1
|
Anand A, Shrivastava A, Singh K, Barik R, Gayakwad D, Jailani S, Shamim, Dwivedi S. Neuroprotective Efficacy and Complementary Treatment with Medicinal Herbs: A Comprehensive Review of Recent Therapeutic Approaches in Epilepsy Management. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:60-73. [PMID: 39069797 DOI: 10.2174/0118715273332140240724093837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/30/2024]
Abstract
Central Nervous System (CNS) disorders affect millions of people worldwide, with a significant proportion experiencing drug-resistant forms where conventional medications fail to provide adequate seizure control. This abstract delves into recent advancements and innovative therapies aimed at addressing the complex challenge of CNS-related drug-resistant epilepsy (DRE) management. The idea of precision medicine has opened up new avenues for epilepsy treatment. Herbs such as curcumin, ginkgo biloba, panax ginseng, bacopa monnieri, ashwagandha, and rhodiola rosea influence the BDNF pathway through various mechanisms. These include the activation of CREB, inhibition of NF-κB, modulation of neurotransmitters, reduction of oxidative stress, and anti- inflammatory effects. By promoting BDNF expression and activity, these herbs support neuroplasticity, cognitive function, and overall neuronal health. Novel antiepileptic drugs (AEDs) with distinct mechanisms of action demonstrate efficacy in refractory cases where traditional medications falter. Additionally, repurposing existing drugs for antiepileptic purposes presents a cost-effective strategy to broaden therapeutic choices. Cannabidiol (CBD), derived from cannabis herbs, has garnered attention for its anticonvulsant properties, offering a potential adjunctive therapy for refractory seizures. In conclusion, recent advances and innovative therapies represent a multifaceted approach to managing drug-resistant epilepsy. Leveraging precision medicine, neurostimulation technologies, novel pharmaceuticals, and complementary therapies, clinicians can optimize treatment outcomes and improve the life expectancy of patients living with refractory seizures. Genetic testing and biomarker identification now allow for personalized therapeutic approaches tailored to individual patient profiles. Utilizing next-generation sequencing techniques, researchers have elucidated genetic mutations.
Collapse
Affiliation(s)
- Amit Anand
- Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Aman Shrivastava
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Rakesh Barik
- GITAM School of Pharmacy, GITAM University, Hyderabad, Telangana, India
| | - Devshree Gayakwad
- Acropolis Institute of Pharmaceutical Education and Research, Indore, Madhya Pradesh, India
| | - S Jailani
- Formulation R&D Department, Alpha Pharma, KAEC, Rabigh, Kingdom of Saudi Arabia
| | - Shamim
- IIMT College of Medical Sciences, IIMT University, Ganga Nagar, Meerut, Uttar Pradesh, India
| | - Sumeet Dwivedi
- Acropolis Institute of Pharmaceutical Education and Research, Indore, Madhya Pradesh, India
| |
Collapse
|
2
|
Kim JH, Lee RM, Oh HB, Kim TY, Rhim H, Choi YK, Kim JH, Oh S, Kim DG, Cho IH, Nah SY. Atypical formations of gintonin lysophosphatidic acids as new materials and their beneficial effects on degenerative diseases. J Ginseng Res 2024; 48:1-11. [PMID: 38223830 PMCID: PMC10785247 DOI: 10.1016/j.jgr.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/12/2023] [Indexed: 02/23/2023] Open
Abstract
Fresh ginseng is prone to spoilage due to its high moisture content. For long-term storage, most fresh ginsengs are dried to white ginseng (WG) or steamed for hours at high temperature/pressure and dried to form Korean Red ginseng (KRG). They are further processed for ginseng products when subjected to hot water extraction/concentration under pressure. These WG or KRG preparation processes affect ginsenoside compositions and also other ginseng components, probably during treatments like steaming and drying, to form diverse bioactive phospholipids. It is known that ginseng contains high amounts of gintonin lysophosphatidic acids (LPAs). LPAs are simple lipid-derived growth factors in animals and humans and act as exogenous ligands of six GTP-binding-protein coupled LPA receptor subtypes. LPAs play diverse roles ranging from brain development to hair growth in animals and humans. LPA-mediated signaling pathways involve various GTP-binding proteins to regulate downstream pathways like [Ca2+]i transient induction. Recent studies have shown that gintonin exhibits anti-Alzheimer's disease and anti-arthritis effects in vitro and in vivo mediated by gintonin LPAs, the active ingredients of gintonin, a ginseng-derived neurotrophin. However, little is known about how gintonin LPAs are formed in high amounts in ginseng compared to other herbs. This review introduces atypical or non-enzymatic pathways under the conversion of ginseng phospholipids into gintonin LPAs during steaming and extraction/concentration processes, which exert beneficial effects against degenerative diseases, including Alzheimer's disease and arthritis in animals and humans via LPA receptors.
Collapse
Affiliation(s)
- Ji-Hun Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ra Mi Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyo-Bin Oh
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jeollabuk-do, Republic of Korea
| | - Tae-Young Kim
- Department of Efficacy Study, Institute of Jinan Red Ginseng, Jeollabuk-do, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Bio/Molecular Informatics Center, Republic of Korea
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Jeollabuk-do, Republic of Korea
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Do-Geun Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Arreola J, Pérez-Cornejo P, Segura-Covarrubias G, Corral-Fernández N, León-Aparicio D, Guzmán-Hernández ML. Function and Regulation of the Calcium-Activated Chloride Channel Anoctamin 1 (TMEM16A). Handb Exp Pharmacol 2024; 283:101-151. [PMID: 35768554 DOI: 10.1007/164_2022_592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Various human tissues express the calcium-activated chloride channel Anoctamin 1 (ANO1), also known as TMEM16A. ANO1 allows the passive chloride flux that controls different physiological functions ranging from muscle contraction, fluid and hormone secretion, gastrointestinal motility, and electrical excitability. Overexpression of ANO1 is associated with pathological conditions such as hypertension and cancer. The molecular cloning of ANO1 has led to a surge in structural, functional, and physiological studies of the channel in several tissues. ANO1 is a homodimer channel harboring two pores - one in each monomer - that work independently. Each pore is activated by voltage-dependent binding of two intracellular calcium ions to a high-affinity-binding site. In addition, the binding of phosphatidylinositol 4,5-bisphosphate to sites scattered throughout the cytosolic side of the protein aids the calcium activation process. Furthermore, many pharmacological studies have established ANO1 as a target of promising compounds that could treat several illnesses. This chapter describes our current understanding of the physiological roles of ANO1 and its regulation under physiological conditions as well as new pharmacological compounds with potential therapeutic applications.
Collapse
Affiliation(s)
- Jorge Arreola
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | - Patricia Pérez-Cornejo
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Guadalupe Segura-Covarrubias
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Nancy Corral-Fernández
- Department of Physiology and Biophysics, School of Medicine of Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Daniel León-Aparicio
- Physics Institute, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | | |
Collapse
|
4
|
Choi NR, Kwon MJ, Choi WG, Kim SC, Park JW, Nam JH, Kim BJ. The traditional herbal medicines mixture, Banhasasim-tang, relieves the symptoms of irritable bowel syndrome via modulation of TRPA1, NaV1.5 and NaV1.7 channels. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116499. [PMID: 37059250 DOI: 10.1016/j.jep.2023.116499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The cause of irritable bowel syndrome (IBS), a functional gastrointestinal (GI) disorder, remains unclear. Banhasasim-tang (BHSST), a traditional herbal medicines mixture, mainly used to treat GI-related diseases, may have a potential in IBS treatment. IBS is characterized by abdominal pain as the main clinical symptom, which seriously affects the quality of life. AIM OF THE STUDY We conducted a study to evaluate the effectiveness of BHSST and its mechanisms of action in treating IBS. MATERIALS AND METHODS We evaluated the efficacy of BHSST in a zymosan-induced diarrhea-predominant animal model of IBS. Electrophysiological methods were used to confirm modulation of transient receptor potential (TRP) and voltage-gated Na+ (NaV) ion channels, which are associated mechanisms of action. RESULTS Oral administration of BHSST decreased colon length, increased stool scores, and increased colon weight. Weight loss was also minimized without affecting food intake. In mice administered with BHSST, the mucosal thickness was suppressed, making it similar to that of normal mice, and the degree of tumor necrosis factor-α was severely reduced. These effects were similar to those of the anti-inflammatory drug-sulfasalazine-and antidepressant-amitriptyline. Moreover, pain-related behaviors were substantially reduced. Additionally, BHSST inhibited TRPA1, NaV1.5, and NaV1.7 ion channels associated with IBS-mediated visceral hypersensitivity. CONCLUSIONS In summary, the findings suggest that BHSST has potential beneficial effects on IBS and diarrhea through the modulation of ion channels.
Collapse
Affiliation(s)
- Na Ri Choi
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, 50612, Republic of Korea.
| | - Min Ji Kwon
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, 50612, Republic of Korea.
| | - Woo-Gyun Choi
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, 50612, Republic of Korea.
| | - Sang Chan Kim
- College of Oriental Medicine Daegu Haany University, Gyeongsan, 38610, Republic of Korea
| | - Jae-Woo Park
- Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Kyungju, 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang, 10326, Republic of Korea.
| | - Byung Joo Kim
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
5
|
Park KT, Jo H, Kim B, Kim W. Red Ginger Extract Prevents the Development of Oxaliplatin-Induced Neuropathic Pain by Inhibiting the Spinal Noradrenergic System in Mice. Biomedicines 2023; 11:432. [PMID: 36830967 PMCID: PMC9953630 DOI: 10.3390/biomedicines11020432] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023] Open
Abstract
Oxaliplatin is a well-known chemotherapeutic drug that is widely used to treat colorectal cancer. However, it can induce acute side effects in up to 90% of patients. Serotonin and norepinephrine reuptake inhibitors (SNRIs) are used as first-choice drugs; however, even SNRIs are known to be effective only in treatment and not for prevention. Therefore, finding a drug that can prevent the development of cold and mechanical forms of allodynia induced by oxaliplatin is needed. This study demonstrated that multiple oral administrations of 100 mg/kg and 300 mg/kg of red ginger extract could significantly prevent pain development in mice. The role of the noradrenergic system was investigated as an underlying mechanism of action. Both the spinal α1- and α2-adrenergic receptors were significantly downregulated after treatment. Furthermore, the noradrenaline levels in the serum and spinal cord were upregulated and downregulated after treatment with paclitaxel and red ginger, respectively. As the active sub-component of red ginger, ginsenoside Rg3 (Rg3) was identified and quantified using HPLC. Moreover, multiple intraperitoneal injections of Rg3 prevented the development of pain in paclitaxel-treated mice, suggesting that RG3 may induce the effect of red ginger extract.
Collapse
Affiliation(s)
- Keun-Tae Park
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Heejoon Jo
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Bonglee Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Hong J, Geem KR, Kim J, Jo IH, Yang TJ, Shim D, Ryu H. Prolonged Exposure to High Temperature Inhibits Shoot Primary and Root Secondary Growth in Panax ginseng. Int J Mol Sci 2022; 23:11647. [PMID: 36232949 PMCID: PMC9569605 DOI: 10.3390/ijms231911647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022] Open
Abstract
High temperature is one of the most significant abiotic stresses reducing crop yield and quality by inhibiting plant growth and development. Global warming has recently increased the frequency of heat waves, which negatively impacts agricultural fields. Despite numerous studies on heat stress responses and signal transduction in model plant species, the molecular mechanism underlying thermomorphogenesis in Panax ginseng remains largely unknown. Here, we investigated the high temperature response of ginseng at the phenotypic and molecular levels. Both the primary shoot growth and secondary root growth of ginseng plants were significantly reduced at high temperature. Histological analysis revealed that these decreases in shoot and root growth were caused by decreases in cell elongation and cambium stem cell activity, respectively. Analysis of P. ginseng RNA-seq data revealed that heat-stress-repressed stem and root growth is closely related to changes in photosynthesis, cell wall organization, cell wall loosening, and abscisic acid (ABA) and jasmonic acid (JA) signaling. Reduction in both the light and dark reactions of photosynthesis resulted in defects in starch granule development in the storage parenchymal cells of the main tap root. Thus, by combining bioinformatics and histological analyses, we show that high temperature signaling pathways are integrated with crucial biological processes that repress stem and root growth in ginseng, providing novel insight into the heat stress response mechanism of P. ginseng.
Collapse
Affiliation(s)
- Jeongeui Hong
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Kyoung Rok Geem
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| | - Jaewook Kim
- Department of Biological Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Ick-Hyun Jo
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong 27709, Korea
| | - Tae-Jin Yang
- Department of Agriculture, Forestry and Bioresources, Plant Genomics and Breeding Institute, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Donghwan Shim
- Department of Biological Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Hojin Ryu
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
7
|
Zha Z, Liu S, Liu Y, Li C, Wang L. Potential Utility of Natural Products against Oxidative Stress in Animal Models of Multiple Sclerosis. Antioxidants (Basel) 2022; 11:antiox11081495. [PMID: 36009214 PMCID: PMC9404913 DOI: 10.3390/antiox11081495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-mediated degenerative disease of the central nervous system (CNS) characterized by immune cell infiltration, demyelination and axonal injury. Oxidative stress-induced inflammatory response, especially the destructive effect of immune cell-derived free radicals on neurons and oligodendrocytes, is crucial in the onset and progression of MS. Therefore, targeting oxidative stress-related processes may be a promising preventive and therapeutic strategy for MS. Animal models, especially rodent models, can be used to explore the in vivo molecular mechanisms of MS considering their similarity to the pathological processes and clinical signs of MS in humans and the significant oxidative damage observed within their CNS. Consequently, these models have been used widely in pre-clinical studies of oxidative stress in MS. To date, many natural products have been shown to exert antioxidant effects to attenuate the CNS damage in animal models of MS. This review summarized several common rodent models of MS and their association with oxidative stress. In addition, this review provides a comprehensive and concise overview of previously reported natural antioxidant products in inhibiting the progression of MS.
Collapse
|
8
|
Jin Y, Liu D, Lu Z, Yang L, Chen J, Zhou X, Qiu Z, Jin Y. Preparation and Evaluation of Liposomes and Niosomes Containing Total Ginsenosides for Anti-Photoaging Therapy. Front Bioeng Biotechnol 2022; 10:874827. [PMID: 35464721 PMCID: PMC9019747 DOI: 10.3389/fbioe.2022.874827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/14/2022] [Indexed: 12/31/2022] Open
Abstract
Ginsenosides are the principal bioactive compounds of ginseng. Total ginsenosides (GS) contain a variety of saponin monomers, which have potent anti-photoaging activity and improve the skin barrier function. To enhance the efficiency of GS transdermal absorption, GS liposomes (GSLs) and GS niosomes (GSNs) were formulated as delivery vehicles. Based on the clarified and optimized formulation process, GSL and GSN were prepared. The structure, cumulative transmittance, skin retention, total transmittance, and bioactivity of GSLs and GSNs were characterized. GSL and GSN were shown to inhibit lipid peroxidation and increase the contents of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in human keratinocytes (HaCaTs). In addition, HaCAT cell migration, proliferation, and GS cellular uptake were significantly increased. The therapeutic effects of GSL and GSN were also evaluated in a rat model of photoaging. Histopathological changes were assessed in rat skin treated with GSL, GSN, or GS by hematoxylin–eosin (H&E) and aldehyde fuchsine staining. Malondialdehyde (MDA), SOD, GSH-Px, matrix metalloproteinases (MMPs), interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) expression levels were determined. Results indicated that the optimal formulation of GSL used soybean lecithin (SPC) as the phospholipid, with a lipid–drug ratio of 1:0.4 and a phospholipid–cholesterol ratio of 1:3.5. The optimal temperature for the preparation process of GSN by ethanol injection was 65°C, with a ratio of the organic phase to aqueous phase of 1:9. It was demonstrated that the cumulative release rate, skin retention rate, and total transmission rate of GSL-7 at 24 h were higher than those of GSN-4 and GS. GSL-7 significantly inhibited skin lipid peroxidation caused by ultraviolet (UV) radiation. In addition, GSL-7 reduced the contents of MMPs and inflammatory cytokines in skin tissue. In conclusion, GSL-7 may reduce skin aging caused by UV radiation and contribute to skin tissue repair.
Collapse
Affiliation(s)
- Yuanyuan Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Zhen Lu
- School of Environment and Quality Testing, Chongqing Chemical Industry Vocational College, Chongqing, China
| | - Lubing Yang
- Department of Pharmacy, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Jiangli Chen
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xuyan Zhou
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Zhidong Qiu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Zhidong Qiu, ; Ye Jin,
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Zhidong Qiu, ; Ye Jin,
| |
Collapse
|
9
|
Xu X, Jia L, Ma X, Li H, Sun C. Application Potential of Plant-Derived Medicines in Prevention and Treatment of Platinum-Induced Peripheral Neurotoxicity. Front Pharmacol 2022; 12:792331. [PMID: 35095502 PMCID: PMC8793340 DOI: 10.3389/fphar.2021.792331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/23/2021] [Indexed: 11/23/2022] Open
Abstract
As observed with other chemotherapeutic agents, the clinical application of platinum agents is a double-edged sword. Platinum-induced peripheral neuropathy (PIPN) is a common adverse event that negatively affects clinical outcomes and patients’ quality of life. Considering the unavailability of effective established agents for preventing or treating PIPN and the increasing population of cancer survivors, the identification and development of novel, effective interventions are the need of the hour. Plant-derived medicines, recognized as ideal agents, can not only help improve PIPN without affecting chemotherapy efficacy, but may also produce synergy. In this review, we present a brief summary of the mechanisms of platinum agents and PIPN and then focus on exploring the preventive or curative effects and underlying mechanisms of plant-derived medicines, which have been evaluated under platinum-induced neurotoxicity conditions. We identified 11 plant extracts as well as 17 plant secondary metabolites, and four polyherbal preparations. Their effects against PIPN are focused on oxidative stress and mitochondrial dysfunction, glial activation and inflammation response, and ion channel dysfunction. Also, ten clinical trials have assessed the effect of herbal products in patients with PIPN. The understanding of the molecular mechanism is still limited, the quality of clinical trials need to be further improved, and in terms of their efficacy, safety, and cost effectiveness studies have not provided sufficient evidence to establish a standard practice. But plant-derived medicines have been found to be invaluable sources for the development of natural agents with beneficial effects in the prevention and treatment of PIPN.
Collapse
Affiliation(s)
- Xiaowei Xu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqun Jia
- Oncology Department of Integrative Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoran Ma
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.,College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|
10
|
Lee R, Lee BH, Choi SH, Cho YJ, Cho HS, Kim HC, Rhim H, Cho IH, Rhee MH, Nah SY. Effects of Gintonin-enriched fraction on the gene expression of six lysophosphatidic receptor subtypes. J Ginseng Res 2021; 45:583-590. [PMID: 34803428 PMCID: PMC8587509 DOI: 10.1016/j.jgr.2021.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/17/2021] [Indexed: 11/03/2022] Open
Abstract
Background Gintonin, isolated from ginseng, acts as a ginseng-derived lysophosphatidic acid (LPA) receptor ligand and elicits the [Ca2+]i transient through six LPA receptor subtypes (LPARSs). However, the long-term effects of gintonin-enriched fraction (GEF) on the gene expression of six LPARSs remain unknown. We examined changes in the gene expression of six LPA receptors in the mouse whole brain, heart, lungs, liver, kidneys, spleen, small intestine, colon, and testis after long-term oral GEF administration. Methods C57BL/6 mice were divided into two groups: control vehicle and GEF (100 mg/kg, p.o.). After 21-day saline or GEF treatment, total RNA was extracted from nine mouse organs. Quantitative-real-time PCR (qRT-PCR) and western blot were performed to quantify changes in the gene and protein expression of the six LPARSs, respectively. Results qRT-PCR analysis before GEF treatment revealed that the LPA6 RS was predominant in all organs except the small intestine. The LPA2 RS was most abundant in the small intestine. Long-term GEF administration differentially regulated the six LPARSs. Upon GEF treatment, the LPA6 RS significantly increased in the liver, small intestine, colon, and testis but decreased in the whole brain, heart, lungs, and kidneys. Western blot analysis of the LPA6 RS confirmed the differential effects of GEF on LPA6 receptor protein levels in the whole brain, liver, small intestine, and testis. Conclusion The LPA6 receptor was predominantly expressed in all nine organs examined; long-term oral GEF administration differentially regulated LPA3, LPA4, and LPA6 receptors in the whole brain, heart, lungs, liver, kidneys, small intestine, and testis.
Collapse
Affiliation(s)
- Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Byung-Hwan Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yeon-Jin Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Han-Sung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Man Hee Rhee
- Laboratory of Veterinary Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Dorman G, Flores I, Gutiérrez C, Castaño RF, Aldecoa M, Kim L. Medicinal herbs and nutritional supplements for dementia therapy: potential therapeutic targets and clinical evidence. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:26-51. [PMID: 34370647 DOI: 10.2174/1871527320666210809121230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/20/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Spices and herbs have been used for medicinal purposes for centuries. Also, in the last decades, the use of different nutritional supplements has been implemented to treat all kinds of diseases, including those that present an alteration in cognitive functioning. Dementia is a clinical syndrome in which a person's mental and cognitive capacities gradually decline. As the disease progresses, the person's autonomy diminishes. As there is not an effective treatment to prevent progressive deterioration in many of these pathologies, nutritional interventions have been, and still are, one of the most widely explored therapeutic possibilities. In this review, we have discussed a great number of potentially interesting plants, nutritional derivatives and probiotics for the treatment of dementia around the world. Their action mechanisms generally involve neuroprotective effects via anti-inflammatory, antioxidant, anti-apoptotic, b-amyloid and tau anti-aggregate actions; brain blood flow improvement, and effects on synaptic cholinergic and dopaminergic neurotransmission, which may optimize cognitive performance in patients with cognitive impairment. As for their efficacy in patients with cognitive impairment and/or dementias, evidence is still scarce and/or their outcomes are controversial. We consider that many of these substances have promising therapeutic properties. Therefore, the scientific community has to continue with a more complete research focused on both identifying possible action mechanisms and carrying out clinical trials, preferably randomized double-blind ones, with a greater number of patients, a long-term follow-up, dose standardization and the use of current diagnosis criteria.
Collapse
Affiliation(s)
- Guido Dorman
- Division of Neurology, Ramos Mejia Hospital. Argentina
| | - Ignacio Flores
- Neuroscience Institute, Favaloro Foundation Hospital. Argentina
| | | | | | - Mayra Aldecoa
- Division of Neurology, Ramos Mejia Hospital. Argentina
| | - Leandro Kim
- Division of Neurology, Ramos Mejia Hospital. Argentina
| |
Collapse
|
12
|
Choi SH, Lee R, Nam SM, Kim DG, Cho IH, Kim HC, Cho Y, Rhim H, Nah SY. Ginseng gintonin, aging societies, and geriatric brain diseases. Integr Med Res 2021; 10:100450. [PMID: 32817818 PMCID: PMC7426447 DOI: 10.1016/j.imr.2020.100450] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A dramatic increase in aging populations and low birth rates rapidly drive aging societies and increase aging-associated neurodegenerative diseases. However, functional food or medicinal formulations to prevent geriatric brain disorders are not readily available. Panax ginseng is a candidate, since ginseng has long-been consumed as a rejuvenating agent. However, the underlying molecular mechanisms and the components of ginseng that are responsible for brain rejuvenation and human longevity are unknown. Accumulating evidence shows that gintonin is a candidate for the anti-aging ingredient of ginseng, especially in brain senescence. METHODS Gintonin, a glycolipoprotein complex, contains three lipid-derived G protein-coupled receptor ligands: lysophosphatidic acids (LPAs), lysophosphatidylinositols (LPIs), and linoleic acid (LA). LPA, LPI, and LA act on six LPA receptor subtypes, GPR55, and GPR40, respectively. These G protein-coupled receptors are distributed within the nervous and non-nervous systems of the human body. RESULTS Gintonin-enriched fraction (GEF) exhibits anti-brain senescence and effects against disorders such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). Oral administration of gintonin in animal models of d-galactose-induced brain aging, AD, HD, and PD restored cognitive and motor functions. The underlying molecular mechanisms of gintonin-mediated anti-brain aging and anti-neurodegenerative diseases include neurogenesis, autophagy stimulation, anti-apoptosis, anti-oxidative stress, and anti-inflammatory activities. This review describes the characteristics of gintonin and GEF, and how gintonin exerts its effects on brain aging and brain associated-neurodegenerative diseases. CONCLUSION Finally, we describe how GEF can be applied to improve the quality of life of senior citizens in aging societies.
Collapse
Affiliation(s)
- Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sung Min Nam
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Do-Geun Kim
- Neurovascular Biology Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Yoonjeong Cho
- Center for Neuroscience Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Tian M, Li LN, Zheng RR, Yang L, Wang ZT. Advances on hormone-like activity of Panax ginseng and ginsenosides. Chin J Nat Med 2021; 18:526-535. [PMID: 32616193 DOI: 10.1016/s1875-5364(20)30063-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Indexed: 12/26/2022]
Abstract
Traditional Chinese medicine (TCM) has been paid much attentions due to the prevention and treatment of steroid hormone disorders. Ginseng, the root of Panax ginseng C. A. Meyer (Araliaceae), is one of the most valuable herbs in complementary and alternative medicines around the world. A series of dammarane triterpenoid saponins, also known as phytosteroids, were reported as the primary ingredients of Ginseng, and indicated broad spectral pharmacological actions, including anti-cancer, anti-inflammation and anti-fatigue. The skeletons of the dammarane triterpenoid aglycone are structurally similar to the steroid hormones. Both in vitro and in vivo studies showed that Ginseng and its active ingredients have beneficial hormone-like role in hormonal disorders. This review thus summarizes the structural similarities between hormones and dammarane ginsenosides and integrates the analogous effect of Ginseng and ginsenosides on prevention and treatment of hormonal disorders published in recent twenty years (1998-2018). The review may provide convenience for anticipate structure-function relationship between saponins structure and hormone-like effect.
Collapse
Affiliation(s)
- Mei Tian
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lin-Nan Li
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rui-Rong Zheng
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Yang
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zheng-Tao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
14
|
Verstraeten SL, Lorent JH, Mingeot-Leclercq MP. Lipid Membranes as Key Targets for the Pharmacological Actions of Ginsenosides. Front Pharmacol 2020; 11:576887. [PMID: 33041822 PMCID: PMC7518029 DOI: 10.3389/fphar.2020.576887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/18/2020] [Indexed: 12/27/2022] Open
Abstract
In this review, we will focus on the activity of ginsenosides on membranes and their related effects, from physicochemical, biophysical, and pharmacological viewpoints. Ginsenosides are a class of saponins with a large structural diversity and a wide range of pharmacological effects. These effects can at least partly be related to their activity on membranes which results from their amphiphilic character. Some ginsenosides are able to interact with membrane lipids and associate into nanostructures, making them possible adjuvants for vaccines. They are able to modulate membrane biophysical properties such as membrane fluidity, permeability or the formation of lateral domains with some degree of specificity towards certain cell types such as bacteria, fungi, or cancer cells. In addition, they have shown antioxidant properties which protect membranes from lipid oxidation. They further displayed some activity on membrane proteins either through direct or indirect interaction. We investigate the structure activity relationship of ginsenosides on membranes and discuss the implications and potential use as anticancer, antibacterial, and antifungal agents.
Collapse
Affiliation(s)
- Sandrine L Verstraeten
- Cellular & Molecular Pharmacology Unit (FACM), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), Brussels, Belgium
| | - Joseph H Lorent
- Cellular & Molecular Pharmacology Unit (FACM), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), Brussels, Belgium.,Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Marie-Paule Mingeot-Leclercq
- Cellular & Molecular Pharmacology Unit (FACM), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCL), Brussels, Belgium
| |
Collapse
|
15
|
Lee R, Lee NE, Choi SH, Nam SM, Kim HC, Rhim H, Cho IH, Hwang SH, Nah SY. Effects of gintonin-enriched fraction on hippocampal gene expressions. Integr Med Res 2020; 10:100475. [PMID: 33134079 PMCID: PMC7588706 DOI: 10.1016/j.imr.2020.100475] [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: 03/30/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023] Open
Abstract
Background Recently, gintonin and gintonin-enriched fraction (GEF) have been isolated from ginseng, a herbal medicine. Gintonin induces [Ca2+]i transition in cultured hippocampal neurons and stimulates acetylcholine release through LPA receptor activation. Oral administration of GEF is linked to hippocampus-dependent cognitive enhancement and other neuroprotective effects; however, effects of its long-term administration on hippocampal gene expression remains unknown. Here, we used next-generation sequence (NGS) analysis to examine changes in hippocampal gene expressions after long-term oral administration of GEF. Methods C57BL/6 mice were divided into three groups: control group, GEF50 (GEF 50 mg/kg, p.o.), and GEF100 (GEF 100 mg/kg, p.o.). After 22 days, total RNA was extracted from mouse hippocampal tissues. NGS was used for gene expression profiling; quantitative-real-time PCR and western blot were performed to quantify the changes in specific genes and to confirm the protein expression levels in treatment groups. Results NGS analysis screened a total of 23,282 genes, analyzing 11-related categories. We focused on the neurogenesis category, which includes four genes for candidate markers: choline acetyltransferase (ChAT) gene, β3-adrenergic receptor (Adrb3) gene, and corticotrophin-releasing hormone (Crh) gene, and tryptophan 2,3-dioxygenase (Tdo2) gene. Real-time PCR showed a marked overexpression of ChAT, Adrb3, and Crh genes, while reduced expression of Tdo2. Western blot analysis also confirmed increased ChAT and decreased Tdo2 protein levels. Conclusion We found that GEF affects mouse hippocampal gene expressions, associated with memory, cognitive, anti-stress and anti-anxiety functions, and neurodegeneration at differential degree, that might explain the genetic bases of GEF-mediated neuroprotective effects.
Collapse
Affiliation(s)
- Rami Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Na-Eun Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sung Min Nam
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuro Psychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Department of Science in Korean Medicine and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Karunasagara S, Hong GL, Park SR, Lee NH, Jung DY, Kim TW, Jung JY. Korean red ginseng attenuates hyperglycemia-induced renal inflammation and fibrosis via accelerated autophagy and protects against diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112693. [PMID: 32112899 DOI: 10.1016/j.jep.2020.112693] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/10/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax ginseng C.A. Mey. (Korean ginseng) has been widely used in traditional medicine to treat diabetes mellitus for thousands of years. It also plays a key role in health maintenance owing to its anti-oxidant and anti-fatigue properties, and is quite popular as a dietary supplement. AIM OF THE STUDY This study was designed to offer a complementary and alternative medicine to manage the diabetic kidney disease (DKD), which causes long-term damage to the renal structure. We also investigated the regulation of the autophagy mechanism, which is the underlying the pathogenesis of DKD. MATERIALS AND METHODS The effect of Korean red ginseng (KRG) on DKD was evaluated using human kidney proximal tubular cells and streptozotocin (STZ)-treated Sprague-Dawley rat models. In vitro experiments were conducted to evaluate the proteins related to fibrosis and autophagy. This was followed by in vivo experiments involving rats treated with single intraperitoneal administration of STZ (60 mg/kg) and then with KRG solution orally for 4 weeks. Proteins related to renal injury, fibrosis, and autophagy were determined by immunoblotting. Hematoxylin and eosin (H&E), Periodic acid-Schiff (PAS), Sirius red, and immunostaining were processed for histological studies. RESULTS KRG diminished the levels of metabolic measurements and blood parameters. Western blotting showed a decreased expression of proteins, such as TGF-β1, KIM1, and AGE, which are responsible for renal inflammation, injury, and fibrosis. Histological studies also supported these results and revealed that the KRG-treated groups recovered from renal injury and fibrosis. Furthermore, the autophagy marker, LC3, was upregulated, whereas p62 was downregulated. The levels of proteins related to the autophagy mechanism, such as ATG7, increased, while mammalian target of rapamycin (mTOR) decreased with the KRG treatment and exhibited accelerated autophagy compared to the STZ alone group. CONCLUSIONS KRG can suppress renal inflammation, injury, and fibrosis by blocking TGF-β1 activation and can induce cellular autophagy. Therefore, this study strongly suggests that KRG exhibits a renoprotective effect against the STZ-induced DKD.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Geum-Lan Hong
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Se-Ra Park
- Samsung Biomedical Research Institute, Seoul, 06351, Republic of Korea
| | - Na-Hyun Lee
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Da-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Tae-Won Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
17
|
Protopanaxadiol ginsenoside Rd protects against NMDA receptor-mediated excitotoxicity by attenuating calcineurin-regulated DAPK1 activity. Sci Rep 2020; 10:8078. [PMID: 32415270 PMCID: PMC7228936 DOI: 10.1038/s41598-020-64738-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroprotective strategies in the treatment of stroke have been attracting a great deal of attentions. Our previous clinical and basic studies have demonstrated that protopanaxadiol ginsenoside-Rd (Rd), a monomer compound extracted from Panax ginseng or Panax notoginseng, has neuroprotective effects against ischemic stroke, probably due to its ability to block Ca2+ overload, an usual consequence of the overactivation of NMDA receptor (NMDAR). As an extending study, we explored here whether Rd exerted its neuroprotection as a novel NMDAR blocker. Our whole-cell patch-clamp results showed that Rd reduced NMDAR currents of cultured rat cortical neurons (EC50 = 7.7 μM) dose-dependently by acting on extrasynaptic NMDAR NR2b subunit. However, unexpectedly, cell transfection and radioligand binding assays revealed that Rd did not bind to the NMDAR channel directly. Alternatively, it inhibited the phosphorylation of NR2b at Ser-1303, a target of death associated protein kinase 1 (DAPK1). Moreover, cell-based and cell-free enzymatic assays showed that Rd did not inhibit the activity of DAPK1 directly, but blocked the activity of calcineurin, a key phosphatase for activating DAPK1. Importantly, other protopanaxadiol ginsenosides were also found to have potential inhibitory effects on calcineurin activity. Furthermore, as expected, calcineurin inhibition by cyclosporin A could mimic Rd's effects and protect against NMDA-, oxygen glucose deprivation- or transient ischemic stroke-induced neuronal injury. Therefore, our present study provided the first evidence that Rd could exert an inhibitive effect on NMDAR-triggered currents and sequential excitotoxicity through mitigation of DAPK1-mediated NR2b phosphorylation by attenuating calcineurin activity.
Collapse
|
18
|
Huang-Pu-Tong-Qiao Formula Ameliorates Tau Phosphorylation by Inhibiting the CaM-CaMKIV Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8956071. [PMID: 34046075 PMCID: PMC8128057 DOI: 10.1155/2020/8956071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease. It is a chronic, lethal disease in which brain function is severely impaired and neuronal damage is irreversible. Huang-Pu-Tong-Qiao (HPTQ), a formula from traditional Chinese medicine, has been used in the clinical treatment of AD for many years, with remarkable effects. However, the neuroprotective mechanisms of HPTQ in AD have not yet been investigated. In the present study, we used AD models in vivo and in vitro, to investigate both the neuroprotective effect of HPTQ water extracts (HPTQ-W) and the potential mechanisms of this action. For the in vivo study, after HPTQ intervention, the Morris water maze test was used to examine learning and memory in rats. Transmission electron microscopy and immunofluorescence methods were then used to investigate neuronal damage. For the in vitro experiments, rat primary hippocampal neurons were cultured and cell viability was examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. Additionally, mRNA levels of CaM, CaMKK, CaMKIV, and tau were examined using qRT-PCR, and protein expression of CaM, CaMKK, p-CaMKIV, and p-tau were examined using western blot. In vivo, we revealed that HPTQ significantly improved learning and memory deficits and attenuated neuronal damage in the AD rat model. Furthermore, in vitro results showed that HPTQ significantly increased cell viability in the AD cell model. We also demonstrated that HPTQ significantly decreased the mRNA levels of CaM, CaMKK, CaMKIV, and tau and significantly decreased the protein expressions of CaM, CaMKK, p-CaMKIV, and p-tau. In conclusion, our results indicated that HPTQ improved cognition and ameliorated neuronal damage in AD models and implicated a reduction in tau phosphorylation caused by inhibition of the CaM-CaMKIV pathway as a possible mechanism.
Collapse
|
19
|
Nam SM, Choi JH, Choi SH, Cho HJ, Cho YJ, Rhim H, Kim HC, Cho IH, Kim DG, Nah SY. Ginseng gintonin alleviates neurological symptoms in the G93A-SOD1 transgenic mouse model of amyotrophic lateral sclerosis through lysophosphatidic acid 1 receptor. J Ginseng Res 2020; 45:390-400. [PMID: 34025132 PMCID: PMC8134849 DOI: 10.1016/j.jgr.2020.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 01/11/2023] Open
Abstract
Background We recently showed that gintonin, an active ginseng ingredient, exhibits antibrain neurodegenerative disease effects including multiple target mechanisms such as antioxidative stress and antiinflammation via the lysophosphatidic acid (LPA) receptors. Amyotrophic lateral sclerosis (ALS) is a spinal disease characterized by neurodegenerative changes in motor neurons with subsequent skeletal muscle paralysis and death. However, pathophysiological mechanisms of ALS are still elusive, and therapeutic drugs have not yet been developed. We investigate the putative alleviating effects of gintonin in ALS. Methods The G93A-SOD1 transgenic mouse ALS model was used. Gintonin (50 or 100 mg/kg/day, p.o.) administration started from week seven. We performed histological analyses, immunoblot assays, and behavioral tests. Results Gintonin extended mouse survival and relieved motor dysfunctions. Histological analyses of spinal cords revealed that gintonin increased the survival of motor neurons, expression of brain-derived neurotrophic factors, choline acetyltransferase, NeuN, and Nissl bodies compared with the vehicle control. Gintonin attenuated elevated spinal NAD(P) quinone oxidoreductase 1 expression and decreased oxidative stress-related ferritin, ionized calcium-binding adapter molecule 1-immunoreactive microglia, S100β-immunoreactive astrocyte, and Olig2-immunoreactive oligodendrocytes compared with the control vehicle. Interestingly, we found that the spinal LPA1 receptor level was decreased, whereas gintonin treatment restored decreased LPA1 receptor expression levels in the G93A-SOD1 transgenic mouse, thereby attenuating neurological symptoms and histological deficits. Conclusion Gintonin-mediated symptomatic improvements of ALS might be associated with the attenuations of neuronal loss and oxidative stress via the spinal LPA1 receptor regulations. The present results suggest that the spinal LPA1 receptor is engaged in ALS, and gintonin may be useful for relieving ALS symptoms.
Collapse
Affiliation(s)
- Sung Min Nam
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea.,Department of Anatomy, School of Medicine and Institute for Environmental Science, Wonkwang University, Iksan, Republic of Korea
| | - Jong Hee Choi
- Department of Science in Korean Medicine, Brain Korea 21 Plus Program, Department of Conversions Medical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hee-Jung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yeon-Jin Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Ik-Hyun Cho
- Department of Science in Korean Medicine, Brain Korea 21 Plus Program, Department of Conversions Medical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Do-Geun Kim
- Neurovascular Biology Laboratory, Department of Structure and Function of Neural Network, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Zhang Y, Wu F, Jia A, He T, Qu M, Xiao C, Cole J, Wang W, Qiu Z. Adsorption and desorption characteristics of ginsenosides from
Panax ginseng C. A. Meyer
on middle‐pressure chromatogram isolated gels. J Sep Sci 2020; 43:2436-2446. [DOI: 10.1002/jssc.201901050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Yan‐Fei Zhang
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Fa‐Jie Wu
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Ai‐Ling Jia
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Tian‐Zhu He
- School of Basic Medical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Mo Qu
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Chun‐Ping Xiao
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Janét Cole
- Department of PharmaceuticsCenter for Pharmaceutical Engineering and Sciences, School of PharmacyVirginia Commonwealth University Richmond Virginia 23298 USA
| | - Wei‐Nan Wang
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| | - Zhi‐Dong Qiu
- School of Pharmaceutical sciencesChangchun University of Chinese Medicine Changchun Jilin 130117 P. R. China
| |
Collapse
|
21
|
Kim MS, Jeon SJ, Youn SJ, Lee H, Park YJ, Kim DO, Kim BY, Kim W, Baik MY. Enhancement of Minor Ginsenosides Contents and Antioxidant Capacity of American and Canadian Ginsengs ( Panax quinquefolius) by Puffing. Antioxidants (Basel) 2019; 8:antiox8110527. [PMID: 31694256 PMCID: PMC6912307 DOI: 10.3390/antiox8110527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/02/2019] [Accepted: 11/04/2019] [Indexed: 01/04/2023] Open
Abstract
The effects of puffing on ginsenosides content and antioxidant activities of American and Canadian ginsengs, Panax quinquefolius, were investigated. American and Canadian ginsengs puffed at different pressures were extracted using 70% ethanol. Puffing formed a porous structure, inducing the efficient elution of internal compounds that resulted in significant increases in extraction yields and crude saponin content. The content of minor ginsenosides (Rg2, Rg3, compound K) increased with increasing puffing pressure, whereas that of major ginsenosides (Rg1, Re, Rf, Rb1, Rc, Rd) decreased, possibly due to their deglycosylation and pyrolysis. Furthermore, 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) radical scavenging activity, total phenolic content, total flavonoid content, amount of Maillard reaction products, and acidic polysaccharides content increased with increasing puffing pressure, but 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging activity did not. There was no substantial difference in the results between puffed American and Canadian ginsengs. Consequently, these results suggest that puffing can be a promising novel technology for processing P. quinquefolius to achieve higher levels of minor ginsenosides and obtain value-added products.
Collapse
Affiliation(s)
- Min-Soo Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
| | - Sung-Joon Jeon
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
| | - So Jung Youn
- Department of Food Engineering, Dankook University, Cheonan 31116, Korea
| | - Hyungjae Lee
- Department of Food Engineering, Dankook University, Cheonan 31116, Korea
| | - Young-Joon Park
- Department of Science in Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Dae-Ok Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
| | - Byung-Yong Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
| | - Wooki Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
- Correspondence: (W.K.); (M.-Y.B.); Tel.: +82-31-201-3482 (W.K.); +82-31-201-2625 (M.-Y.B.)
| | - Moo-Yeol Baik
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin 17104, Korea
- Correspondence: (W.K.); (M.-Y.B.); Tel.: +82-31-201-3482 (W.K.); +82-31-201-2625 (M.-Y.B.)
| |
Collapse
|
22
|
Antidepressant effects of ginsenoside Rf on behavioral change in the glial degeneration model of depression by reversing glial loss. J Ginseng Res 2019; 44:603-610. [PMID: 32617040 PMCID: PMC7322760 DOI: 10.1016/j.jgr.2019.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 07/08/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022] Open
Abstract
Background Depression is a common neuropsychiatric disease that shows astrocyte pathology. Ginsenoside Rf (G-Rf) is a saponin found in Panax ginseng which has been used to treat neuropsychiatric diseases. We aimed to investigate antidepressant properties of G-Rf when introduced into the L-alpha-aminoadipic acid (L-AAA)–infused mice model which is representative of a major depressive disorder that features diminished astrocytes in the brain. Methods L-AAA was infused into the prefrontal cortex (PFC) of mice to induce decrease of astrocytes. Mice were orally administered G-Rf (20 mg/kg) as well as vehicle only or imipramine (20 mg/kg) as controls. Depression-like behavior of mice was evaluated using forced swimming test (FST) and tail suspension test (TST). We observed recovery of astroglial impairment and increased proliferative cells in the PFC and its accompanied change in the hippocampus by Western blot and immunohistochemistry to assess the effect of G-Rf. Results After injection of L-AAA into the PFC, mice showed increased immobility time in FST and TST and loss of astrocytes without significant neuronal change in the PFC. G-Rf–treated mice displayed significantly more decreased immobility time in FST and TST than did vehicle-treated mice, and their immobility time almost recovered to those of the sham mice and imipramine-treated mice. G-Rf upregulated glial fibrillary acidic protein (GFAP) expression and Ki-67 expression in the PFC reduced by L-AAA and also alleviated astroglial change in the hippocampus. Conclusion G-Rf markedly reversed depression-like behavioral changes and exhibited protective effect against the astrocyte ablation in the PFC induced by L-AAA. These protective properties suggest that G-Rf might be a therapeutic agent for major depressive disorders.
Collapse
|
23
|
An X, Fu R, Ma P, Ma X, Fan D. Ginsenoside Rk1 inhibits cell proliferation and promotes apoptosis in lung squamous cell carcinoma by calcium signaling pathway. RSC Adv 2019; 9:25107-25118. [PMID: 35528653 PMCID: PMC9069870 DOI: 10.1039/c9ra05037j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/01/2019] [Indexed: 12/02/2022] Open
Abstract
Ginsenoside Rk1 (Rk1) is a rare saponin extracted from Sun Ginseng (SG) and has been shown to have an anti-tumor effect; however, the potential role of its in lung squamous cell carcinoma remains elusive. In this study, we investigated the anti-proliferative activity and involved mechanism of Rk1 against lung squamous cell carcinoma in vitro and in vivo. First, MTT assay, cell colony formation assay and cell cycle assay showed that Rk1 effectively inhibited cell proliferation and colony formation, and induced cell arrest at G1 phase. Following AV/PI staining, JC-10 staining, Western blot and immunohistochemistry indicated that Rk1 induced caspase-dependent apoptosis. In addition, Rk1 induced ER stress, causing the release of Ca2+, resulting in intracellular calcium and mitochondrial calcium overload. Intracellular calcium overload activated the calpain-caspase-12 and calpain-caspase-7-PARP pathways, while mitochondrial calcium overload caused mitochondrial membrane potential reduced, and the release of cytochrome c. BAPTA-AM (Ca2+ scavengers) and calpeptin (calpain inhibitors) significantly attenuated Rk1-induced apoptosis. Moreover, Rk1 significantly inhibited the growth of SK-MES-1 xenograft tumors with low toxic side effects. In summary, this study for the first time demonstrated that Rk1 had significant antitumor effects against lung squamous cell carcinoma and great potential to serve as a novel anticancer agent.
Collapse
Affiliation(s)
- Xining An
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China +86-29-8830-5118 +86-29-8830-5118
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
- Biotech. & Biomed. Research Institute, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
| | - Rongzhan Fu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China +86-29-8830-5118 +86-29-8830-5118
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
- Biotech. & Biomed. Research Institute, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
| | - Pei Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China +86-29-8830-5118 +86-29-8830-5118
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
- Biotech. & Biomed. Research Institute, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
| | - Xiaoxuan Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China +86-29-8830-5118 +86-29-8830-5118
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
- Biotech. & Biomed. Research Institute, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China +86-29-8830-5118 +86-29-8830-5118
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
- Biotech. & Biomed. Research Institute, Northwest University 229 North Taibai Road Xi'an Shaanxi 710069 China
| |
Collapse
|
24
|
Shin SJ, Jeon SG, Kim JI, Jeong YO, Kim S, Park YH, Lee SK, Park HH, Hong SB, Oh S, Hwang JY, Kim HS, Park H, Nam Y, Lee YY, Kim JJ, Park SH, Kim JS, Moon M. Red Ginseng Attenuates Aβ-Induced Mitochondrial Dysfunction and Aβ-mediated Pathology in an Animal Model of Alzheimer's Disease. Int J Mol Sci 2019; 20:E3030. [PMID: 31234321 PMCID: PMC6627470 DOI: 10.3390/ijms20123030] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/05/2019] [Accepted: 06/19/2019] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and is characterized by neurodegeneration and cognitive deficits. Amyloid beta (Aβ) peptide is known to be a major cause of AD pathogenesis. However, recent studies have clarified that mitochondrial deficiency is also a mediator or trigger for AD development. Interestingly, red ginseng (RG) has been demonstrated to have beneficial effects on AD pathology. However, there is no evidence showing whether RG extract (RGE) can inhibit the mitochondrial deficit-mediated pathology in the experimental models of AD. The effects of RGE on Aβ-mediated mitochondrial deficiency were investigated in both HT22 mouse hippocampal neuronal cells and the brains of 5XFAD Aβ-overexpressing transgenic mice. To examine whether RGE can affect mitochondria-related pathology, we used immunohistostaining to study the effects of RGE on Aβ accumulation, neuroinflammation, neurodegeneration, and impaired adult hippocampal neurogenesis in hippocampal formation of 5XFAD mice. In vitro and in vivo findings indicated that RGE significantly improves Aβ-induced mitochondrial pathology. In addition, RGE significantly ameliorated AD-related pathology, such as Aβ deposition, gliosis, and neuronal loss, and deficits in adult hippocampal neurogenesis in brains with AD. Our results suggest that RGE may be a mitochondria-targeting agent for the treatment of AD.
Collapse
Affiliation(s)
- Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Jin-Il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Korea.
| | - Yu-On Jeong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Seong-Kyung Lee
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Hyun Ha Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Sua Oh
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Ji-Young Hwang
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - HyunHee Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro, Shinseong-dong, Yuseong-gu, Daejeon 34128, Korea.
| | - Jwa-Jin Kim
- Department of Nephrology, School of Medicine, Chungnam National University, Daejeon 35015, Korea.
| | - Sun-Hyun Park
- R&D center for Advanced Pharmaceuticals & Evaluation, Korea Institute of toxicology, 141, Gajeong-ro, Yuseong-gu, Daejeon 34114, Korea.
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon 35365, Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Korea.
| |
Collapse
|
25
|
Lee NE, Park SD, Hwang H, Choi SH, Lee RM, Nam SM, Choi JH, Rhim H, Cho IH, Kim HC, Hwang SH, Nah SY. Effects of a gintonin-enriched fraction on hair growth: an in vitro and in vivo study. J Ginseng Res 2019; 44:168-177. [PMID: 32095099 PMCID: PMC7033365 DOI: 10.1016/j.jgr.2019.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/18/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022] Open
Abstract
Background Ginseng has been widely used as a health-promoting tonic. Gintonin present in ginseng acts as a lysophosphatidic acid (LPA) receptor ligand that activates six LPA receptor subtypes. The LPA6 subtype plays a key role in normal hair growth, and mutations in the LPA6 receptor impair normal human hair growth. Currently, human hair loss and alopecia are concerning issues that affect peoples' social and day-to-day lives. Objective We investigated the in vitro and in vivo effects of a gintonin-enriched fraction (GEF) on mouse hair growth. Methods Human hair follicle dermal papilla cells (HFDPCs) and six-week-old male C57BL/6 mice were used. The mice were divided into the four groups: control, 1% minoxidil, 0.75% GEF, and 1.5% GEF. The dorsal hair was removed to synchronize the telogen phase. Each group was treated topically, once a day, for 15 days. We analyzed hair growth activity and histological changes. Results GEF induced transient [Ca2+]i, which stimulated HFDPC proliferation and caused 5-bromo-2'-deoxyuridine (BrdU) incorporation in a concentration-dependent manner. GEF-mediated HFDPC proliferation was blocked by the LPA receptor antagonist and Ca2+ chelator. HFDPC treatment with GEF stimulated vascular endothelial growth factor release. Topical application of GEF and minoxidil promoted hair growth in a dose-dependent manner. Histological analysis showed that GEF and minoxidil increased the number of hair follicles and hair weight. Conclusion Topical application of GEF promotes mouse hair growth through HFDPC proliferation. GEF could be one of the main components of ginseng that promote hair growth and could be used to treat human alopecia.
Collapse
Affiliation(s)
- Na-Eun Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sang-Deuk Park
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hongik Hwang
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ra Mi Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sung Min Nam
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jong Hee Choi
- Department of Convergence Medical Science, Department of Science in Korean Medicine, and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Department of Science in Korean Medicine, and Brain Korea 21 Plus Program, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Li G, Zhang N, Geng F, Liu G, Liu B, Lei X, Li G, Chen X. High-throughput metabolomics and ingenuity pathway approach reveals the pharmacological effect and targets of Ginsenoside Rg1 in Alzheimer's disease mice. Sci Rep 2019; 9:7040. [PMID: 31065079 PMCID: PMC6504884 DOI: 10.1038/s41598-019-43537-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/23/2019] [Indexed: 01/23/2023] Open
Abstract
Ginsenoside Rg1, a natural triterpenoid saponins compound isolated from the Panax species, has been found to possess neuroprotective properties in neurodegenerative diseases such as Alzheimer's disease (AD). However, its pharmacological mechanism on AD has not been studied. In this study, an ultra-performance liquid chromatography combined with quadrupole time of-flight mass spectrometry (UPLC-Q/TOF-MS) based non-targeted metabolomics strategy was performed to explore the mechanism of Ginsenoside Rg1 protecting against AD mice by characterizing metabolic biomarkers and regulation pathways changes. A total of nineteen potential metabolites in serum were discovered and identified to manifest the difference between wild-type mice and triple transgenic mice in control and model group, respectively. Fourteen potential metabolites involved in ten metabolic pathways such as linoleic acid metabolism, arachidonic acid metabolism, tryptophan metabolism and sphingolipid metabolism were affected by Rg1. From the ingenuity pathway analysis (IPA) platform, the relationship between gene, protein, metabolites alteration and protective activity of ginsenoside Rg1 in AD mice are deeply resolved, which refers to increased level of albumin, amino acid metabolism and molecular transport. In addition, quantitative analysis of key enzymes in the disturbed pathways by proteomics parallel reaction was employed to verify changed metabolic pathway under Ginsenoside Rg1. The UPLC-Q/TOF-MS based serum metabolomics method brings about new insights into the pharmacodynamic studies of Ginsenoside Rg1 on AD mice.
Collapse
Affiliation(s)
- Ge Li
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Xuanwei Avenue 138, Jinghong City, 666100, Yunnan Province, China
| | - Ning Zhang
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Guanghua Street 39, Qianjin District, Jiamusi City, 154007, Heilongjiang Province, China
| | - Fang Geng
- College of Chemistry & Chemical Engineering, Harbin Normal University, Shida Road No. 1, Limin Economic Development Zone, Harbin, 150025, Heilongjiang Province, China
| | - Guoliang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Bin Liu
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Guanghua Street 39, Qianjin District, Jiamusi City, 154007, Heilongjiang Province, China
| | - Xia Lei
- College of Jiamusi, Heilongjiang University of Chinese Medicine, Jiamusi, Guanghua Street 39, Qianjin District, Jiamusi City, 154007, Heilongjiang Province, China
| | - Guang Li
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Xuanwei Avenue 138, Jinghong City, 666100, Yunnan Province, China
| | - Xi Chen
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Xuanwei Avenue 138, Jinghong City, 666100, Yunnan Province, China.
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
27
|
Reay JL, van Schaik P, Wilson CJ. A systematic review of research investigating the physiological and psychological effects of combining Ginkgo biloba and Panax ginseng into a single treatment in humans: Implications for research design and analysis. Brain Behav 2019; 9:e01217. [PMID: 30729756 PMCID: PMC6422825 DOI: 10.1002/brb3.1217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 12/17/2018] [Accepted: 12/22/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The traditional herbal supplements Panax ginseng and Ginkgo biloba are self-medicated by members of the general public and prescribed by healthcare professionals in some EU countries for numerous health complaints. Clinical evidence is mixed and mechanisms of action are not fully understood. There is clinical interest into the synergistic effects of combining both herbs. METHODS We systematically review the literature investigating the effects of combination treatments on physiological and psychological outcomes in humans. We identified all studies meeting inclusion criteria: (a) written in English; (b) peer-reviewed; (c) conducted in humans; (d) including either a proprietary Panax ginseng/Ginkgo biloba treatment or a study preparation containing both; (e) placebo-controlled; (f) utilizing standardized extracts. We critically discuss each trial; calculate standardized effect sizes where possible and provide recommendations for research design and analysis. RESULTS Eight studies were identified and all investigated a proprietary combination treatment, Gincosan® . Studies are of high quality and robust; however, practice effects, choice of statistical model, and reliance upon null-hypothesis significance testing hinder generalized estimates of effect. The most consistent results are benefits to aspects of the circulatory/cardiovascular system in patient populations and "secondary memory" performance in patient and healthy populations. Two studies demonstrate synergy in healthy populations following a single dose; however, synergy in patient populations and following repeated dosing has not yet been directly tested. CONCLUSIONS A Panax ginseng and Ginkgo biloba combination treatment can improve aspects of physiological and cognitive function in humans; however, evidence for synergy requires further investigation and future research should directly investigate synergy following repeated dosing.
Collapse
Affiliation(s)
- Jonathon Lee Reay
- Department of Psychology, School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| | - Paul van Schaik
- Department of Psychology, School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| | - Christopher James Wilson
- Department of Psychology, School of Social Sciences, Humanities and Law, Teesside University, Middlesbrough, UK
| |
Collapse
|
28
|
Majdi Seghinsara A, Shoorei H, Hassanzadeh Taheri MM, Khaki A, Shokoohi M, Tahmasebi M, Khaki AA, Eyni H, Ghorbani S, Riahi Rad KH, Kalarestaghi H, Roshangar L. Panax ginseng Extract Improves Follicular Development after Mouse Preantral Follicle 3D Culture. CELL JOURNAL 2019; 21:210-219. [PMID: 30825295 PMCID: PMC6397605 DOI: 10.22074/cellj.2019.5733] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Abstract
Objective Panax ginseng is a popular traditional herb that has been used in complementary and alternative medicine
in eastern Asia, and it possesses pharmacologically active compounds like ginsenosides (GSs). This study aimed to
investigate the impact of Panax ginseng extract (PGE) at different concentrations on in vitro follicular function and
development in a three-dimensional (3D) culture system fabricated using sodium alginate after 12 days of culture.
Materials and Methods In this experimental study, preantral follicles (n=661) were mechanically isolated from the
ovaries of 14-day-old female NMRI mice using 29-gauge insulin syringes. Follicles were individually capsulated within
sodium alginate, and divided into four groups including control and experimental groups 1, 2, and 3. Then, they were
cultured for 12 days in the medium supplemented with different concentrations of PGE (0, 50, 100, and 500 µg/
mL, for control groups and groups 1, 2 and 3, respectively). At the end of the culture period, the mean diameter and
maturation of follicles, follicular steroid production, mRNA expression level of proliferating cell nuclear antigen (PCNA)
and follicle stimulating hormone receptor (FSH-R), and reactive oxygen species (ROS) levels in collected metaphase-II
(MII) oocytes were determined.
Results The mean diameter of follicles in group 2 was significantly increased as compared to other groups (P<0.001).
The percentages of the survival and maturation rate and levels of secreted hormones were higher in group 2 than
the other groups (P<0.05). Follicles cultured in the presence of PGE 100 µg/mL had higher levels of proliferation cell
nuclear antigen (PCNA) and follicle stimulating hormone receptor (FSH-R) mRNA expression in comparison to other
groups (P<0.05). Moreover, oocytes collected from groups 2 and 3 had lower levels of ROS as compared to other
groups (P<0.05).
Conclusion Our results suggest that PGE at the concentration of 100 µg/mL induces higher follicular function and
development in the 3D culture system.
Collapse
Affiliation(s)
- Abbas Majdi Seghinsara
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. Electronic Address:
| | | | - Arash Khaki
- Department of Pathology, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Majid Shokoohi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moloud Tahmasebi
- Department of Anatomical Sciences, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amir Afshin Khaki
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Eyni
- Department of Anatomical Sciences, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Ghorbani
- Department of Anatomical Sciences, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran
| | - K Hadijeh Riahi Rad
- Department of Horticulture Science, Tarbiat Modares University, Tehran, Iran
| | - Hossein Kalarestaghi
- Research laboratory for Embryology and Stem Cells, Department of Anatomical Sciences and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Dhuna K, Felgate M, Bidula SM, Walpole S, Bibic L, Cromer BA, Angulo J, Sanderson J, Stebbing MJ, Stokes L. Ginsenosides Act As Positive Modulators of P2X4 Receptors. Mol Pharmacol 2018; 95:210-221. [PMID: 30545933 PMCID: PMC6334005 DOI: 10.1124/mol.118.113696] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022] Open
Abstract
We investigated the selectivity of protopanaxadiol ginsenosides from Panax ginseng acting as positive allosteric modulators on P2X receptors. ATP-induced responses were measured in stable cell lines overexpressing human P2X4 using a YOPRO-1 dye uptake assay, intracellular calcium measurements, and whole-cell patch-clamp recordings. Ginsenosides CK and Rd were demonstrated to enhance ATP responses at P2X4 by ∼twofold, similar to potentiation by the known positive modulator ivermectin. Investigations into the role of P2X4 in mediating a cytotoxic effect showed that only P2X7 expression in HEK-293 cells induces cell death in response to high concentrations of ATP, and that ginsenosides can enhance this process. Generation of a P2X7-deficient clone of BV-2 microglial cells using CRISPR/Cas9 gene editing enabled an investigation of endogenous P2X4 in a microglial cell line. Compared with parental BV-2 cells, P2X7-deficient BV-2 cells showed minor potentiation of ATP responses by ginsenosides, and insensitivity to ATP− or ATP+ ginsenoside-induced cell death, indicating a primary role for P2X7 receptors in both of these effects. Computational docking to a homology model of human P2X4, based on the open state of zfP2X4, yielded evidence of a putative ginsenoside binding site in P2X4 in the central vestibule region of the large ectodomain.
Collapse
Affiliation(s)
- Kshitija Dhuna
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Matthew Felgate
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Stefan M Bidula
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Samuel Walpole
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Lucka Bibic
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Brett A Cromer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Jesus Angulo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Julie Sanderson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Martin J Stebbing
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| | - Leanne Stokes
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia (K.D., B.A.C., M.J.S., L.S.); School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, United Kingdom (M.F., S.M.B., S.W., L.B., J.A., J.S., L.S.); Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia (B.A.C.); and Florey Institute of Neuroscience and Mental Health, Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia (M.J.S.)
| |
Collapse
|
30
|
Kim HJ, Jung SW, Kim SY, Cho IH, Kim HC, Rhim H, Kim M, Nah SY. Panax ginseng as an adjuvant treatment for Alzheimer's disease. J Ginseng Res 2018; 42:401-411. [PMID: 30337800 PMCID: PMC6190533 DOI: 10.1016/j.jgr.2017.12.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 11/29/2017] [Accepted: 12/12/2017] [Indexed: 01/22/2023] Open
Abstract
Longevity in medicine can be defined as a long life without mental or physical deficits. This can be prevented by Alzheimer's disease (AD). Current conventional AD treatments only alleviate the symptoms without reversing AD progression. Recent studies demonstrated that Panax ginseng extract improves AD symptoms in patients with AD, and the two main components of ginseng might contribute to AD amelioration. Ginsenosides show various AD-related neuroprotective effects. Gintonin is a newly identified ginseng constituent that contains lysophosphatidic acids and attenuates AD-related brain neuropathies. Ginsenosides decrease amyloid β-protein (Aβ) formation by inhibiting β- and γ-secretase activity or by activating the nonamyloidogenic pathway, inhibit acetylcholinesterase activity and Aβ-induced neurotoxicity, and decrease Aβ-induced production of reactive oxygen species and neuroinflammatory reactions. Oral administration of ginsenosides increases the expression levels of enzymes involved in acetylcholine synthesis in the brain and alleviates Aβ-induced cholinergic deficits in AD models. Similarly, gintonin inhibits Aβ-induced neurotoxicity and activates the nonamyloidogenic pathway to reduce Aβ formation and to increase acetylcholine and choline acetyltransferase expression in the brain through lysophosphatidic acid receptors. Oral administration of gintonin attenuates brain amyloid plaque deposits, boosting hippocampal cholinergic systems and neurogenesis, thereby ameliorating learning and memory impairments. It also improves cognitive functions in patients with AD. Ginsenosides and gintonin attenuate AD-related neuropathology through multiple routes. This review focuses research demonstrating that ginseng constituents could be a candidate as an adjuvant for AD treatment. However, clinical investigations including efficacy and tolerability analyses may be necessary for the clinical acceptance of ginseng components in combination with conventional AD drugs.
Collapse
Key Words
- AChE, acetylcholinesterase
- AD, Alzheimer's disease
- APP, amyloid precursor protein
- Adjuvant
- Alzheimer's disease
- Aβ, amyloid β-protein
- BDNF, brain-derived neurotrophic factor
- EGF, Epidermal growth factor
- GLP151, ginseng major latex-like protein 151
- Ginsenoside
- Gintonin
- LPA, Lysophosphatidic acid
- NGF, nerve growth factor
- NMDA, n-methyl-d-aspartic acid
- PI3K, phosphoinositide-3 kinase
- PPARγ, peroxisome proliferator-activated receptor-γ
- Panax ginseng
- ROS, reactive oxygen species
- sAPPα, soluble amyloid precursor protein α
Collapse
Affiliation(s)
- Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seok-Won Jung
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seog-Young Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine and Institute of Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Manho Kim
- Department of Neurology, Neuroscience Research Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Ginsenosides: the need to move forward from bench to clinical trials. J Ginseng Res 2018; 43:361-367. [PMID: 31308807 PMCID: PMC6606839 DOI: 10.1016/j.jgr.2018.09.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 12/02/2022] Open
Abstract
Panax ginseng, known as Koran ginseng, one of the most commonly used traditional plants, has been demonstrated to show a wide range of pharmacological applications. Ginsenosides are the major active ingredients found in ginseng and are responsible for the biological and pharmacological activities, such as antioxidation, antiinflammation, vasorelaxation, and anticancer actions. Existing studies have mostly focused on identifying and purifying single ginsenosides and investigating pharmacological activities and molecular mechanisms in cells and animal models. However, ginsenoside studies based on clinical trials have been very limited. Therefore, this review aimed to discuss the currently available clinical trials on ginsenosides and provide insights and future directions for developing ginsenosides as efficacious and safe drugs for human disease.
Collapse
|
32
|
A Comprehensive In Vivo and In Vitro Assessment of the Drug Interaction Potential of Red Ginseng. Clin Ther 2018; 40:1322-1337. [PMID: 30078466 DOI: 10.1016/j.clinthera.2018.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 11/20/2022]
Abstract
Purpose: Red ginseng is one of the world's most popular herbal medicines; it exhibits a wide range of pharmacologic activities and is often co-ingested with other herbal and conventional medicines. This open-label, randomized, 3-period study investigated the in vivo herb-drug interaction potential for red ginseng extract with cytochrome P-450 (CYP) enzymes and organic anion-transporting polypeptide (OATP) 1B1. METHODS Fifteen healthy male volunteers (22-28 years; 57.1-80.8 kg) were administered a single dose of cocktail probe substrates (caffeine 100 mg, losartan 50 mg, omeprazole 20 mg, dextromethorphan 30 mg, midazolam 2 mg, and pitavastatin 2 mg) and single or multiple doses of red ginseng extract for 15 days. FINDINGS The pharmacokinetic profiles of the probe substrates and metabolites after single- or multiple-dose administration of red ginseng extracts were comparable to the corresponding profiles of the control group. The geometric mean ratio of AUC0-t and 90% CIs for the probe substrate drugs between the control and multiple doses of red ginseng for 15 days were within 0.8 to 1.25 (CYP2C9, CYP3A4, and OATP1B1 probe substrates) or slightly higher (CYP1A2, CYP2C19, and CYP2D6 probe substrates). Additional assessments of the in vitro drug interaction potential of red ginseng extracts and the ginsenoside Rb1 on drug-metabolizing enzymes and transporters using human liver microsomes, cryopreserved human hepatocytes, and transporter-overexpressed cells were negative. IMPLICATIONS Red ginseng poses minimal risks for clinically relevant CYP- or OATP-mediated drug interactions and is well tolerated. Clinical Research Information Service registry no.
Collapse
|
33
|
Ning C, Gao X, Wang C, Kong Y, Liu Z, Sun H, Sun P, Huo X, Ma X, Meng Q, Liu K. Ginsenoside Rg1 protects against acetaminophen-induced liver injury via activating Nrf2 signaling pathway in vivo and in vitro. Regul Toxicol Pharmacol 2018; 98:58-68. [PMID: 30030101 DOI: 10.1016/j.yrtph.2018.07.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022]
Abstract
Acetaminophen (APAP) is a worldwide used drug for treating fever and pain. However, APAP overdose is the leading cause of drug-induced liver injury. The purpose of the current study is to evaluate the hepatoprotective effect of ginsenoside Rg1 (Rg1), the main pharmacologically active compounds of Panax ginseng, against APAP-induced acute liver injury, and further to elucidate the involvement of Nrf2 signaling pathway by in vivo and in vitro experiments. Male C57BL/6 mice were treated with Rg1 for 3 days before injection of APAP. Serum and liver tissue samples were collected 6 h later. The results indicated that Rg1 significantly attenuated APAP-induced hepatotoxicity and oxidative stress in a dose-dependent manner. Rg1 effectively enhanced antioxidant and detoxification capacity, which is largely dependent on up-regulating Nrf2 nuclear translocation, reducing Keap1 protein expression and up-regulating Nrf2 target genes including GCLC, GCLM, HO-1, NQO1, Ugt1a1, Ugt1a6, Ugt2b1, Sult2a1, Mrp2, Mrp3 and Mrp4. Furthermore, Rg1 repressed the activities of Cyp2e1, Cyp3a11, Cyp1a2, which are important enzymes in the formation of APAP toxic metabolite N-acetyl-p-benzoquinone imine. However, the changes in transporters and enzymes, as well as ameliorative liver histology induced by Rg1 were abrogated by Nrf2 antagonist all-transretinoic acid in vivo and Nrf2 siRNA in vitro. In conclusion, Rg1 produced hepatoprotective effects against APAP-induced acute liver injury via Nrf2 signaling pathway. Rg1 might be an effective approach for the prevention against acute liver injury.
Collapse
Affiliation(s)
- Chenqing Ning
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiaoguang Gao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Yulong Kong
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zhihao Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Pengyuan Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Xiaokui Huo
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Xiaodong Ma
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China.
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Key Laboratory of Pharmacokinetics and Transport of Liaoning Province, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
34
|
Kim DG, Jang M, Choi SH, Kim HJ, Jhun H, Kim HC, Rhim H, Cho IH, Nah SY. Gintonin, a ginseng-derived exogenous lysophosphatidic acid receptor ligand, enhances blood-brain barrier permeability and brain delivery. Int J Biol Macromol 2018; 114:1325-1337. [DOI: 10.1016/j.ijbiomac.2018.03.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
|
35
|
The anxiolytic-like effects of ginsenoside Rg3 on chronic unpredictable stress in rats. Sci Rep 2018; 8:7741. [PMID: 29773855 PMCID: PMC5958129 DOI: 10.1038/s41598-018-26146-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022] Open
Abstract
The present study is to evaluate the anxiolytic-like activities underlying ginsenoside Rg3 (GRg3). The anxiolytic-like activities were induced by GRg3 (20 and 40 mg/kg, i.g), evidenced by blocking the decreased time and entries in the open arms in elevated plus maze test and by reversing the increased latency to feed in novelty-suppressed feeding test. In addition, the decreased levels on progesterone, allopregnanolone, serotonin (5-HT) in the prefrontal cortex and hippocampus of chronic unpredictable stress (CUS) were blocked by GRg3 (20 and 40 mg/kg, i.g). Furthermore, the increased corticotropin releasing hormone, corticosterone and adrenocorticotropic hormone were blocked by GRg3 (20 and 40 mg/kg, i.g). Collectively, the anxiolytic-like effects produced by GRg3 were associated with the normalization of neurosteroids biosynthesis, serotonergic system as well as HPA axis dysfunction.
Collapse
|
36
|
Kong D, Tian X, Li Y, Zhang S, Cheng Y, Huo L, Ma H, Yang Z, Ren L, Zhang M, Zhang W. Revealing the Inhibitory Effect of Ginseng on Mitochondrial Respiration through Synaptosomal Proteomics. Proteomics 2018; 18:e1700354. [PMID: 29687596 DOI: 10.1002/pmic.201700354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/19/2018] [Indexed: 12/25/2022]
Abstract
Ginseng, the active ingredients of which are ginsenosides, is the most popular herbal medicine and has potential merit in the treatment of cerebral disorders. To better understand the function of Ginseng in the cerebral system, we examined changes in the protein expression profiles of synaptosomes extracted from the cerebral cortical and hippocampal tissues of rats administered a high or low dose of Ginseng for 2 weeks. More than 5000 proteins belonging to synaptosomes were simultaneously identified and quantitated by an approach combining tandem mass tags with 2D liquid chromatography-mass spectrometry (LC-MS). Regarding differentially expressed proteins, downregulated proteins were much more highly induced than upregulators in the cerebral cortical and hippocampal synaptosomes, regardless of the dose of Ginseng. Bioinformatic analysis indicated the majority of the altered proteins to be located in the mitochondria, directly or indirectly affecting mitochondrial oxidative respiration. Further functional experiments using the substrate-uncoupler inhibitor titration approach confirmed that three representative ginsenosides were able to inhibit oxidative phosphorylation in mitochondria. Our results demonstrate that Ginseng can regulate the function of mitochondria and alter the energy metabolism of cells, which may be useful for the treatment of central nervous disorders.
Collapse
Affiliation(s)
- Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Xiaolin Tian
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Yunshan Li
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Saihang Zhang
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Yiru Cheng
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Lifang Huo
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Huanhuan Ma
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Zuxiao Yang
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Leiming Ren
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| | - Mingquan Zhang
- Department of Basic Theory of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050017, P. R. China
| | - Wei Zhang
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, P. R. China
| |
Collapse
|
37
|
Kim HJ, Kim JY, Lee BH, Choi SH, Rhim H, Kim HC, Ahn SY, Jeong SW, Jang M, Cho IH, Nah SY. Gintonin, an exogenous ginseng-derived LPA receptor ligand, promotes corneal wound healing. J Vet Sci 2018; 18:387-397. [PMID: 27586470 PMCID: PMC5639092 DOI: 10.4142/jvs.2017.18.3.387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/03/2016] [Accepted: 08/26/2016] [Indexed: 01/25/2023] Open
Abstract
Ginseng gintonin is an exogenous ligand of lysophosphatidic acid (LPA) receptors. Accumulating evidence shows LPA helps in rapid recovery of corneal damage. The aim of this study was to evaluate the therapeutic efficacy of gintonin in a rabbit model of corneal damage. We investigated the signal transduction pathway of gintonin in human corneal epithelium (HCE) cells to elucidate the underlying molecular mechanism. We next evaluated the therapeutic effects of gintonin, using a rabbit model of corneal damage, by undertaking histochemical analysis. Treatment of gintonin to HCE cells induced transient increases of [Ca2+]i in concentration-dependent and reversible manners. Gintonin-mediated mobilization of [Ca2+]i was attenuated by LPA1/3 receptor antagonist Ki16425, phospholipase C inhibitor U73122, inositol 1,4,5-triphosphate receptor antagonist 2-APB, and intracellular Ca2+ chelator BAPTA-AM. Gintonin facilitated in vitro wound healing in a concentration-dependent manner. When applied as an eye-drop to rabbits with corneal damage, gintonin rapidly promoted recovery. Histochemical analysis showed gintonin decreased corneal apoptosis and increased corneal cell proliferation. We demonstrated that LPA receptor activation by gintonin is linked to in vitro and in vivo therapeutic effects against corneal damage. Gintonin can be applied as a clinical agent for the rapid healing of corneal damage.
Collapse
Affiliation(s)
- Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Joon Young Kim
- Veterinary Medical Teaching Hospital, Konkuk University, Seoul 05029, Korea
| | - Byung-Hwan Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Hyewon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and toxicology program, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Seoung-Yob Ahn
- Veterinary Medical Teaching Hospital, Konkuk University, Seoul 05029, Korea
| | - Soon-Wuk Jeong
- Veterinary Medical Teaching Hospital, Konkuk University, Seoul 05029, Korea
| | - Minhee Jang
- Department of Convergence Medical Science, College of Oriental Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Oriental Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
38
|
Xu L, Li Y, Dai Y, Peng J. Natural products for the treatment of type 2 diabetes mellitus: Pharmacology and mechanisms. Pharmacol Res 2018; 130:451-465. [PMID: 29395440 DOI: 10.1016/j.phrs.2018.01.015] [Citation(s) in RCA: 278] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 02/06/2023]
Abstract
Epidemiological studies have implied that diabetes mellitus (DM) will become an epidemic accompany with metabolic and endocrine disorders worldwide. Most of DM patients are affected by type 2 diabetes mellitus (T2DM) with insulin resistance and insulin secretion defect. Generally, the strategies to treat T2DM are diet control, moderate exercise, hypoglycemic and lipid-lowing agents. Despite the therapeutic benefits for the treatment of T2DM, most of the drugs can produce some undesirable side effects. Considering the pathogenesis of T2DM, natural products (NPs) have become the important resources of bioactive agents for anti-T2DM drug discovery. Recently, more and more natural components have been elucidated to possess anti-T2DM properties, and many efforts have been carried out to elucidate the possible mechanisms. The aim of this paper was to overview the activities and underlying mechanisms of NPs against T2DM. Developments of anti-T2DM agents will be greatly promoted with the increasing comprehensions of NPs for their multiple regulating effects on various targets and signal pathways.
Collapse
Affiliation(s)
- Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yue Li
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Dai
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
39
|
Tewari D, Stankiewicz AM, Mocan A, Sah AN, Tzvetkov NT, Huminiecki L, Horbańczuk JO, Atanasov AG. Ethnopharmacological Approaches for Dementia Therapy and Significance of Natural Products and Herbal Drugs. Front Aging Neurosci 2018; 10:3. [PMID: 29483867 PMCID: PMC5816049 DOI: 10.3389/fnagi.2018.00003] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022] Open
Abstract
Dementia is a clinical syndrome wherein gradual decline of mental and cognitive capabilities of an afflicted person takes place. Dementia is associated with various risk factors and conditions such as insufficient cerebral blood supply, toxin exposure, mitochondrial dysfunction, oxidative damage, and often coexisting with some neurodegenerative disorders such as Alzheimer's disease (AD), Huntington's disease (HD), and Parkinson's disease (PD). Although there are well-established (semi-)synthetic drugs currently used for the management of AD and AD-associated dementia, most of them have several adverse effects. Thus, traditional medicine provides various plant-derived lead molecules that may be useful for further medical research. Herein we review the worldwide use of ethnomedicinal plants in dementia treatment. We have explored a number of recognized databases by using keywords and phrases such as “dementia”, “Alzheimer's,” “traditional medicine,” “ethnopharmacology,” “ethnobotany,” “herbs,” “medicinal plants” or other relevant terms, and summarized 90 medicinal plants that are traditionally used to treat dementia. Moreover, we highlight five medicinal plants or plant genera of prime importance and discuss the physiological effects, as well as the mechanism of action of their major bioactive compounds. Furthermore, the link between mitochondrial dysfunction and dementia is also discussed. We conclude that several drugs of plant origin may serve as promising therapeutics for the treatment of dementia, however, pivotal evidence for their therapeutic efficacy in advanced clinical studies is still lacking.
Collapse
Affiliation(s)
- Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Nainital, India
| | - Adrian M Stankiewicz
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Andrei Mocan
- Department of Pharmaceutical Botany, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,ICHAT and Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Archana N Sah
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Nainital, India
| | - Nikolay T Tzvetkov
- Department of Molecular Biology and Biochemical Pharmacology, Institute of Molecular Biology Roumen Tsanev, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Lukasz Huminiecki
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Jarosław O Horbańczuk
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland.,Department of Pharmacognosy, University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Xie M, Liu J, Yan Z, Li X, Yang X, Jin H, Su A, Qin B. Bio-guided isolation of plant growth regulators from allelopathic plant-Codonopsis pilosula: phyto-selective activities and mechanisms. RSC Adv 2018; 8:13649-13655. [PMID: 35539311 PMCID: PMC9079819 DOI: 10.1039/c7ra12072a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 03/25/2018] [Indexed: 11/21/2022] Open
Abstract
Bio-guided isolation of the aerial waste part of typical allelopathic plant-Codonopsis pilosulaled to six active compounds being produced.
Collapse
Affiliation(s)
- Min Xie
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Jingkun Liu
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Zhiqiang Yan
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Xiuzhuang Li
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Xiaoyan Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Hui Jin
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| | - Anxiang Su
- Institute for the Control of Agrochemicals
- Ministry of Agriculture (ICAMA)
- Beijing
- PR China
| | - Bo Qin
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources
- Key Laboratory for Natural Medicine of Gansu Province
- Lanzhou Institute of Chemical Physics
- Chinese Academy of Sciences
- Lanzhou 730000
| |
Collapse
|
41
|
Cho HJ, Choi SH, Kim HJ, Lee BH, Rhim H, Kim HC, Hwang SH, Nah SY. Bioactive lipids in gintonin-enriched fraction from ginseng. J Ginseng Res 2017; 43:209-217. [PMID: 30962735 PMCID: PMC6437394 DOI: 10.1016/j.jgr.2017.11.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 02/08/2023] Open
Abstract
Background Ginseng is a traditional herbal medicine for human health. Ginseng contains a bioactive ligand named gintonin. The active ingredient of gintonin is lysophosphatidic acid C18:2 (LPA C18:2). We previously developed a method for gintonin-enriched fraction (GEF) preparation to mass-produce gintonin from ginseng. However, previous studies did not show the presence of other bioactive lipids besides LPAs. The aim of this study was to quantify the fatty acids, lysophospholipids (LPLs), and phospholipids (PLs) besides LPAs in GEF. Methods We prepared GEF from white ginseng. We used gas chromatography-mass spectrometry for fatty acid analysis and liquid chromatography-tandem mass spectrometry for PL analysis, and quantified the fatty acids, LPLs, and PLs in GEF using respective standards. We examined the effect of GEF on insulin secretion in INS-1 cells. Results GEF contains about 7.5% linoleic (C18:2), 2.8% palmitic (C16:0), and 1.5% oleic acids (C18:1). GEF contains about 0.2% LPA C18:2, 0.06% LPA C16:0, and 0.02% LPA C18:1. GEF contains 0.08% lysophosphatidylcholine, 0.03% lysophosphatidylethanolamine, and 0.13% lysophosphatidylinositols. GEF also contains about 1% phosphatidic acid (PA) 16:0-18:2, 0.5% PA 18:2-18:2, and 0.2% PA 16:0-18:1. GEF-mediated insulin secretion was not blocked by LPA receptor antagonist. Conclusion We determined four characteristics of GEF through lipid analysis and insulin secretion. First, GEF contains a large amount of linoleic acid (C18:2), PA 16:0-18:2, and LPA C18:2 compared with other lipids. Second, the main fatty acid component of LPLs and PLs is linoleic acid (C18:2). Third, GEF stimulates insulin secretion not through LPA receptors. Finally, GEF contains bioactive lipids besides LPAs.
Collapse
Affiliation(s)
- Hee-Jung Cho
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Sun-Hye Choi
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Byung-Hwan Lee
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyewon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Jiang S, Fang DF, Chen Y. Involvement of N-Methyl-D-Aspartic Acid Receptor and DL-α-Amino-3-Hydroxy-5- Methyl-4-Isoxazole Propionic Acid Receptor in Ginsenosides Rb1 and Rb3 against Oxygen-Glucose Deprivation-Induced Injury in Hippocampal Slices from Rat. Pharmacology 2017; 101:133-139. [PMID: 29207398 DOI: 10.1159/000481710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/22/2017] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Ginsenosides, Rb1 and Rb3, are the major protopanaxadiol components of ginseng saponin. In the present study, the influences of ginsenosides Rb1 and Rb3 on N-methyl-D-aspartic acid (NMDA) receptor or DL-α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor-mediated synaptic transmission after oxygen-glucose deprivation (OGD) were investigated. METHODS NMDA receptor population spike (NMDA-PS) or AMPA receptor-mediated population spike (AMPA-PS) was recorded in the CA1 pyramidal cell layer of rat hippocampal slices by electrophysiological techniques. RESULTS Under normal conditions, ginsenosides Rb3 and Rb1 depressed glutamate receptors-mediated synaptic transmission. Fourteen min of OGD resulted in a poor recovery amplitude of NMDA-PS or AMPA-PS after reoxygenation. Ginsenoside Rb3 significantly delayed the appearance of transient recovery of PS during OGD, and improved the recovery amplitudes of NMDA-PS and AMPA-PS after reoxygenation. However, the similar protective effects of ginsenoside Rb1 were observed only on NMDA-PS but not AMPA-PS. CONCLUSION These results suggest that ginsenosides Rb1 and Rb3 have the different inhibitions on NMDA and AMPA receptors-mediated response, which may partially explain the different protective effects of these agents on ischemic neuronal death.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Anatomy and Physiology, Lianyungang Branch of Traditional Chinese Medicine, Jiangsu Union Technical Institute, Lianyungang, China
| | - De-Fang Fang
- Department of Anatomy and Physiology, Lianyungang Branch of Traditional Chinese Medicine, Jiangsu Union Technical Institute, Lianyungang, China
| | - Ying Chen
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
43
|
Mitu SA, Bose U, Suwansa-Ard S, Turner LH, Zhao M, Elizur A, Ogbourne SM, Shaw PN, Cummins SF. Evidence for a Saponin Biosynthesis Pathway in the Body Wall of the Commercially Significant Sea Cucumber Holothuria scabra. Mar Drugs 2017; 15:E349. [PMID: 29112144 PMCID: PMC5706039 DOI: 10.3390/md15110349] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 01/31/2023] Open
Abstract
The sea cucumber (phylum Echinodermata) body wall is the first line of defense and is well known for its production of secondary metabolites; including vitamins and triterpenoid glycoside saponins that have important ecological functions and potential benefits to human health. The genes involved in the various biosynthetic pathways are unknown. To gain insight into these pathways in an echinoderm, we performed a comparative transcriptome analysis and functional annotation of the body wall and the radial nerve of the sea cucumber Holothuria scabra; to define genes associated with body wall metabolic functioning and secondary metabolite biosynthesis. We show that genes related to signal transduction mechanisms were more highly represented in the H. scabra body wall, including genes encoding enzymes involved in energy production. Eight of the core triterpenoid biosynthesis enzymes were found, however, the identity of the saponin specific biosynthetic pathway enzymes remains unknown. We confirm the body wall release of at least three different triterpenoid saponins using solid phase extraction followed by ultra-high-pressure liquid chromatography-quadrupole time of flight-mass spectrometry. The resource we have established will help to guide future research to explore secondary metabolite biosynthesis in the sea cucumber.
Collapse
Affiliation(s)
- Shahida Akter Mitu
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Utpal Bose
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
- CSIRO Agriculture and Food, St Lucia, Brisbane 4067, Queensland, Australia.
- School of Pharmacy, The University of Queensland, Brisbane 4067, Queensland, Australia.
| | - Saowaros Suwansa-Ard
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Luke H Turner
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Min Zhao
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Abigail Elizur
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Steven M Ogbourne
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| | - Paul Nicholas Shaw
- School of Pharmacy, The University of Queensland, Brisbane 4067, Queensland, Australia.
| | - Scott F Cummins
- Genecology Research Center, Faculty of Science, Health, Engineering and Education, University of the Sunshine Coast, Maroochydore DC 4558, Queensland, Australia.
| |
Collapse
|
44
|
Gardner A, Wu W, Thomson S, Zangerl-Plessl EM, Stary-Weinzinger A, Sanguinetti MC. Molecular Basis of Altered hERG1 Channel Gating Induced by Ginsenoside Rg3. Mol Pharmacol 2017; 92:437-450. [PMID: 28705808 DOI: 10.1124/mol.117.108886] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/12/2017] [Indexed: 11/22/2022] Open
Abstract
Outward current conducted by human ether-à-go-go-related gene type 1 (hERG1) channels is a major determinant of action potential repolarization in the human ventricle. Ginsenoside 20(S)-Rg3 [Rg3; (2S,3R,4S,5S,6R)-2-[(2R,3R,4S,5S,6R)-4,5-dihydroxy-2-[[(3S,5R,8R,9R,10R,12R,13R,14R,17S)-12-hydroxy-17-[(2S)-2-hydroxy-6-methylhept-5-en-2-yl]-4,4,8,10,14-pentamethyl-2,3,5,6,7,9,11,12,13,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-3-yl]oxy]-6-(hydroxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)oxane-3,4,5-triol], an alkaloid isolated from the root of Panax ginseng, slows the rate of hERG1 deactivation, induces channels to open at more negative potentials than normal, and increases current magnitude. The onset of Rg3 action is extremely fast, suggesting that it binds to an extracellular accessible site on the channel to alter its gating. Here we used a scanning mutagenesis approach to identify residues in the extracellular loops and transmembrane segments of hERG1 that might interact with Rg3. Single or multiple residues of hERG1 were mutated to Ala or Cys and the resulting mutant channels were heterologously expressed in Xenopus oocytes. The effects of Rg3 on the voltage dependence of activation and the deactivation rate of mutant channel currents were characterized using the two-microelectrode voltage clamp technique. Mutation to Ala of specific residues in the S1 (Tyr420), S2 (Leu452, Phe463), and S4 (Ile521, Lys525) segments partially inhibited the effects of Rg3 on hERG1. The double mutant Y420A/L452A nearly eliminated the effects of Rg3 on voltage-dependent channel gating but did not prevent the increase in current magnitude. These findings together with molecular modeling suggest that Rg3 alters the gating of hERG1 channels by interacting with and stabilizing the voltage sensor domain in an activated state.
Collapse
Affiliation(s)
- Alison Gardner
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| | - Wei Wu
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| | - Steven Thomson
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| | - Eva-Maria Zangerl-Plessl
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| | - Anna Stary-Weinzinger
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute (A.G., W.W., S.T., M.C.S.) and Division of Cardiovascular Medicine, Department of Internal Medicine, (M.C.S.), University of Utah, Salt Lake City, Utah; and Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria (E.-M.Z.-P., A.S.-W.)
| |
Collapse
|
45
|
Kochan E, Szymczyk P, Kuźma Ł, Lipert A, Szymańska G. Yeast Extract Stimulates Ginsenoside Production in Hairy Root Cultures of American Ginseng Cultivated in Shake Flasks and Nutrient Sprinkle Bioreactors. Molecules 2017; 22:molecules22060880. [PMID: 28587128 PMCID: PMC6152677 DOI: 10.3390/molecules22060880] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/23/2017] [Indexed: 12/26/2022] Open
Abstract
One of the most effective strategies to enhance metabolite biosynthesis and accumulation in biotechnological systems is the use of elicitation processes. This study assesses the influence of different concentrations of yeast extract (YE) on ginsenoside biosynthesis in Panax quinquefolium (American ginseng) hairy roots cultivated in shake flasks and in a nutrient sprinkle bioreactor after 3 and 7 days of elicitation. The saponin content was determined using HPLC. The maximum yield (20 mg g−1 d.w.) of the sum of six examined ginsenosides (Rb1, Rb2, Rc, Rd, Re and Rg1) in hairy roots cultivated in shake flasks was achieved after application of YE at 50 mg L−1 concentration and 3 day exposure time. The ginsenoside level was 1.57 times higher than that attained in control medium. The same conditions of elicitation (3 day time of exposure and 50 mg L−1 of YE) also favourably influenced the biosynthesis of studied saponins in bioreactor cultures. The total ginsenoside content was 32.25 mg g−1 d.w. and was higher than that achieved in control medium and in shake flasks cultures. Obtained results indicated that yeast extract can be used to increase ginsenoside production in hairy root cultures of P. quinquefolium.
Collapse
Affiliation(s)
- Ewa Kochan
- Pharmaceutical Biotechnology Department, Medical University of Lodz, Muszyńskiego 1, Lodz 90-151, Poland.
| | - Piotr Szymczyk
- Pharmaceutical Biotechnology Department, Medical University of Lodz, Muszyńskiego 1, Lodz 90-151, Poland.
| | - Łukasz Kuźma
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszyńskiego l, Lodz 90-151, Poland.
| | - Anna Lipert
- Department of Sports Medicine, Medical University of Lodz, Pomorska 251, Lodz 92-213, Poland.
| | - Grażyna Szymańska
- Pharmaceutical Biotechnology Department, Medical University of Lodz, Muszyńskiego 1, Lodz 90-151, Poland.
| |
Collapse
|
46
|
Ahuja A, Kim JH, Kim JH, Yi YS, Cho JY. Functional role of ginseng-derived compounds in cancer. J Ginseng Res 2017; 42:248-254. [PMID: 29983605 PMCID: PMC6026353 DOI: 10.1016/j.jgr.2017.04.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/18/2017] [Indexed: 12/19/2022] Open
Abstract
Ginseng is a natural product best known for its curative properties in diverse physiological processes such as cancer, neurodegenerative disorders, hypertension, and maintenance of hemostasis in the immune system. In previous decades, there have been some promising studies into the pharmacology and chemistry of ginseng components and the relationship between their structure and function. The emerging use of modified ginseng and development of new compounds from ginseng for clinical studies have been topics of study for many researchers. The present review deals with the anticancer, anti-inflammatory, antioxidant, and chemopreventive effects, and recent advances in microRNA technology related to red ginseng. The review also summarizes the current knowledge on the effect of ginsenosides in the treatment of cancer.
Collapse
Affiliation(s)
- Akash Ahuja
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Young-Su Yi
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
47
|
Guo S, Chen Y, Pang C, Wang X, Qi J, Mo L, Zhang H, An H, Zhan Y. Ginsenoside Rb1, a novel activator of the TMEM16A chloride channel, augments the contraction of guinea pig ileum. Pflugers Arch 2017; 469:681-692. [PMID: 28124133 DOI: 10.1007/s00424-017-1934-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/25/2016] [Accepted: 01/02/2017] [Indexed: 01/01/2023]
Abstract
Calcium-activated chloride channels (CaCCs) play important roles in many physiological processes, and the molecular basis of CaCCs has been identified as TMEM16A in many cell types. It is well established that TMEM16A is a drug target in many diseases, including cystic fibrosis, hypertension, asthma, and various tumors. Therefore, identifying potent and specific modulators of the TMEM16A channel is crucial. In this study, we identified the first natural activator of TMEM16A from traditional Chinese medicine and explored its mechanism. Our data showed that Ginsenoside Rb1 (GRb1) can activate TMEM16A directly from the intracellular side in a dose-dependent manner at an EC50 of 38.4 ± 2.14 μM. GRb1 specifically activated TMEM16A/B, but not the other previously proposed CaCC mediators such as CFTR and bestrophin. Moreover, GRb1 promoted proliferation of CHO cells stably expressing TMEM16A, in a concentration-dependent manner. Finally, we showed that GRb1 increased the amplitude and frequency of contractions in an isolated guinea pig ileum assay in vivo. In summary, GRb1 can be considered a lead compound for the development of novel drugs for the treatment of diseases caused by TMEM16A dysfunction.
Collapse
Affiliation(s)
- Shuai Guo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.,School of Electrical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Yafei Chen
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Chunli Pang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Xuzhao Wang
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Jinlong Qi
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Pharmacology and Toxicology for New Drug, Hebei Province, Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Li Mo
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China
| | - Hailin Zhang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of Pharmacology and Toxicology for New Drug, Hebei Province, Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.
| | - Yong Zhan
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.
| |
Collapse
|
48
|
El-Sheikh AAK, Kamel MY. Ginsenoside-Rb1 ameliorates lithium-induced nephrotoxicity and neurotoxicity: Differential regulation of COX-2/PGE 2 pathway. Biomed Pharmacother 2016; 84:1873-1884. [PMID: 27847198 DOI: 10.1016/j.biopha.2016.10.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 11/28/2022] Open
Abstract
To investigate the effect of Ginsenoside-Rb1 (GRb1) on lithium (Li+)-induced toxicity, GRb1 was given to rats orally (100mg/kg) for 14days. In independent groups, lithium chloride (4meq/kg/day i.p.) was administered at day 4 of the experiment for 10days, with or without GRb1. Li+ caused significant deterioration of behavioral responses including righting reflex, spontaneous motor activity and catalepsy. Li+ also caused distortion in normal renal, cerebral and cerebellum architecture and significantly worsened all kidney functional parameters tested compared to control. In addition, Li caused oxidative stress in both kidney and brain, evident by significant increase in malondialdehyde and nitric oxide levels, with decrease in reduced glutathione and catalase activity. Administration of GRb1 prior to Li+ significantly improved behavioral responses, renal and brain histopathological picture, kidney function tests and oxidative stress markers compared to sole Li+-treated group. Concomitant administration of GRb1 decreased Li+ levels by about 50% in serum, urine and brain and by 35% in the kidney. Interestingly, Li+ had a differential effect on cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) pathway, as it significantly increased COX-2 expression and PGE2 level in the kidney, while decreasing them in the brain compared to control. On the other hand, administering GRb1 with Li+ suppressed COX-2/PGE2 pathway in both kidney and brain compared to Li+ alone. In conclusion, GRb1 can alter Li+ pharmacokinetics resulting in extensively decreasing its serum and tissue concentrations. Furthermore, COX-2/PGE2 pathway has a mechanistic role in the nephro- and neuro-protective effects of GRb1 against Li+-induced toxicity.
Collapse
Affiliation(s)
| | - Maha Y Kamel
- Pharmacology Department, Faculty of Medicine, Minia University, Egypt
| |
Collapse
|
49
|
Effects of ginseng on two main sex steroid hormone receptors: estrogen and androgen receptors. J Ginseng Res 2016; 41:215-221. [PMID: 28413327 PMCID: PMC5386121 DOI: 10.1016/j.jgr.2016.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/06/2016] [Accepted: 08/15/2016] [Indexed: 01/01/2023] Open
Abstract
Ginseng has been used in China for at least two millennia and is now popular in over 35 countries. It is one of the world's popular herbs for complementary and alternative medicine and has been shown to have helpful effects on cognition and blood circulation, as well as anti-aging, anti-cancer, and anti-diabetic effects, among many others. The pharmacological activities of ginseng are dependent mainly on ginsenosides. Ginsenosides have a cholesterol-like four trans-ring steroid skeleton with a variety of sugar moieties. Nuclear receptors are one of the most important molecular targets of ginseng, and reports have shown that members of the nuclear receptor superfamily are regulated by a variety of ginsenosides. Here, we review the published literature on the effects of ginseng and its constituents on two main sex steroid hormone receptors: estrogen and androgen receptors. Furthermore, we discuss applications for sex steroid hormone receptor modulation and their therapeutic efficacy.
Collapse
|
50
|
Wu W, Gardner A, Sachse FB, Sanguinetti MC. Ginsenoside Rg3, a Gating Modifier of EAG Family K+ Channels. Mol Pharmacol 2016; 90:469-82. [PMID: 27502018 DOI: 10.1124/mol.116.104091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/05/2016] [Indexed: 01/11/2023] Open
Abstract
Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-à-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. However, it is unknown whether this action is unique to hERG1 channels. Here we compare and contrast the mechanisms of actions of Rg3 on hERG1 with three other members of the ether-à-go-go (EAG) K(+) channel gene family, including EAG1 (Kv10.1), ERG3 (Kv11.3), and ELK1 (Kv12.1). All four channel types were heterologously expressed in Xenopus laevis oocytes, and K(+) currents were measured using the two-microelectrode voltage-clamp technique. At a maximally effective concentration, Rg3 shifted the half-point of voltage-dependent activation of currents by -14 mV for ERG1 (EC50 = 414 nM), -20 mV for ERG3 (EC50 = 374 nM), -28 mV for EAG1 (EC50 = 1.18 μM), and more than -100 mV for ELK1 (EC50 = 197 nM) channels. Rg3 also induced slowing of ERG1, ERG3, and ELK1 channel deactivation and accelerated the rate of EAG1 channel activation. A Markov model was developed to simulate gating and the effects of Rg3 on the voltage dependence of activation of hELK1 channels. Understanding the mechanism underlying the action of Rg3 may facilitate the development of more potent and selective EAG family channel activators as therapies for cardiovascular and neural disorders.
Collapse
Affiliation(s)
- Wei Wu
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Alison Gardner
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute (W.W., A.G., F.B.S., M.C.S.), Department of Bioengineering (F.B.S.), Department of Internal Medicine, Division of Cardiovascular Medicine (M.C.S.), University of Utah, Salt Lake City, Utah
| |
Collapse
|