1
|
Berežni S, Mimica-Dukić N, Domina G, Raimondo FM, Orčić D. Anthriscus sylvestris-Noxious Weed or Sustainable Source of Bioactive Lignans? PLANTS (BASEL, SWITZERLAND) 2024; 13:1087. [PMID: 38674496 PMCID: PMC11053937 DOI: 10.3390/plants13081087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Anthriscus sylvestris (L.) Hoffm. (Apiaceae), commonly known as wild chervil, has gained scientific interest owing to its diverse phytochemical profile and potential therapeutic applications. The plant, despite being categorized as a noxious weed, is traditionally used in treating various conditions like headaches, dressing wounds, and as a tonic, antitussive, antipyretic, analgesic, and diuretic. Its pharmacological importance stems from containing diverse bioactive lignans, especially aryltetralins and dibenzylbutyrolactones. One of the main compounds of A. sylvestris, deoxypodophyllotoxin, among its wide-ranging effects, including antitumor, antiproliferative, antiplatelet aggregation, antiviral, anti-inflammatory, and insecticidal properties, serves as a pivotal precursor to epipodophyllotoxin, crucial in the semisynthesis of cytostatic agents like etoposide and teniposide. The main starting compound for these anticancer medicines was podophyllotoxin, intensively isolated from Sinopodophyllum hexandrum, now listed as an endangered species due to overexploitation. Since new species are being investigated as potential sources, A. sylvestris emerges as a highly promising candidate owing to its abundant lignan content. This review summarizes the current knowledge on A. sylvestris, investigating its biological and morphological characteristics, and pharmacological properties. Emphasizing the biological activities and structure-activity relationship, this review underscores its therapeutic potential, thus encouraging further exploration and utilization of this valuable plant resource.
Collapse
Affiliation(s)
- Sanja Berežni
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| | - Neda Mimica-Dukić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| | - Gianniantonio Domina
- Department of Agricultural, Food and Forest Sciences, University of Palermo, Viale delle Scienze, bldg. 4, 90128 Palermo, Italy;
| | - Francesco Maria Raimondo
- PLANTA/Center for Research, Documentation and Training, Via Serraglio Vecchio 28, 90123 Palermo, Italy;
| | - Dejan Orčić
- Department of Chemistry, Biochemistry and Environmental Protection, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia; (N.M.-D.); (D.O.)
| |
Collapse
|
2
|
Shen Y, Sheng R, Guo R. Application of Zebrafish as a Model for Anti-Cancer Activity Evaluation and Toxicity Testing of Natural Products. Pharmaceuticals (Basel) 2023; 16:827. [PMID: 37375774 DOI: 10.3390/ph16060827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Developing natural product-based anti-cancer drugs/agents is a promising way to overcome the serious side effects and toxicity of traditional chemotherapeutics for cancer treatment. However, rapid assessment of the in vivo anti-cancer activities of natural products is a challenge. Alternatively, zebrafish are useful model organisms and perform well in addressing this challenging issue. Nowadays, a growing number of studies have utilized zebrafish models to evaluate the in vivo activities of natural compounds. Herein, we reviewed the application of zebrafish models for evaluating the anti-cancer activity and toxicity of natural products over the past years, summarized its process and benefits, and provided future outlooks for the development of natural product-based anti-cancer drugs.
Collapse
Affiliation(s)
- Yifan Shen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Ruilong Sheng
- CQM-Centro de Química da Madeira, Campus da Penteada, Universidade da Madeira, 9000-390 Funchal, Portugal
| | - Ruihua Guo
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China
| |
Collapse
|
3
|
Zhang A, Kong M, Zhang X, Pei Z. Mechanism of action of CTRP6 in the regulation of tumorigenesis in the digestive system. Oncol Lett 2022; 24:391. [PMID: 36276484 PMCID: PMC9533366 DOI: 10.3892/ol.2022.13511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
Tumors of the digestive system have always received attention, and their occurrence and development are regulated by various mechanisms such as inflammation and immunity, glucose and lipid metabolism, and tumor angiogenesis. Complement Clq/TNF-related protein 6 (CTRP6) is a member of the CTRP family; it is widely expressed in various tissues and cell types, and plays a biological role in a number of mechanisms, such as glucose and lipid metabolism and inflammation. Recent studies have revealed the tumor-promoting effect of CTRP6 in gastric cancer, liver cancer, colorectal cancer and other gastrointestinal tumors, but, to the best of our knowledge, there has been no systematic discussion on the tumor-promoting mechanism of CTRP6. The present study reviews the role of CTRP6 in tumors of the digestive system and its possible mechanisms.
Collapse
Affiliation(s)
- Aimin Zhang
- Department of Nutriology, Shijiazhuang Second Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Mowei Kong
- Department of Cardiology, Guiqian International General Hospital, Guiyang, Guizhou 550018, P.R. China
| | - Xiuyun Zhang
- Department of Nutriology, Shijiazhuang Second Hospital, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhenying Pei
- Department of Cardiology, Guiqian International General Hospital, Guiyang, Guizhou 550018, P.R. China
| |
Collapse
|
4
|
Seo JH, Yoon G, Park S, Shim JH, Chae JI, Jeon YJ. Deoxypodophyllotoxin Induces ROS-Mediated Apoptosis by Modulating the PI3K/AKT and p38 MAPK-Dependent Signaling in Oral Squamous Cell Carcinoma. J Microbiol Biotechnol 2022; 32:1103-1109. [PMID: 36039387 PMCID: PMC9628964 DOI: 10.4014/jmb.2207.07012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022]
Abstract
Deoxypodophyllotoxin (DPT), a naturally occurring flavonolignan, possesses several pharmacological properties, including anticancer property. However, the mechanisms underlying DPT mode of action in oral squamous cell carcinoma (OSCC) remain unknown. This study aimed to investigate the anticancer effects of DPT on OSCC and the underlying mechanisms. Results of the MTT assay revealed that DPT significantly reduced the cell viability in a time- and dose-dependent manner. Flow cytometry analysis revealed that DPT induces apoptosis in OSCC cells in a dose-dependent manner. Moreover, DPT enhanced the production of mitochondrial reactive oxygen species (ROS) in OSCC cells. Mechanistically, DPT induced apoptosis in OSCC cells by suppressing the PI3K/AKT signaling pathway while activating the p38 MAPK signaling to regulate the expression of apoptotic proteins. Treatment with SC79, an AKT activator, reversed the effects of DPT on AKT signaling in OSCC cells. Taken together, these results provide the basis for the use of DPT in combination with conventional chemotherapy for the treatment of oral cancer.
Collapse
Affiliation(s)
- Ji-Hye Seo
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea
| | - Seryoung Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Muan‐Gun, Jeonnam, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea,
J.I. Chae Phone:+82-63-270-4024 Fax:+82-63-270-4037 E-mail:
| | - Young-Joo Jeon
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea,Corresponding authors Y.J. Jeon Phone:+82-42-860-4386 Fax:+82-42-860-8596 E-mail:
| |
Collapse
|
5
|
Ai Y, Zhao Z, Wang H, Zhang X, Qin W, Guo Y, Zhao M, Tang J, Ma X, Zeng J. Pull the plug: Anti‐angiogenesis potential of natural products in gastrointestinal cancer therapy. Phytother Res 2022; 36:3371-3393. [PMID: 35871532 DOI: 10.1002/ptr.7492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/13/2022] [Accepted: 04/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Yanling Ai
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Hengyi Wang
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiaomei Zhang
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Weihan Qin
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Yanlei Guo
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Maoyuan Zhao
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
- Department of Geriatrics Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
6
|
Shan C, Zhao L, Shi Y, Zhang S, Wu H, Yang M, Yang Q, Wu J. Transcriptome analyses reveal the expression profile of genes related to lignan biosynthesis in Anthriscus sylvestris L. Hoffm. Gen. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2022; 28:333-346. [PMID: 35400889 PMCID: PMC8943078 DOI: 10.1007/s12298-022-01156-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/14/2022] [Accepted: 02/27/2022] [Indexed: 06/14/2023]
Abstract
UNLABELLED Anthriscus sylvestris L. Hoffm. Gen (A. sylvestris) is a perennial herb widely used for antitussive and diuretic purposes in traditional Korean and Chinese medicine. Lignans are critical secondary metabolites with widely pharmacological activities in A. sylvestris. Using transcriptome data of A. sylvestris, we identified genes related to lignan biosynthesis. In all, 123,852 unigenes were obtained from the flowers, leaves, roots, and stems of A. sylvestris with the Illumina HiSeq 4000 platform. The average length of unigenes was 1,123 bp and 91,217 (73.65%) of them were annotated in public databases. Differentially expressed genes and root-specific genes were analyzed between roots and the other three tissue types by comparing gene expression profiles. Specifically, the key enzyme genes involved in lignan biosynthesis were identified and analyzed. The expression levels of some of these genes were highest in the roots, consistent with the accumulation of deoxypodophyllotoxin. These expression levels were experimentally verified via quantitative real-time polymerase chain reaction (qRT-PCR). This research provides valuable information on the transcriptome data of A. sylvestris and the identification of candidate genes associated with the biosynthesis of lignans, laying the foundation for further research on genomics in A. sylvestris and related species. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s12298-022-01156-w.
Collapse
Affiliation(s)
- Chunmiao Shan
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Liqiang Zhao
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Yuanyuan Shi
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Shengxiang Zhang
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Huan Wu
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Mo Yang
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
| | - Qingshan Yang
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, 230012 China
| | - Jiawen Wu
- Anhui University of Chinese Medicine and Anhui Academy of Chinese Medicine, Hefei, 230038 China
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038 China
- Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, 230012 China
| |
Collapse
|
7
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
8
|
Dai Y, Amenov A, Ignatyeva N, Koschinski A, Xu H, Soong PL, Tiburcy M, Linke WA, Zaccolo M, Hasenfuss G, Zimmermann WH, Ebert A. Troponin destabilization impairs sarcomere-cytoskeleton interactions in iPSC-derived cardiomyocytes from dilated cardiomyopathy patients. Sci Rep 2020; 10:209. [PMID: 31937807 PMCID: PMC6959358 DOI: 10.1038/s41598-019-56597-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
The sarcomeric troponin-tropomyosin complex is a critical mediator of excitation-contraction coupling, sarcomeric stability and force generation. We previously reported that induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from patients with a dilated cardiomyopathy (DCM) mutation, troponin T (TnT)-R173W, display sarcomere protein misalignment and impaired contractility. Yet it is not known how TnT mutation causes dysfunction of sarcomere microdomains and how these events contribute to misalignment of sarcomeric proteins in presence of DCM TnT-R173W. Using a human iPSC-CM model combined with CRISPR/Cas9-engineered isogenic controls, we uncovered that TnT-R173W destabilizes molecular interactions of troponin with tropomyosin, and limits binding of PKA to local sarcomere microdomains. This attenuates troponin phosphorylation and dysregulates local sarcomeric microdomains in DCM iPSC-CMs. Disrupted microdomain signaling impairs MYH7-mediated, AMPK-dependent sarcomere-cytoskeleton filament interactions and plasma membrane attachment. Small molecule-based activation of AMPK can restore TnT microdomain interactions, and partially recovers sarcomere protein misalignment as well as impaired contractility in DCM TnT-R173W iPSC-CMs. Our findings suggest a novel therapeutic direction targeting sarcomere- cytoskeleton interactions to induce sarcomere re-organization and contractile recovery in DCM.
Collapse
Affiliation(s)
- Yuanyuan Dai
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Asset Amenov
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Nadezda Ignatyeva
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Andreas Koschinski
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Hang Xu
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Poh Loong Soong
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Wolfgang A Linke
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Gerd Hasenfuss
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology, University of Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany
| | - Antje Ebert
- Heart Center, Department of Cardiology and Pneumology, Goettingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site, Goettingen, Germany.
| |
Collapse
|
9
|
Chang Z, Zhang P, Zhang M, Jun F, Hu Z, Yang J, Wu Y, Zhou R. Aloperine suppresses human pulmonary vascular smooth muscle cell proliferation via inhibiting inflammatory response. CHINESE J PHYSIOL 2019; 62:157-165. [PMID: 31535631 DOI: 10.4103/cjp.cjp_27_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abnormal pulmonary arterial vascular smooth muscle cells (PASMCs) proliferation is critical pathological feature of pulmonary vascular remodeling that acts as driving force in the initiation and development of pulmonary arterial hypertension (PAH), ultimately leading to pulmonary hypertension. Aloperine is a main active alkaloid extracted from the traditional Chinese herbal Sophora alopecuroides and possesses outstanding antioxidation and anti-inflammatory effects. Our group found Aloperine has protective effects on monocroline-induced pulmonary hypertension in rats by inhibiting oxidative stress in previous researches. However, the anti-inflammation effects of Aloperine on PAH remain unclear. Therefore, to further explore whether the beneficial role of Aloperine on PAH was connected with its anti-inflammatory effects, we performed experiments in vitro. Aloperine significantly inhibited the proliferation and DNA synthesis of human pulmonary artery smooth muscle cells (HPASMCs) induced by platelet-derived growth factor-BB, blocked progression through G0/G1to S phase of the cell cycle and promoted total ratio of apoptosis. In summary, these results suggested that Aloperine negatively regulated nuclear factor-κB signaling pathway activity to exert protective effects on PAH and suppressed HPASMCs proliferation therefore has a potential value in the treatment of pulmonary hypertension by negatively modulating pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Zhi Chang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Peng Zhang
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Min Zhang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Feng Jun
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Zhiqiang Hu
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Jiamei Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yuhua Wu
- General Hospital of Ningxia Medical University, Yinchuan 750004, PR China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan 750004, PR China
| |
Collapse
|
10
|
Zhang M, Chang Z, Zhang P, Jing Z, Yan L, Feng J, Hu Z, Xu Q, Zhou W, Ma P, Hao Y, Zhou R. Protective effects of 18β-glycyrrhetinic acid on pulmonary arterial hypertension via regulation of Rho A/Rho kinsase pathway. Chem Biol Interact 2019; 311:108749. [DOI: 10.1016/j.cbi.2019.108749] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 11/28/2022]
|
11
|
Tavares WR, Seca AML. The Current Status of the Pharmaceutical Potential of Juniperus L. Metabolites. MEDICINES 2018; 5:medicines5030081. [PMID: 30065158 PMCID: PMC6165314 DOI: 10.3390/medicines5030081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022]
Abstract
Background: Plants and their derived natural compounds possess various biological and therapeutic properties, which turns them into an increasing topic of interest and research. Juniperus genus is diverse in species, with several traditional medicines reported, and rich in natural compounds with potential for development of new drugs. Methods: The research for this review were based in the Scopus and Web of Science databases using terms combining Juniperus, secondary metabolites names, and biological activities. This is not an exhaustive review of Juniperus compounds with biological activities, but rather a critical selection taking into account the following criteria: (i) studies involving the most recent methodologies for quantitative evaluation of biological activities; and (ii) the compounds with the highest number of studies published in the last four years. Results: From Juniperus species, several diterpenes, flavonoids, and one lignan were emphasized taking into account their level of activity against several targets. Antitumor activity is by far the most studied, being followed by antibacterial and antiviral activities. Deoxypodophyllotoxin and one dehydroabietic acid derivative appears to be the most promising lead compounds. Conclusions: This review demonstrates the Juniperus species value as a source of secondary metabolites with relevant pharmaceutical potential.
Collapse
Affiliation(s)
- Wilson R Tavares
- Faculty of Sciences and Technology, University of Azores, 9501-801 Ponta Delgada, Portugal.
| | - Ana M L Seca
- Department of Chemistry & QOPNA-Organic Chemistry, Natural Products and Food Stuffs, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
- cE3c-Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group & Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9501-321 Ponta Delgada, Portugal.
| |
Collapse
|
12
|
Zang X, Wang G, Cai Q, Zheng X, Zhang J, Chen Q, Wu B, Zhu X, Hao H, Zhou F. A Promising Microtubule Inhibitor Deoxypodophyllotoxin Exhibits Better Efficacy to Multidrug-Resistant Breast Cancer than Paclitaxel via Avoiding Efflux Transport. Drug Metab Dispos 2018; 46:542-551. [PMID: 29523600 DOI: 10.1124/dmd.117.079442] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/22/2018] [Indexed: 02/13/2025] Open
Abstract
Multidrug resistance (MDR) is a common limitation for the clinical use of microtubule-targeting chemotherapeutic agents, and it is the main factor for poor prognoses in cancer therapy. Here, we report on deoxypodophyllotoxin (DPT), a promising microtubule inhibitor in phase 1, as a promising candidate to circumvent this obstacle. DPT remarkably suppressed tumor growth in xenograft mice bearing either paclitaxel (PTX)-sensitive MCF-7/S or acquired resistance MCF-7/Adr (MCF-7/A) cells. Also, DPT exhibited similar accumulation in both tumors, whereas PTX displayed much a lower accumulation in the resistant tumors. In vitro, DPT exhibited a much lower resistance index (0.552) than those of PTX (754.5) or etoposide (38.94) in both MCF-7/S and MCF-7/A cells. Flow cytometry analysis revealed that DPT (5 and 10 nM) caused arrest of the G2/M phase in the two cell lines, whereas PTX (up to 10 nM) had no effect on cell-cycle progression of the MCF-7/A cells. Microtubule dynamics assays revealed that DPT destabilized microtubule assembly in a different mode. Cellular pharmacokinetic assays indicated comparable intracellular and subcellular accumulations of DPT in the two cell lines but a much lower retention of PTX in the MCF-7/A cells. Additionally, transport assays revealed that DPT was not the substrate of P-glycoprotein, breast cancer resistance protein, or MDR-associated protein 2, indicating a lower occurrence rate of MDR. DPT might be a promising microtubule inhibitor for breast cancer therapy, especially for treatment of drug-resistant tumors.
Collapse
Affiliation(s)
- Xiaojie Zang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Qingyun Cai
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Xiao Zheng
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Jingwei Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Qianying Chen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Baojin Wu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Xiong Zhu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Haiping Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines (X.Za., G.W., Q.Ca., X.Zhe., J.Z., Q.Ch., H.H., F.Z.), and Medical and Chemical Institute (B.W., X.Zhu.), China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Eriocalyxin B, a natural diterpenoid, inhibited VEGF-induced angiogenesis and diminished angiogenesis-dependent breast tumor growth by suppressing VEGFR-2 signaling. Oncotarget 2018; 7:82820-82835. [PMID: 27756875 PMCID: PMC5347735 DOI: 10.18632/oncotarget.12652] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 10/02/2016] [Indexed: 12/29/2022] Open
Abstract
Eriocalyxin B (EriB), a natural ent-kaurane diterpenoid isolated from the plant Isodon eriocalyx var. laxiflora, has emerged as a promising anticancer agent. The effects of EriB on angiogenesis were explored in the present study. Here we demonstrated that the subintestinal vein formation was significantly inhibited by EriB treatment (10, 15 μM) in zebrafish embryos, which was resulted from the alteration of various angiogenic genes as shown in transcriptome profiling. In human umbilical vein endothelial cells, EriB treatment (50, 100 nM) could significantly block vascular endothelial growth factors (VEGF)-induced cell proliferation, tube formation, cell migration and cell invasion. Furthermore, EriB also caused G1 phase cell cycle arrest which was correlated with the down-regulation of the cyclin D1 and CDK4 leading to the inhibition of phosphorylated retinoblastoma protein expression. Investigation of the signal transduction revealed that EriB inhibited VEGF-induced phosphorylation of VEGF receptor-2 via the interaction with the ATP-binding sites according to the molecular docking simulations. The suppression of VEGFR-2 downstream signal transduction cascades was also observed. EriB was showed to inhibit new blood vessel formation in Matrigel plug model and mouse 4T1 breast tumor model. EriB (5 mg/kg/day) treatment was able to decrease tumor vascularization and suppress tumor growth and angiogenesis. Taken together, our findings suggested that EriB is a novel inhibitor of angiogenesis through modulating VEGFR-2 signaling pathway, which could be developed as a promising anti-angiogenic agent for treatment of angiogenesis-related human diseases, such as cancer.
Collapse
|
14
|
Okuda KS, Lee HM, Velaithan V, Ng MF, Patel V. Utilizing Zebrafish to Identify Anti-(Lymph)Angiogenic Compounds for Cancer Treatment: Promise and Future Challenges. Microcirculation 2018; 23:389-405. [PMID: 27177346 DOI: 10.1111/micc.12289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/11/2016] [Indexed: 12/13/2022]
Abstract
Cancer metastasis which predominantly occurs through blood and lymphatic vessels, is the leading cause of death in cancer patients. Consequently, several anti-angiogenic agents have been approved as therapeutic agents for human cancers such as metastatic renal cell carcinoma. Also, anti-lymphangiogenic drugs such as monoclonal antibodies VGX-100 and IMC-3C5 have undergone phase I clinical trials for advanced and metastatic solid tumors. Although anti-tumor-associated angiogenesis has proven to be a promising therapeutic strategy for human cancers, this approach is fraught with toxicities and development of drug resistance. This emphasizes the need for alternative anti-(lymph)angiogenic drugs. The use of zebrafish has become accepted as an established model for high-throughput screening, vascular biology, and cancer research. Importantly, various zebrafish transgenic lines have now been generated that can readily discriminate different vascular compartments. This now enables detailed in vivo studies that are relevant to both human physiological and tumor (lymph)angiogenesis to be conducted in zebrafish. This review highlights recent advancements in the zebrafish anti-vascular screening platform and showcases promising new anti-(lymph)angiogenic compounds that have been derived from this model. In addition, this review discusses the promises and challenges of the zebrafish model in the context of anti-(lymph)angiogenic compound discovery for cancer treatment.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Hui Mei Lee
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Vithya Velaithan
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Mei Fong Ng
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| | - Vyomesh Patel
- Drug Discovery, Cancer Research Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
15
|
Jeon D, Kim SJ, Kim HS. Anti-inflammatory evaluation of the methanolic extract of Taraxacum officinale in LPS-stimulated human umbilical vein endothelial cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:508. [PMID: 29187173 PMCID: PMC5707789 DOI: 10.1186/s12906-017-2022-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/20/2017] [Indexed: 12/27/2022]
Abstract
Background Atherosclerosis is a chronic vascular inflammatory disease. Since even low-level endotoxemia constitutes a powerful and independent risk factor for the development of atherosclerosis, it is important to find therapies directed against the vascular effects of endotoxin to prevent atherosclerosis. Taraxacum officinale (TO) is used for medicinal purposes because of its choleretic, diuretic, antioxidative, anti-inflammatory, and anti-carcinogenic properties, but its anti-inflammatory effect on endothelial cells has not been established. Methods We evaluated the anti-inflammatory activity of TO filtered methanol extracts in LPS-stimulated human umbilical vein endothelial cells (HUVECs) by monocyte adhesion and western blot assays. HUVECs were pretreated with 100 μg/ml TO for 1 h and then incubated with 1 μg/ml LPS for 24 h. The mRNA and protein expression levels of the targets (pro-inflammatory cytokines and adhesion molecules) were analyzed by real-time PCR and western blot assays. We also preformed HPLC analysis to identify the components of the TO methanol extract. Results The TO filtered methanol extracts dramatically inhibited LPS-induced endothelial cell–monocyte interactions by reducing vascular cell adhesion molecule-1 and monocyte chemoattractant protein-1, and pro-inflammatory cytokine expression. TO suppressed the LPS-induced nuclear translocation of NF-κB, whereas it did not affect MAPK activation. Conclusions Our findings demonstrated that methanol extracts of TO could attenuate LPS-induced endothelial cell activation by inhibiting the NF-κB pathway. These results indicate the potential clinical benefits and applications of TO for the prevention of vascular inflammation and atherosclerosis. Electronic supplementary material The online version of this article (10.1186/s12906-017-2022-7) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Wu F, Yao W, Yang J, Zhang M, Xu Y, Hao Y, Yan L, Niu Y, Sun T, Yu J, Zhou R. Protective effects of aloperin on monocroline-induced pulmonary hypertension via regulation of Rho A/Rho kinsase pathway in rats. Biomed Pharmacother 2017; 95:1161-1168. [PMID: 28926926 DOI: 10.1016/j.biopha.2017.08.126] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/22/2017] [Accepted: 08/29/2017] [Indexed: 12/30/2022] Open
Abstract
Pulmonary hypertension (PH) is fatal disease which closely involves Rho A/ Rho kinsase (ROCK) pathway. Aloperine is a main active alkaloid extracted from Sophora alopecuroides, which is a traditional Chinese herbal medicine that has been used widely. However, the effects of this alkaloid on pulmonary hypertension and its mechanisms remain unclear. Therefore, this study is designed to investigate whether aloperine has protective effects on PH induced by monocrotaline, whether these effects may be related to regulation of RhoA/ROCK pathway in rats. Pulmonary hypertension was induced by monocrotaline (60mg/kg), and subsequently oral administration of aloperine (25, 50, 100mg/kg/day) for 21 days. At the end of the experiment, rats were underwent hemodynamic and morphologic assessments. At same time, the expression of Rho A, ROCK1, ROCK2, as well as activities of ROCK in the lung of rat has been detected. Afterwards, the expression of p27kip1, Bax, Bcl-2, which was the downstream proliferation and apoptosis factors of ROCK, were tested. The result indicted that aloperine treatment showed significantly improvement in hemodynamic and pathomorphologic data. Moreover, the reduction in expression of Rho A, ROCK1, ROCK2, and suppression in activities of ROCK were found in rat lungs after aloperine treatment. Furthermore, aloperine also alleviated the MCT-induced changes of p27kip1, Bax and Bcl-2. In summary, this study indicates that aloperine have protective effects on monocrotaline-induced PH. And these effects may be partially related to RhoA/ROCK pathway. Thus, aloperine could be considered a possible therapeutic strategy for PH.
Collapse
Affiliation(s)
- Fan Wu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Wanxia Yao
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Jiamei Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Min Zhang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Yanping Xu
- Echocardiogram Room, Heart Center, General Hospital of Ningxia Medical University, Yinchuan, 750004, PR China
| | - Yinju Hao
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Lin Yan
- College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Yang Niu
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, 750004, PR China.
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, PR China; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, PR China; Ningxia Hui Medicine Modern Engineering Research Center and Collaborative Innovation Center, Ningxia Medical University, Yinchuan, 750004, PR China.
| |
Collapse
|
17
|
Pérez-Pérez MJ, Priego EM, Bueno O, Martins MS, Canela MD, Liekens S. Blocking Blood Flow to Solid Tumors by Destabilizing Tubulin: An Approach to Targeting Tumor Growth. J Med Chem 2016; 59:8685-8711. [DOI: 10.1021/acs.jmedchem.6b00463] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | - Eva-María Priego
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Oskía Bueno
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | | | - María-Dolores Canela
- Instituto de Química Médica (IQM-CSIC), Juan de la Cierva 3, E-28006 Madrid, Spain
| | - Sandra Liekens
- Rega
Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|