1
|
Wang Y, Wen J, Liu F, Peng X, Xu G, Zhang M, Huang Z. Traditional usages, chemical metabolites, pharmacological activities, and pharmacokinetics of Boesenbergia rotunda (L.) Mansf.: a comprehensive review. Front Pharmacol 2025; 16:1527210. [PMID: 40176912 PMCID: PMC11962002 DOI: 10.3389/fphar.2025.1527210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
Boesenbergia rotunda: (L.) Mansf. (family Zingiberaceae), also known as fingerroot, is a medicinal and food plant that is widely distributed in southern China, Southeast Asia, and South Asia. It is a traditional herb and spice that is also known for its beneficial effects on Qi, appetite, stagnation and pain relief. The objective of this study is to conduct a comprehensive and systematic review of the botanical characteristics, traditional applications, phytochemical metabolites, pharmacological properties, toxicology, quality control measures, pharmacokinetics, and clinical applications of B. rotunda. A bibliometric analysis of current studies on B. rotunda was also conducted to facilitate further exploration and utilization of B. rotunda in the functional food and pharmaceutical industries. These data were collected from PubMed, Web of Science, Google Scholar, China National Knowledge Infrastructure doctoral and master's theses and other books and scientific databases by searching the keywords Boesenbergia rotunda. Phytochemical analysis has revealed the presence of flavonoids, monoterpenes, alkaloids, aromatic metabolites, phenols, and other metabolites in B. rotunda, exhibiting a wide range of biological activities such as anti-cancer, nephroprotective, anti-inflammatory, anti-bacterial, hepatoprotective, anti-obesity, and anti-oxidant effects, both in vivo and in vitro. In this paper, the research of B. rotunda is discussed in depth by combining traditional application and modern pharmacological research, aiming to provide valuable reference for the future research and practical application of B. rotunda.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
| | - Juanjuan Wen
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
| | - Feng Liu
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
- Shaanxi Buchang Pharmaceutical Co. Ltd, Xi’an, Shaanxi, China
| | - Xiujuan Peng
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
| | - Gang Xu
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
| | - Mingliang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhuangzhuang Huang
- Key Laboratory of Chemical Substances and Biological Effects in Traditional Chinese Medicine, College of Medicine and Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi’an, Shaanxi, China
- Shaanxi Buchang Pharmaceutical Co. Ltd, Xi’an, Shaanxi, China
| |
Collapse
|
2
|
Monton C, Kulvanich P, Chankana N, Rangsimawong W, Suksaeree J, Charoenchai L, Theanphong O. Cluster analysis of fingerroot cultivated in different regions across Thailand and implementation of Quality by Design approach for R&D of fingerroot extract tablet. Food Res Int 2025; 202:115728. [PMID: 39967104 DOI: 10.1016/j.foodres.2025.115728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/13/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025]
Abstract
Fingerroot has been used as food, traditional medicine, and a dietary supplement. This study aimed to cluster fingerroot cultivated in different regions across Thailand, based on their flavonoids. Subsequently, the extract of fingerroot was formulated into tablet form using a Quality by Design (QbD) approach. Initially, 32 fingerroot samples were included in this study. They were analyzed using a validated high-performance liquid chromatographic method. The results indicated that the rhizomes of fingerroot had higher flavonoid content (pinocembrin, pinostrobin, and panduratin A) compared to the roots. Cluster analysis revealed that fingerroot from Surat Thani, Nong Bua Lamphu, Kalasin, and Sisaket Provinces formed a high flavonoid content group. Ethanolic extract of defatted fingerroot from Sisaket Province was then used to prepare tablets. Following the QbD approach, the quality target product profile, critical quality attributes, initial risk assessment, and formulation design were investigated. A Box-Behnken design was applied to optimize the tablet formulation. Design spaces were constructed, and the optimal formulation was verified. The optimal formulation comprised 3 % hydroxypropyl methylcellulose F4M without spray-dried lactose, compressed using 1,000 psi force. The tablets had an individual weight of 600 mg, a thickness of 4.22 mm, a diameter of 12.6 mm, a hardness of 6.33 kP, a disintegration time of 2.02 min, and a friability of 0.05 %. Each tablet contained 120 mg of fingerroot extract, consisting of 9.77 mg pinocembrin, 23.00 mg pinostrobin, and 11.27 mg panduratin A. Dissolution tests demonstrated that the three flavonoids could dissolve in a 0.5 % sodium lauryl sulfate solution at levels of 99.34 %, 103.81 %, and 61.44 % within 2 h, which was superior to a commercial product. In summary, this study provides a guide for selecting fingerroot cultivated in Thailand with high flavonoid content. Furthermore, the QbD approach was successfully implemented in the development of a fingerroot extract tablet with desired properties.
Collapse
Affiliation(s)
- Chaowalit Monton
- Drug and Herbal Product Research and Development Center, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand; Department of Pharmacognosy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand.
| | - Poj Kulvanich
- Industrial Pharmacy Program, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Natawat Chankana
- Sun Herb Thai Chinese Manufacturing, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Worranan Rangsimawong
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand
| | - Jirapornchai Suksaeree
- Department of Pharmaceutical Chemistry, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Laksana Charoenchai
- Drug and Herbal Product Research and Development Center, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Orawan Theanphong
- Department of Pharmacognosy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| |
Collapse
|
3
|
Kiatsoonthon K, Phimthong N, Potikanond S, Wikan N, Nimlamool W. Panduratin A Inhibits TNF Alpha-Stimulated Endothelial Cell Activation Through Suppressing the NF-κB Pathway. Biomolecules 2024; 15:34. [PMID: 39858429 PMCID: PMC11762725 DOI: 10.3390/biom15010034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway may help prevent the risk of developing the disease. The current study elucidated the inhibitory effect of panduratin A (PA) on TNF-α-induced endothelial activation and monocyte adhesion. We discovered that PA reduced the level of pro-inflammatory cytokine IL-6 and chemokine MCP-1 in the media collected from endothelial cells stimulated with TNF-α. In addition, PA inhibited the expression of ICAM-1 and VCAM-1 on the surface of TNF-α-induced endothelial cells resulting in a decrease in the number of monocytes attached to endothelial cell surface. Mechanistically, PA prevented IκB degradation and specifically suppressed NF-κB phosphorylation and nuclear translocation in endothelial cells. However, PA had no inhibitory effect on the phosphorylation of AKT, ERK1/2, p38, and JNK. Taken together, PA blocked the production of cytokine and chemokine, adhesion molecules, and monocyte adhesion in response to TNF-α stimulation, in part, through NF-κB inhibition. Our study suggests that PA may possibly be effective in blocking the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Kriangkrai Kiatsoonthon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitchakarn Phimthong
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
- PhD’s Degree Program in Pharmacology, Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.K.); (N.P.); (S.P.)
| |
Collapse
|
4
|
Alqudah A, Qnais E, Gammoh O, Bseiso Y, Wedyan M, Oqal M. Panduratin A mitigates inflammation and oxidative stress in DSS-induced colitis mice model. Future Sci OA 2024; 10:2428129. [PMID: 39559852 PMCID: PMC11581177 DOI: 10.1080/20565623.2024.2428129] [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: 01/06/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
AIM This study explored Panduratin A's protective effects against DSS-induced colitis in mice, focusing on reducing inflammation and oxidative stress in the colon. METHODS Mice were treated with dextran sodium sulfate (DSS) and Panduratin A (3, 6, 18 mg/kg), and changes in body weight, colon length, Disease Activity Index (DAI), histopathology, inflammation markers including tumor necrosis factor- α (TNF-α), Interleukin-1 β (IL-1β), Myeloperoxidase (MPO), and oxidative stress, Malondialdehyde (MDA) were evaluated. RESULTS Panduratin A significantly reversed DSS-induced symptoms, including body weight loss, colonic length shortening, and DAI increase, while reducing histopathological damage. It lowered inflammatory markers and oxidative stress, suppressed NF-κB activation, and enhanced Nrf2 and HO-1 expression. CONCLUSION Panduratin A shows promise as a colitis treatment, warranting further research for broader clinical application.
Collapse
Affiliation(s)
- Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | - Esam Qnais
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Yousra Bseiso
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mohammed Wedyan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Muna Oqal
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
5
|
Techapichetvanich P, Tangpanithandee S, Supannapan K, Wongwiwatthananukit S, Chang LC, Khemawoot P. Oral sub-chronic toxicity of fingerroot ( Boesenbergia rotunda) rhizome extract formulation in Wistar rats. Toxicol Rep 2024; 12:224-233. [PMID: 38328737 PMCID: PMC10847838 DOI: 10.1016/j.toxrep.2024.01.013] [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: 12/08/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 02/09/2024] Open
Abstract
Background Boesenbergia rotunda (fingerroot) rhizome extract contains two major bioactive components, panduratin A and pinostrobin. In our previous study, we found the anti-inflammatory effects of the fingerroot extract against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in golden Syrian hamsters. In the present study, we evaluated the sub-chronic toxicity of a fingerroot extract formulation over 90 consecutive days of oral administration. Methods We enhanced the water solubility of a fingerroot extract by formulating it with cyclodextrin, containing panduratin A (29% w/w) and pinostrobin (32% w/w). This formulation was administered to male and female Wistar rats at doses of 25, 50, or 100 mg/kg/day for a duration of 90 days. Additionally, two recovery groups, comprising a control group and a high-dose group, were designated for a 14-day observation period to assess the persistence and reversibility of potential adverse effects. Throughout the experiment, we performed clinical and health observations, followed by hematological testing, clinical biochemistry analysis, necropsy examination, and histopathological evaluation at the end of the experiment. Results The administration of the fingerroot extract formulation at doses of 25, 50, or 100 mg/kg/day did not result in mortality or clinical signs of toxicity. No clinically significant findings were associated with the oral administration of the fingerroot extract formulation. Conclusion The fingerroot extract formulation showed no serious adverse effects at doses up to 100 mg/kg/day in Wistar rats under the experimental condition. Consequently, the No Observed Adverse Effect Level (NOAEL) was considered to be 100 mg/kg/day. This finding contributes significance for future developments involving fingerroot extract in herbal medicinal products targeting chronic inflammation.
Collapse
Affiliation(s)
- Pinnakarn Techapichetvanich
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Supawit Tangpanithandee
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand
| | | | | | - Leng Chee Chang
- The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, USA
| | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand
| |
Collapse
|
6
|
Li G, Feng X, Wang W, Li J, Shi Y, Wang L, Hu C. Synthesis and biological evaluation of chromanone-based derivatives as potential anti-neuroinflammatory agents. Bioorg Chem 2023; 139:106767. [PMID: 37552914 DOI: 10.1016/j.bioorg.2023.106767] [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: 05/26/2023] [Revised: 07/29/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
As a privileged scaffold, chromanone has been extensively introduced in the design of drug leads with diverse pharmacological features, particularly in the area of inflammatory diseases. Herein, the preparation of chromanone-based derivatives (4a-4i) was smoothly achieved, and their structures were characterized using 1H NMR, 13C NMR, and ESI-HRMS spectroscopy techniques. Out of them, analogue 4e exhibited the most potent inhibitory capacity against the NO release and iNOS expression, without apparent cytotoxicity. Our observations showed that 4e could dramatically prevent the translocation of NF-κB from the cytoplasm to nucleus, and decrease the production of proinflammatory cytokines TNF-α, IL-6 and IL-1β in LPS-induced BV-2 cells. Mechanistically, 4e significantly deactivated NF-κB by disturbing TLR4-mediated TAK1/NF-κB and PI3K/Akt signaling cascades. Consistent with the in vitro study, 4e could effectively mitigate the inflammation response of hippocampal tissue in LPS-induced mouse model by inhibiting microglial activation. Collectively, these results revealed 4e as a prospective neuroprotective candidate for the therapy of neuroinflammation-related disorders.
Collapse
Affiliation(s)
- Guoxun Li
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Xiaoqing Feng
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Wenqian Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Jian Li
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China; Analysis and Testing Center, NERC Biomass of Changzhou University, Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Yeye Shi
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Lin Wang
- College of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Caijuan Hu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China.
| |
Collapse
|
7
|
Grekhnev DA, Kruchinina AA, Vigont VA, Kaznacheyeva EV. The Mystery of EVP4593: Perspectives of the Quinazoline-Derived Compound in the Treatment of Huntington's Disease and Other Human Pathologies. Int J Mol Sci 2022; 23:15724. [PMID: 36555369 PMCID: PMC9778905 DOI: 10.3390/ijms232415724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Quinazoline derivatives have various pharmacological activities and are widely used in clinical practice. Here, we reviewed the proposed mechanisms of the physiological activity of the quinazoline derivative EVP4593 and perspectives for its clinical implication. We summarized the accumulated data about EVP4593 and focused on its activities in different models of Huntington's disease (HD), including patient-specific iPSCs-based neurons. To make a deeper insight into its neuroprotective role in HD treatment, we discussed the ability of EVP4593 to modulate calcium signaling and reduce the level of the huntingtin protein. Moreover, we described possible protective effects of EVP4593 in other pathologies, such as oncology, cardiovascular diseases and parasite invasion. We hope that comprehensive analyses of the molecular mechanisms of EVP4593 activity will allow for the expansion of the scope of the EVP4593 application.
Collapse
|