a post metabolism compound (PMC) of ingested ginsenosides, may be responsible for the antidepressant activity of ginseng. To test this hypothesis, antidepressant-like activity of orally given S111 was measured in animal tests including tail suspension test, forced swimming test and rat olfactory bulbectomy depression model. In all those tests, S111 demonstrated antidepressant-like activity as potent as fluoxetine. S111 treated bulbectomy animals had higher levels of monoamine neurotransmitters in the brain and in vitro reuptake assay showed that S111 had a mild inhibitory effect. Furthermore, S111 but not fluoxetine significantly reduced brain oxidative stress and down-regulated serum corticosterone concentration in bulbectomy animals. No disturbance to central nervous system (CNS) normal functions were found in S111 treated animals. These results suggest that the ginseng active metabolite S111 is a potential antidepressant. Since the monoamine reuptake activity of this compound is rather weak, it remains to be investigated whether its antidepressant-like effect is by mechanisms that are different from current antidepressants. Furthermore, this study has demonstrated that post metabolism compounds (PMCs) of herb medicines such as S111 may be a novel source for drug discovery from medicinal herbs.
Collapse
Affiliation(s)
- Changjiang Xu
- Shanghai Innovative Research Centre of Traditional Chinese Medicine, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Gong J, Liu W, Dong J, Wang Y, Xu H, Wei W, Zhong J, Xi Q, Chen J. Developmental iodine deficiency and hypothyroidism impair neural development in rat hippocampus: involvement of doublecortin and NCAM-180. BMC Neurosci 2010; 11:50. [PMID: 20412599 PMCID: PMC2876162 DOI: 10.1186/1471-2202-11-50] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 04/23/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Developmental iodine deficiency results in inadequate thyroid hormone (TH), which damages the hippocampus. Here, we explored the roles of hippocampal doublecortin and neural cell adhesion molecule (NCAM)-180 in developmental iodine deficiency and hypothyroidism. METHODS Two developmental rat models were established with either an iodine-deficient diet, or propylthiouracil (PTU)-adulterated water (5 ppm or 15 ppm) to impair thyroid function, in pregnant rats from gestational day 6 until postnatal day (PN) 28. Silver-stained neurons and protein levels of doublecortin and NCAM-180 in several hippocampal subregions were assessed on PN14, PN21, PN28, and PN42. RESULTS The results show that nerve fibers in iodine-deficient and 15 ppm PTU-treated rats were injured on PN28 and PN42. Downregulation of doublecortin and upregulation of NCAM-180 were observed in iodine-deficient and 15 ppm PTU-treated rats from PN14 on. These alterations were irreversible by the restoration of serum TH concentrations on PN42. CONCLUSION Developmental iodine deficiency and hypothyroidism impair the expression of doublecortin and NCAM-180, leading to nerve fiber malfunction and thus impairments in hippocampal development.
Collapse
Affiliation(s)
- Jian Gong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Selenoproteins and the aging brain. Mech Ageing Dev 2010; 131:253-60. [DOI: 10.1016/j.mad.2010.02.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 01/11/2010] [Accepted: 02/20/2010] [Indexed: 01/18/2023]
|
24
|
Lee B, Park J, Kwon S, Park MW, Oh SM, Yeom MJ, Shim I, Lee HJ, Hahm DH. Effect of wild ginseng on scopolamine-induced acetylcholine depletion in the rat hippocampus. J Pharm Pharmacol 2010; 62:263-71. [DOI: 10.1211/jpp.62.02.0015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract
Objectives
The ameliorating effects of wild ginseng on learning and memory deficits were investigated in rats.
Methods
Rats were treated daily with wild ginseng or cultivated ginseng for 7 days at 30 min before scopolamine injection (2 mg/kg, i.p.). After inducing cognitive impairment by the administration of scopolamine, behavioural assessment using the Morris water maze was performed. Changes in cholinergic system reactivity were also examined by measuring the immunoreactive neurons of choline acetyltransferase and the reactivity of acetylcholinesterase in the hippocampus.
Key findings
Scopolamine injection induced impaired performance in the water maze test and severe cell losses in hippocampal cholinergic neurons, as indicated by decreased choline acetyltransferase immunoreactivity and increased acetylcholinesterase reactivity. Daily administration of wild ginseng produced a significant improvement in the escape latency for finding the platform in the Morris water maze and reduced the loss of cholinergic immunoreactivity in the hippocampus. The reduced expression of brain-derived neurotrophic factor mRNA due to the scopolamine injection was recovered to normal levels by the administration of wild ginseng.
Conclusions
Wild ginseng demonstrates a significant neuroprotective effect against scopolamine-induced neuronal and cognitive impairment.
Collapse
Affiliation(s)
- Bombi Lee
- Department of Oriental Medicine, Kyung Hee University, Republic of Korea
| | - Jongbong Park
- Acupuncture and Meridian Science Research Center, Republic of Korea
| | - Sunoh Kwon
- Acupuncture and Meridian Science Research Center, Republic of Korea
| | - Moo-Won Park
- Acupuncture and Meridian Science Research Center, Republic of Korea
- Department of Oriental Medicine, Kyung Hee University, Republic of Korea
| | - Sang-Muk Oh
- Department of Biochemistry, College of Medical Science, Konyang University, Daejeon, Republic of Korea
| | - Mi-Jung Yeom
- Acupuncture and Meridian Science Research Center, Republic of Korea
| | - Insop Shim
- Department of Integrative Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hye-Jung Lee
- Acupuncture and Meridian Science Research Center, Republic of Korea
- Department of Oriental Medicine, Kyung Hee University, Republic of Korea
| | - Dae-Hyun Hahm
- Acupuncture and Meridian Science Research Center, Republic of Korea
| |
Collapse
|
25
|
Gong J, Dong J, Wang Y, Xu H, Wei W, Zhong J, Liu W, Xi Q, Chen J. Developmental iodine deficiency and hypothyroidism impair neural development, up-regulate caveolin-1 and down-regulate synaptophysin in rat hippocampus. J Neuroendocrinol 2010; 22:129-39. [PMID: 20025630 DOI: 10.1111/j.1365-2826.2009.01943.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Developmental iodine deficiency leads to inadequate thyroid hormone, which damages the hippocampus. In the present study, we implicate hippocampal caveolin-1 and synaptophysin in developmental iodine deficiency and hypothyroidism. Two developmental rat models were established: pregnant rats were administered either an iodine-deficient diet or propylthiouracil (PTU)-adulterated (5 p.p.m. or 15 p.p.m.) drinking water from gestational day 6 until postnatal day (PN) 28. Nissl staining and the levels of caveolin-1 and synaptophysin in several hippocampal subregions were assessed on PN14, PN21, PN28 and PN42. The results obtained show that surviving cells in the iodine-deficient and PTU-treated rats were lower than in controls. Up-regulation of caveolin-1 and down-regulation of synaptophysin were observed in the iodine-deficient and PTU-treated rats. Our findings implicate decreases in the number of surviving cells and alterations in the levels of caveolin-1 and synaptophysin in the impairments in neural development induced by developmental iodine deficiency and hypothyroidism.
Collapse
Affiliation(s)
- J Gong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | | | | | | | | | | | | | | | | |
Collapse
|