1
|
Li L, Yang Z, Li Q, Guo Q, Wu X, Wang Y, Shen X, Chen Y, Tao L. Ameliorating vascular endothelial injury for lipolysacharide-induced via mitochondrial targeting function of octaarginine-modified essential oil from Fructus Alpiniae zerumbet (EOFAZ) lipid microspheres. CHINESE HERBAL MEDICINES 2025; 17:340-351. [PMID: 40256722 PMCID: PMC12009099 DOI: 10.1016/j.chmed.2024.11.004] [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: 03/16/2024] [Revised: 09/04/2024] [Accepted: 11/19/2024] [Indexed: 03/18/2025] Open
Abstract
OBJECTIVE To investigate the therapeutic potential of octaarginine (R8)-modified essential oil from Fructus Alpiniae zerumbet (EOFAZ) lipid microspheres (EOFAZ@R8LM) for cardiovascular therapy. METHODS EOFAZ@R8LM was developed by leveraging the volatilization of EOFAZ and integrating it with the oil phase of LM, followed by surface modification with cell-penetrating peptide R8 to target the site of vascular endothelial injury. The therapeutic effects of this formulation in alleviating lipopolysaccharide-induced vascular endothelial inflammation were evaluated by assessing mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) levels, as well as inflammatory factors interleukin-6 (IL-6) and interleukin-1β (IL-1β) levels. RESULTS EOFAZ@R8LM effectively delivered EOFAZ to the site of injury and specifically targeted the mitochondria in vascular endothelial cells, thereby ameliorating mitochondrial dysfunction through regulation of MMP and reduction of intracellular ROS levels. Moreover, it attenuated the expression levels of IL-6 and IL-1β, exerting protective effects on the vascular endothelium. CONCLUSION Our findings highlight the significant therapeutic potential of EOFAZ@R8LM in cardiovascular therapy, providing valuable insights for developing novel dosage forms utilizing EOFAZ for effective treatment against cardiovascular diseases.
Collapse
Affiliation(s)
- Lingyan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Zengqiu Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Qiqi Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Qianqian Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Xingjie Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Yu’e Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Ying Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Ling Tao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
2
|
Li J, Liu Y, Geng K, Lu X, Shen X, Guo Q. ROS-Responsive Nanoparticles with Antioxidative Effect for the treatment of Diabetic Retinopathy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:440-461. [PMID: 39316729 DOI: 10.1080/09205063.2024.2406628] [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: 02/19/2024] [Accepted: 04/29/2024] [Indexed: 09/26/2024]
Abstract
Diabetic retinopathy (DR) is a common microvascular complication of diabetes necessitating early intervention to impede progression, despite current clinical treatments focusing on advanced stages. Essential oils from Fructus Alpiniae zerumbet (EOFAZ) have demonstrated efficacy in protecting against high glucose (HG)-induced Müller cell activation and DR development. This study introduced a reactive oxidative species (ROS)-responsive drug delivery system (NPSPHE@EOFAZ) targeting early DR stages and oxidative stress. Our engineered nanoparticles effectively deliver EOFAZ into HG-exposed Müller cells by detecting and responding to elevated oxidative stress levels. The NPSPHE@EOFAZ significantly inhibited abnormal cell growth, reduced oxidative stress, and alleviated inflammation in vitro. In vivo experiments on diabetic mice with DR revealed that NPSPHE@EOFAZ mitigated early pathological changes by reducing oxidative stress and inflammation while also alleviating organ damage in the heart, liver, spleen, lung, and kidney. These findings underscore the potential of NPSPHE@EOFAZ as a promising antioxidant for early intervention in DR pathogenesis.
Collapse
Affiliation(s)
- Jinjin Li
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yujia Liu
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Kedui Geng
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xin Lu
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xiangchun Shen
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qianqian Guo
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
3
|
Wang YE, Chen J, Yang H, He J, Varier KM, Chen Y, Wu X, Guo Q, Liang Y, Shen X, Wei M, Li W, Tao L. Polysialic acid driving cardiovascular targeting co-delivery 1,8-cineole and miR-126 to synergistically alleviate lipopolysaccharide-induced acute cardiovascular injury. Int J Biol Macromol 2024; 280:135970. [PMID: 39332566 DOI: 10.1016/j.ijbiomac.2024.135970] [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: 11/24/2023] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Infection-induced cardiovascular damage is the primary pathological mechanism underlying septic cardiac dysfunction. This condition affects the majority of patients in intensive care unit and has an unfavorable prognosis due to the lack of effective therapies available. Vascular cell adhesion molecule-1 (VCAM-1) plays a vital role in coordinating the inflammatory response and recruitment of leukocytes in cardiac tissue, making it a potential target for developing novel therapies. MicroRNA-126 (miR-126) has been shown to downregulate VCAM-1 expression in endothelial cells, reducing leukocyte adhesion and exerting anti-inflammatory effects. Therefore, this work described a polysialic acid (PSA) modified ROS-responsive nanosystem to targeted co-delivery 1,8-Cineole and miR-126 for mitigating septic cardiac dysfunction. The nanosystem consists of 1,8-Cineole nanoemulsion (CNE) conjugated with PEI/miR126 complex by a ROS-sensitive linker, with PSA on its surface to facilitate targeted delivery via specific interactions with selectins on endothelial cells. CNE has demonstrated protective effects against inflammation in the cardiovascular system and synergistic anti-inflammatory effects when combined with miR-126. The targeted nanosystem successfully delivered miR-126 and 1,8-Cineole to the injured heart tissues and vessels, reducing inflammatory responses and improving cardiac function. In summary, this work provides a promising therapy for alleviating the inflammatory response in sepsis while boosting cardiovascular protection.
Collapse
Affiliation(s)
- Yu-E Wang
- Department of Cardiovascular medicine, Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang 550025, China; The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Jianbo Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Hong Yang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Jinggang He
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Krishnapriya M Varier
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Ying Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Xingjie Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Yuanxian Liang
- School of Clinical Medicine, Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| | - Maochen Wei
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| | - Wei Li
- Department of Cardiovascular medicine, Affiliated Hospital of Guizhou Medical University, Beijing Road, Yunyan District, Guiyang 550025, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, Guizhou Medical University, NO. 6 Ankang avenue, Guian New District, 561113, Guizhou, China; The Department of Pharmacology (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), Guizhou Medical University, No. 6 Ankang avenue, Guian New District, 561113, Guizhou, China.
| |
Collapse
|
4
|
Wang Y, Li Y, Lu Y, Li J. Biomimetic Nanoparticles for the Diagnosis and Therapy of Atherosclerosis. CHEM REC 2024; 24:e202400087. [PMID: 39148157 DOI: 10.1002/tcr.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/23/2024] [Indexed: 08/17/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammation of blood vessels, which often has no obvious symptoms in the early stage of the disease, but when atherosclerotic plaques are formed, they often cause lumen blockage, and even plaque rupture leads to thrombosis, that is the essential factor of cardiovascular events, for example myocardial infarction, cerebral infarction, and renal atrophy. Therefore, it is considerably significant for the early recognition and precise therapy of plaque. Biomimetic nanoparticles (BNPs), especially those coated with cell membranes, can retain the biological function of cell membranes or cells, which has led to extensive research and application in the diagnosis and treatment of AS in recent years. In this review, we summarized the roles of various key cells in AS progression, the construction of biomimetic nanoparticles based on these key cells as well as their applications in AS diagnosis and therapy. Furthermore, we give a challenge and prospect of biomimetic nanoparticles in AS, hoping to elevate their application quality and the possibility of clinical translation.
Collapse
Affiliation(s)
- Yan Wang
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yize Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yuqing Lu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Jingjing Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| |
Collapse
|
5
|
Jiang F, Wu G, Yang H, Zhang Y, Shen X, Tao L. Diethylaminoethyl-dextran and monocyte cell membrane coated 1,8-cineole delivery system for intracellular delivery and synergistic treatment of atherosclerosis. Int J Biol Macromol 2023; 253:127365. [PMID: 37827418 DOI: 10.1016/j.ijbiomac.2023.127365] [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: 07/09/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
We have developed a biomimetic delivery system termed the Monocyte Cell Membrane-Coated 1,8-Cineole Biomimetic Delivery System (MM-CIN-BDS or BDS), which integrates diethylaminoethyl-dextran (DEAE) and monocyte cell membrane (MM). This innovative approach enhances the cellular uptake efficiency of 1,8-cineole (CIN) and facilitates targeted therapy for atherosclerosis. Our findings demonstrate the successful modification of the drug carrier with DEAE and MM, as validated by measurements of particle size, zeta potential, microscopic morphology, and western blotting analyses. Notably, cellular uptake experiments unveil a significant enhancement in cellular uptake efficiency due to DEAE modification. However, the introduction of monocyte cell membranes diminishes this effect in normal human umbilical vein endothelial cells (HUVECs), although this efficiency is notably restored in HUVECs activated with lipopolysaccharide (LPS). Through in vivo imaging investigations, we observe that the MM coating augments distribution in the spleen, brain, and atherosclerotic plaques, while concurrently diminishing distribution in the heart and kidneys. Animal studies corroborate these findings, illustrating that MM-CIN-BDS treatment curtails lipid parameters, dampens the expression of inflammatory factors and proteins, mitigates vascular tissue damage, and ultimately reduces the extent of atherosclerotic lesion areas. To encapsulate, DEAE emerges as an especially adept agent for modifying drug carriers with suboptimal cellular uptake efficiency in the realm of cardiovascular diseases. The potential therapeutic promise of MM-CIN-BDS for atherosclerosis treatment is evident from our research.
Collapse
Affiliation(s)
- Feng Jiang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, The Department of Pharmacology of Materia Medical, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Guoping Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, The Department of Pharmacology of Materia Medical, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Hong Yang
- Department of Pharmacy, Guiyang Maternal and Child Health Care Hospital, Guiyang 550003, Guizhou, China
| | - Yanyan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, The Department of Pharmacology of Materia Medical, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, The Department of Pharmacology of Materia Medical, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmaceutical Sciences, The Department of Pharmacology of Materia Medical, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China.
| |
Collapse
|
6
|
Hoch CC, Petry J, Griesbaum L, Weiser T, Werner K, Ploch M, Verschoor A, Multhoff G, Bashiri Dezfouli A, Wollenberg B. 1,8-cineole (eucalyptol): A versatile phytochemical with therapeutic applications across multiple diseases. Biomed Pharmacother 2023; 167:115467. [PMID: 37696087 DOI: 10.1016/j.biopha.2023.115467] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
1,8-cineole (Eucalyptol), a naturally occurring compound derived from botanical sources such as eucalyptus, rosemary, and camphor laurel, has a long history of use in traditional medicine and exhibits an array of biological properties, including anti-inflammatory, antioxidant, antimicrobial, bronchodilatory, analgesic, and pro-apoptotic effects. Recent evidence has also indicated its potential role in managing conditions such as Alzheimer's disease, neuropathic pain, and cancer. This review spotlights the health advantages of 1,8-cineole, as demonstrated in clinical trials involving patients with respiratory disorders, including chronic obstructive pulmonary disease, asthma, bronchitis, and rhinosinusitis. In addition, we shed light on potential therapeutic applications of 1,8-cineole in various conditions, such as depression, epilepsy, peptic ulcer disease, diarrhea, cardiac-related heart diseases, and diabetes mellitus. A comprehensive understanding of 1,8-cineole's pharmacodynamics and safety aspects as well as developing effective formulations, might help to leverage its therapeutic value. This thorough review sets the stage for future research on diverse health benefits and potential uses of 1,8-cineole in tackling complex medical conditions.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Julie Petry
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Lena Griesbaum
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Tobias Weiser
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Kathrin Werner
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | | | - Admar Verschoor
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, 81675 Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany; Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Department of Radiation Oncology, Klinikum rechts der Isar, 81675 Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany.
| |
Collapse
|
7
|
Dabravolski SA, Sukhorukov VN, Melnichenko AA, Khotina VA, Orekhov AN. Potential Application of the Plant-Derived Essential Oils for Atherosclerosis Treatment: Molecular Mechanisms and Therapeutic Potential. Molecules 2023; 28:5673. [PMID: 37570643 PMCID: PMC10420188 DOI: 10.3390/molecules28155673] [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: 06/21/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Essential oils (EOs) are complex secondary metabolites identified in many plant species. Plant-derived EOs have been widely used in traditional medicine for centuries for their health-beneficial effects. Some EOs and their active ingredients have been reported to improve the cardiovascular system, in particular to provide an anti-atherosclerotic effect. The objective of this review is to highlight the recent research investigating the anti-inflammatory, anti-oxidative and lipid-lowering properties of plant-derived EOs and discuss their mechanisms of action. Also, recent clinical trials exploring anti-inflammatory and anti-oxidative activities of EOs are discussed. Future research on EOs has the potential to identify new bioactive compounds and invent new effective agents for treatment of atherosclerosis and related diseases such as diabetes, metabolic syndrome and obesity.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel
| | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Alexandra A. Melnichenko
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Victoria A. Khotina
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| | - Alexander N. Orekhov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia; (V.N.S.); (A.A.M.); (V.A.K.); (A.N.O.)
| |
Collapse
|
8
|
Chen Y, Wang J, Xu J, Zhang J, Xu S, Zhang Q, Huang J, Peng J, Xu H, Du Q, Gong Z. Fabrication of a Polysaccharide-Protein/Protein Complex Stabilized Oral Nanoemulsion to Facilitate the Therapeutic Effects of 1,8-Cineole on Atherosclerosis. ACS NANO 2023; 17:9090-9109. [PMID: 37172004 DOI: 10.1021/acsnano.2c12230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Atherosclerosis (AS) is a systemic disease characterized by lipid deposition in the blood vessel wall that urgently requires effective and safe therapeutic drugs for long-term treatment. An essential oil monomer-1,8-cineole (CIN) with ameliorative effects on vascular injuries has considerable potential for preventing the progression of AS because of its antioxidant, anti-inflammation, and cholesterol regulatory effects. However, the high volatility and instability of CIN result in low oral bioavailability and a short half-life, thereby limiting its clinical application. We formulated a nanoemulsion using a polysaccharide-protein/protein complex (dextran-bovine serum albumin/protamine, DEX5k-BSA/PTM) as an emulsifier, with vitamin B12 (VB12) as the ligand to facilitate the transportation across the small intestine. An emulsion preparation method using a microjet followed by ultraviolet irradiation was developed to obtain the CIN-loaded oral nanoemulsion CIN@DEX5k-BSA/PTM/VB12. The nanoemulsion improved the stability of CIN both in vitro and in vivo, prolonged the retention time in the gastrointestinal tract (GIT), and enhanced the permeability across the mucus layer and intestinal epithelial cells to increase oral bioavailability and plaque accumulation of CIN. Validated in an AS mouse model, CIN@DEX5k-BSA/PTM/VB12 achieved prominent therapeutic efficacy combating AS. This study highlights the advantages of DEX5k-BSA/PTM and VB12 in the development of nanoemulsions for CIN and provides a promising oral nanoplatform for the delivery of essential oils.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | | | | |
Collapse
|
9
|
Venkataraman B, Almarzooqi S, Raj V, Bhongade BA, Patil RB, Subramanian VS, Attoub S, Rizvi TA, Adrian TE, Subramanya SB. Molecular Docking Identifies 1,8-Cineole (Eucalyptol) as A Novel PPARγ Agonist That Alleviates Colon Inflammation. Int J Mol Sci 2023; 24:ijms24076160. [PMID: 37047133 PMCID: PMC10094723 DOI: 10.3390/ijms24076160] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Inflammatory bowel disease, comprising Crohn's disease (CD) and ulcerative colitis (UC), is often debilitating. The disease etiology is multifactorial, involving genetic susceptibility, microbial dysregulation, abnormal immune activation, and environmental factors. Currently, available drug therapies are associated with adverse effects when used long-term. Therefore, the search for new drug candidates to treat IBD is imperative. The peroxisome proliferator-activated receptor-γ (PPARγ) is highly expressed in the colon. PPARγ plays a vital role in regulating colonic inflammation. 1,8-cineole, also known as eucalyptol, is a monoterpene oxide present in various aromatic plants which possess potent anti-inflammatory activity. Molecular docking and dynamics studies revealed that 1,8-cineole binds to PPARγ and if it were an agonist, that would explain the anti-inflammatory effects of 1,8-cineole. Therefore, we investigated the role of 1,8-cineole in colonic inflammation, using both in vivo and in vitro experimental approaches. Dextran sodium sulfate (DSS)-induced colitis was used as the in vivo model, and tumor necrosis factor-α (TNFα)-stimulated HT-29 cells as the in vitro model. 1,8-cineole treatment significantly decreased the inflammatory response in DSS-induced colitis mice. 1,8-cineole treatment also increased nuclear factor erythroid 2-related factor 2 (Nrf2) translocation into the nucleus to induce potent antioxidant effects. 1,8-cineole also increased colonic PPARγ protein expression. Similarly, 1,8-cineole decreased proinflammatory chemokine production and increased PPARγ protein expression in TNFα-stimulated HT-29 cells. 1,8-cineole also increased PPARγ promoter activity time-dependently. Because of its potent anti-inflammatory effects, 1,8-cineole may be valuable in treating IBD.
Collapse
Affiliation(s)
- Balaji Venkataraman
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Saeeda Almarzooqi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Vishnu Raj
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Bhoomendra A Bhongade
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Rajesh B Patil
- Department of Pharmaceutical Chemistry, Sinhgad College of Pharmacy, Vadgaon (BK), Pune 411 041, India
| | | | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Tahir A Rizvi
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Thomas E Adrian
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates
| | - Sandeep B Subramanya
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
10
|
Bahrami T, Yaghmaei P, Yousofvand N. The effects of Ibuprofen and 1, 8- cineol on anxiety and spatial memory in hyperammonemic rats. Metab Brain Dis 2023; 38:613-620. [PMID: 36346500 DOI: 10.1007/s11011-022-01093-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/29/2022] [Indexed: 11/11/2022]
Abstract
In hepatic encephalopathy, hyperammonemia (HA) causes cognitive impairment and anxiety by causing neuroinflammation. Ibuprofen and 1,8- cineol have anti-inflammatory and antioxidant properties, respectively. The aim of this study was to evaluate the effects of ibuprofen alone and in combination with 1,8- cineol on anxiety and oxidative stress in a HA rat animal model. For this purpose, 36 rats were divided into six groups (n = 6) including the HA (received intraperitoneally (IP) ammonium acetate 2.5 mg/kg for four week), ibuprofen (induced HA rats that received 15 mg/kg, IP), cineol (induced HA rats that received 5 and 10 mg/kg, IP), Ib + cineol (induced HA rats that received 15 and 10 mg/kg, respectively, IP), and the control groups (received normal saline, IP). Except the HA group, all other groups received the aforementioned treatment for two weeks.. The Morris water maze and elevated plus maze were used to assess cognitive function and anxiety in the animals, respectively. Superoxide dismutase (SOD) activity was measured to evaluate oxidative stress. The mRNA expression levels of interleukin (IL)-6 and IL-1β was assessed by real-time PCR in the animal's brain. The results showed a significant improvement in spatial memory and anxiety of the Ib group compared to the HA group (P < 0.01), but no significant change was observed in SOD activity (P > 0.05). There was a significant improvement in spatial memory and anxiety as well as a significant increase in SOD activity in the Ib + cineol group (P < 0.01) compared to the HA group. These results indicate that the Ib + cineol, not only improve cognitive function and reduce anxiety, also reduce oxidative stress, therefore, the simultaneous use of these two compounds may be useful in improving HA-induced cognitive disorders and anxiety.
Collapse
Affiliation(s)
- Tayebeh Bahrami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Namdar Yousofvand
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| |
Collapse
|
11
|
Di Y, Cao A, Zhang Y, Li J, Sun Y, Geng S, Li Y, Zhang L. Effects of Dietary 1,8-Cineole Supplementation on Growth Performance, Antioxidant Capacity, Immunity, and Intestine Health of Broilers. Animals (Basel) 2022; 12:ani12182415. [PMID: 36139274 PMCID: PMC9495220 DOI: 10.3390/ani12182415] [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: 07/22/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
This study was conducted to investigate the effects of 1,8-cineole on antioxidant capacity, immunity, and intestinal health of broilers. A total of 540 1-day-old Arbor Acres (AA) male broilers were randomly divided into five treatments with six replicates per treatment, and 18 broilers per replicate for 42 days. Dietary treatments were a corn−soybean meal basal diet supplemented with 0, and 10, 20, 30, and 40 mg/kg 1,8-cineole, respectively. Dietary supplementation with 20~30 mg/kg of 1,8-cineole increased the ADG from d 22 to 42 and d 1 to 42 (p < 0.05), and decreased the FCR (p < 0.05). Dietary supplementation of 10~40 mg/kg of 1,8-cineole increased total antioxidant capacity (TAOC) in serum (p < 0.05), and decreased malondialdehyde (MDA) level in the liver on day 21 (p < 0.05). The supplementation of 20~30 mg/kg of 1,8-cineole increased the activity of total superoxide dismutase (T-SOD) in the serum and liver and TAOC in the serum and the liver (p < 0.05), and decreased the level of MDA in the serum and the liver (p < 0.05) on day 42. Dietary supplementation with 20~30 mg/kg of 1,8-cineole increased serum immunoglobulin A, immunoglobulin G, and immunoglobulin M contents on day 21 (p < 0.05). On day 21, dietary supplementation of 20~30 mg/kg of 1,8-cineole increased the VH and VH/CD (p < 0.05) in the jejunum and ileum. The supplementation of 20~30 mg/kg of 1,8-cineole increased the content of secretory immunoglobulin A in the duodenum and ileum mucosa on d 42 (p < 0.05). In conclusion, dietary supplementation of 1,8-cineole improves the growth performance of broilers by enhancing antioxidant capacity, immunity, and intestinal morphology.
Collapse
|
12
|
de Oliveira MC, Bruschi ML. Self-Emulsifying Systems for Delivery of Bioactive Compounds from Natural Origin. AAPS PharmSciTech 2022; 23:134. [PMID: 35534702 DOI: 10.1208/s12249-022-02291-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
Nature has been used as therapeutic resources in the treatment of diseases for many years. However, some natural compounds have poor water solubility. Therefore, physicochemical strategies and technologies are necessary for development of systems for carrying these substances. The self-emulsifying drug delivery systems (SEDDS) have been used as carriers of hydrophobic compounds in order to increase the solubility and absorption, improving their bioavailability. SEDDS are constituted with a mixture of oils and surfactants which, when come into contact with an aqueous medium under mild agitation, can form emulsions. In the last years, a wide variety of self-emulsifying formulations containing bioactive compounds from natural origin has been developed. This review provides a comprehensive overview of the main excipients and natural bioactive compounds composing SEDDS. In addition, applications, new technologies and innovation are reviewed as well. Examples of self-emulsifying formulations administered in different sites are also considered for a better understanding of the use of this strategy to modify the delivery of compounds from natural origin.
Collapse
|
13
|
5,2′-Dibromo-2,4′,5′-trihydroxydiphenylmethanone Inhibits LPS-Induced Vascular Inflammation by Targeting the Cav1 Protein. Molecules 2022; 27:molecules27092884. [PMID: 35566232 PMCID: PMC9101869 DOI: 10.3390/molecules27092884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular inflammation is directly responsible for atherosclerosis. 5,2′-Dibromo-2,4′,5′-trihydroxydiphenylmethanone (TDD), a synthetic bromophenol derivative, exhibits anti-atherosclerosis and anti-inflammatory effects. However, the underlying pathways are not yet clear. In this study, we first examined the effects of TDD on toll-like receptor-4 (TLR4) activity, the signaling receptor for lipopolysaccharide (LPS), and found that TDD does not inhibit LPS-induced TLR4 expression in EA.hy926 cells and the vascular wall in vivo. Next, we investigated the global protein alterations and the mechanisms underlying the action of TDD in LPS-treated EA.hy926 cells using an isobaric tag for the relative and absolute quantification technique. Western blot analysis revealed that TDD inhibited NF-κB activation by regulating the phosphorylation and subsequent degradation IκBα. Among the differentially expressed proteins, TDD concentration-dependently inhibited Caveolin 1(Cav1) expression. The interaction between Cav1 and TDD was determined by using biolayer interference assay, UV-vis absorption spectra, fluorescence spectrum, and molecular docking. We found that TDD can directly bind to Cav1 through hydrogen bonds and van der Waals forces. In conclusion, our results showed that TDD inhibited LPS-induced vascular inflammation and the NF-κB signaling pathway by specifically targeting the Cav1 protein. TDD may be a novel anti-inflammatory compound, especially for the treatment of atherosclerosis.
Collapse
|
14
|
Cai ZM, Peng JQ, Chen Y, Tao L, Zhang YY, Fu LY, Long QD, Shen XC. 1,8-Cineole: a review of source, biological activities, and application. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2021; 23:938-954. [PMID: 33111547 DOI: 10.1080/10286020.2020.1839432] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 06/11/2023]
Abstract
1,8-Cineole (also known as eucalyptol) is mostly extracted from the essential oils of plants, which showed extensively pharmacological properties including anti-inflammatory and antioxidant mainly via the regulation on NF-κB and Nrf2, and was used for the treatment of respiratory diseases and cardiovascular, etc. Although various administration routes have been used in the application of 1.8-cineole, few formulations have been developed to improve its stability and bioavailability. This review retrospects the researches on the source, biological activities, mechanisms, and application of 1,8-cineole since 2000, which provides a view for the further studies on the application and formulations of 1,8-cineole.
Collapse
Affiliation(s)
- Zi-Min Cai
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Jian-Qing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling Tao
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yan-Yan Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling-Yun Fu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Qing-De Long
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Xiang-Chun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550014, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
15
|
Peng J, Jiang Z, Wu G, Cai Z, Du Q, Tao L, Zhang Y, Chen Y, Shen X. Improving protection effects of eucalyptol via carboxymethyl chitosan-coated lipid nanoparticles on hyperglycaemia-induced vascular endothelial injury in rats. J Drug Target 2021; 29:520-530. [PMID: 33263461 DOI: 10.1080/1061186x.2020.1859514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/14/2020] [Accepted: 11/25/2020] [Indexed: 10/22/2022]
Abstract
Hyperglycaemia is responsible for the major pathophysiological factor of diabetes-associated vascular endothelial injury, which mainly resulted from the disturbance of equilibrium between ROS generation and elimination. Eucalyptol was verified with exact anti-oxidation effects via stimulating the secretion of endogenous antioxidant enzymes against ROS. However, the volatility, instability and poor water solubility of eucalyptol limited its pharmacological activities in vivo. In this study, we developed carboxymethyl chitosan-coated lipid nanoparticles for eucalyptol (CMC/ELN) to facilitate oral administration. A thin lipid film dispersion method was used to prepare the ELN. After CMC coating, the diameter of ELN increased from 166 nm to 177 nm and charge reversal was observed. The nanocarrier enhanced the protective effects of eucalyptol both in the high level of glucose (HG)-damaged HUVECs and endothelial injury in type I diabetes mellitus (T1DM) rat model. Furthermore, the mechanism of eucalyptol on the promotion of Nrf2 and HO-1 and reduction on Keap1 expression have been verified both in the in vitro and in vivo model. Besides, the pharmacokinetics data were verified the promotion of the oral eucalyptol absorption by the nanocarrier. Taken together, we established an optimal oral delivery system that promoted oral administration of eucalyptol to exert protective effects on hyperglycaemia-induced vascular endothelial injury.
Collapse
Affiliation(s)
- Jianqing Peng
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Zhaohui Jiang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Guoping Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Zimin Cai
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yanyan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yi Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
16
|
Wang Y, Chen L, Adu‐Frimpong M, Wei C, Weng W, Wang Q, Xu X, Yu J. Preparation, In Vivo and In Vitro Evaluation, and Pharmacodynamic Study of DMY‐Loaded Self‐Microemulsifying Drug Delivery System. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yaping Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Lin Chen
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Michael Adu‐Frimpong
- Department of Applied Chemistry and Biochemistry CK Tedam University of Technology and Applied Science Navrongo, Upper East Region 31011 Ghana
| | - Chunmei Wei
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Wen Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Xi‐Ming Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - JiangNan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| |
Collapse
|
17
|
Improvement of Oral Bioavailability and Anti-Tumor Effect of Zingerone Self-Microemulsion Drug Delivery System. J Pharm Sci 2021; 110:2718-2727. [PMID: 33610568 DOI: 10.1016/j.xphs.2021.01.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
Abstract
This study sought to prepare a self-microemulsion drug delivery system containing zingerone (Z-SMEDDS) to improve the low oral bioavailability of zingerone and anti-tumor effect. Z-SMEDDS was characterized by particle size, zeta potential and encapsulation efficiency, while its pharmacokinetics and anti-tumor effects were also evaluated. Z-SMEDDS had stable physicochemical properties, including average particle size of 17.29 ± 0.07 nm, the zeta potential of -22.81 ± 0.29 mV, and the encapsulation efficiency of 97.96% ± 0.02%. In vitro release studies have shown the release of zingerone released by Z-SMEDDS was significantly higher than free zingerone in different release media. The relative oral bioavailability of Z-SMEDDS was 7.63 times compared with free drug. Meanwhile, the half inhibitory concentration (IC50)of Z-SMEDDS and free zingerone was 8.45 μg/mL and 13.30 μg/mL, respectively on HepG2. This study may provide a preliminary basis for further clinical research and application of Z-SMEDDS.
Collapse
|
18
|
Xiao T, Huang J, Wang X, Wu L, Zhou X, Jiang F, He Z, Guo Q, Tao L, Shen X. Alpinia zerumbet and Its Potential Use as an Herbal Medication for Atherosclerosis: Mechanistic Insights from Cell and Rodent Studies. Lifestyle Genom 2020; 13:138-145. [PMID: 32882697 DOI: 10.1159/000508818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/19/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Alpinia zerumbet (Pers.) Burtt. et Smith has been used as a flavor additive in food and a traditional medicine for centuries, especially in Guizhou Province, China, and it prolongs people's lives with multiple beneficial effects. Thus, one of the aims of this review was to expound the chemical constituents of this plant, especially its fruits. Since cardiovascular diseases, including atherosclerosis, pose a health threat to humans, another aim was to expound the possible mechanisms of its potential use as an herbal medication for atherosclerosis. METHODS In this study, 10 reports are cited to expound the potential bioactive compounds. Moreover, 33 reports explain the antihypertensive and antiatherosclerotic effects of the plant by ameliorating inflammation and endothelial dysfunction, increasing vasodilation, improving hyperlipidemia, downgrading the glucose status, and working as an antioxidant. RESULTS A. zerumbetis rich in terpenes, essential oils, flavonoids, polyphenolics, and sterols. Pharmacological experiments showed that A. zerumbet has antioxidative and anti-inflammatory effects on the NF-κB signaling pathway and can ameliorate oxidative stress in the NOS-NO signaling pathway. Moreover, A. zerumbet demonstrates antihypertensive effects by accelerating vasorelaxant response and increasing 3T3-L1 intracellular cAMP, which has promising antiobesity properties, as well as hypolipidemic and anti-diabetic complication effects. CONCLUSIONS A. zerumbet has potential functions and applications in the prevention of atherosclerosis, but further studies are required before clinical trials.
Collapse
Affiliation(s)
- Ting Xiao
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jiaoyan Huang
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiaowei Wang
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Linjing Wu
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xue Zhou
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Feng Jiang
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Zhiyong He
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qianqian Guo
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling Tao
- Department of Pharmaceutic Preparation of Chinese Medicine, the State Key Laboratory of Functions and Applications of Medicinal Plants, High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiangchun Shen
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China,
- The Department of Pharmacology of Materia Medica, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, Union Key Laboratory of Guiyang City-Guizhou Medical University, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China,
| |
Collapse
|
19
|
Dong J, Zhu XM, Wu FY, Yang BQ, Feng H, Dong YF, Gu W, Chen J. Development of galangal essential oil-based microemulsion gel for transdermal delivery of flurbiprofen: simultaneous permeability evaluation of flurbiprofen and 1,8-cineole. Drug Dev Ind Pharm 2020; 46:91-100. [PMID: 31878816 DOI: 10.1080/03639045.2019.1706548] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Flurbiprofen (FP) is one of the most potent nonsteroidal anti-inflammatory drugs with very low bioavailability of approximately 12% following transdermal administration, compared to that after oral administration. This study aimed to deliver FP as a microemulsion (ME) gel by transdermal administration. Galangal essential oil (GEO) was extracted from Rhizoma Alpiniae Officinarum and identified by GC-MS. The most abundant constituent was determined to be 1,8-cineole (52.06%). Compared to azone, GEO was proved to exert significantly higher (p < .01) penetration enhancement effect and significantly (p < .001) lower skin cell toxicity. The formulation (FP-GEO-ME gel) was prepared using GEO as an oil phase and a penetration enhancer. Compared to that of FP solution, the enhancement ratio (ER) of FP-GEO-ME gel was 4.06. In addition, more than 25% 1,8-cineole permeated through the rat skin. In vivo pharmacokinetic studies revealed that the AUC0-t of FP after transdermal administration of FP-GEO-ME gel was higher by approximately 4.56-fold than that of marketed FP cataplasms. The relative bioavailability of FP and 1,8-cineole after transdermal administration compared to oral administration of FP-GEO-ME were determined to be 96.58% and 85.49%, respectively. FP-GEO-ME gel significantly inhibited carrageenan-induced hind-paw edema and decreased PGE2 levels in rat serum. GEO-ME gel also exhibited significant anti-inflammatory effects at 2 h after the therapy (p < .05). The synergistic effects of FP and GEO were expected for the application of FP-GEO-ME gel. In conclusion, GEO-ME gel may be a promising formulation for transdermal administration of anti-inflammatory hydrophobic drugs, such as FP.
Collapse
Affiliation(s)
- Jie Dong
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue-Min Zhu
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng-Ye Wu
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bing-Qing Yang
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Feng
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Fei Dong
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Gu
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Chen
- Jiangsu Provincial Key Laboratory of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.,Pharmaceutical Research Laboratory, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Linghu KG, Wu GP, Fu LY, Yang H, Li HZ, Chen Y, Yu H, Tao L, Shen XC. 1,8-Cineole Ameliorates LPS-Induced Vascular Endothelium Dysfunction in Mice via PPAR-γ Dependent Regulation of NF-κB. Front Pharmacol 2019; 10:178. [PMID: 30930772 PMCID: PMC6423908 DOI: 10.3389/fphar.2019.00178] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
1,8-Cineole (eucalyptol), a monoterpene, has been widely reported for the anti-inflammatory effects. Our previous data confirmed that 1,8-cineole ameliorated the inflammatory phenotype of human umbilical vein endothelial cells (HUVECs) by mediating NF-κB expression in vitro. At present, we investigated the protection effects of 1,8-cineole on vascular endothelium in lipopolysaccharide (LPS)-induced acute inflammatory injury mice and the potential mechanisms involved in the protection in HUVECs. Results from enzyme linked immunosorbent assays revealed that 1,8-cineole suppressed the secretion of interleukin (IL)-6 and IL-8 and increased the expression of IL-10 in the serum of LPS-induced mice. 1,8-Cineole reduced the inflammatory infiltration and the expression of vascular cell adhesion molecular 1 (VCAM-1) in the sections of thoracic aorta in LPS-induced acute inflammatory mice. Western blotting indicated that 1,8-cineole significantly decreased the phosphorylation of NF-κB p65 and increased the expression of PPAR-γ in the thoracic aorta tissue. 1,8-Cineole increased the expression of PPAR-γ in LPS-induced HUVECs. 1,8-Cineole and rosiglitazone reduced the protein and mRNA levels of VCAM-1, E-selectin, IL-6, and IL-8 in LPS-induced HUVECs, which could be reversed by the action of GW9662 (inhibitor of PPAR-γ). 1,8-Cineole and rosiglitazone blocked the LPS-induced IκBα degradation and NF-κB p65 nucleus translocation, which could be reversed by the pretreatment of GW9662 or silence of PPAR-γ gene. In conclusion, 1,8-cineole attenuated LPS-induced vascular endothelial cells injury via PPAR-γ dependent modulation of NF-κB.
Collapse
Affiliation(s)
- Ke-Gang Linghu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China
| | - Guo-Ping Wu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling-Yun Fu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Hong Yang
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Hai-Zhi Li
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yan Chen
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
- The Department of Pharmaceutics of TCM (the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China
| | - Ling Tao
- The Department of Pharmaceutics of TCM (the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiang-Chun Shen
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
21
|
Akolade JO, Balogun M, Swanepoel A, Ibrahim RB, Yusuf AA, Labuschagne P. Microencapsulation of eucalyptol in polyethylene glycol and polycaprolactone using particles from gas-saturated solutions. RSC Adv 2019; 9:34039-34049. [PMID: 35528904 PMCID: PMC9074077 DOI: 10.1039/c9ra06419b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/29/2019] [Indexed: 12/30/2022] Open
Abstract
Eucalyptol is the natural cyclic ether which constitutes the bulk of terpenoids found in essential oils of Eucalyptus spp. and is used in aromatherapy for treatment of migraine, sinusitis, asthma and stress. It acts by inhibiting arachidonic acid metabolism and cytokine production. Chemical instability and volatility of eucalyptol restrict its therapeutic application and necessitate the need to develop an appropriate delivery system to achieve extended release and enhance its bioactivity. However, the synthesis method of the delivery system must be suitable to prevent loss or inactivation of the drug during processing. In this study, supercritical carbon dioxide (scCO2) was explored as an alternative solvent for encapsulation and co-precipitation of eucalyptol with polyethylene glycol (PEG) and/or polycaprolactone (PCL) using the particles from gas-saturated solution (PGSS) process. Polymers and eucalyptol were pre-mixed and then processed in a PGSS autoclave at 45 °C and 80 bar for 1 h. The mixture in scCO2 was micronized and characterized. The presence of eucalyptol in the precipitated particles was confirmed by infrared spectroscopy, gas chromatography and mass spectrometry. The weight ratios of PEG–PCL blends significantly influenced loading capacity and encapsulation efficiency with 77% of eucalyptol encapsulated in a 4 : 1 composite blend of PEG–PCL. The particle size distribution of the PGSS-micronized particles ranged from 30 to 260 μm. ScCO2 assisted microencapsulation in PEG and PCL reduced loss of the volatile drug during a two-hour vaporization study and addition of PCL extended the mean release time in simulated physiological fluids. Free radical scavenging and lipoxygenase inhibitory activities of eucalyptol formulated in the PGSS-micronized particles was sustained. Findings from this study showed that the scCO2-assisted micronization can be used for encapsulation of volatile drugs in polymeric microparticles without affecting bioactivity of the drug. Application of supercritical carbon dioxide as an alternative solvent for microformulation of the volatile unstable drug, eucalyptol in polymeric composites.![]()
Collapse
Affiliation(s)
- Jubril Olayinka Akolade
- Polymers and Composites, Chemicals Cluster, CSIR
- South Africa
- Biotechnology Advanced Research Centre
- Sheda Science and Technology Complex
- Nigeria
| | | | - Andri Swanepoel
- Polymers and Composites, Chemicals Cluster, CSIR
- South Africa
| | | | | | | |
Collapse
|