1
|
Lin S, Tang RWL, Ye Y, Xia C, Wu J, Duan R, Leung KW, Dong TTX, Tsim KWK. Drug Screening of Flavonoids as Potential VEGF Inhibitors Through Computational Docking and Cell Models. Molecules 2025; 30:257. [PMID: 39860127 PMCID: PMC11767819 DOI: 10.3390/molecules30020257] [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: 12/10/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Vascular endothelial growth factor (VEGF), also known as VEGF-A, has been linked to various diseases, such as wet age-related macular degeneration (wAMD) and cancer. Even though there are VEGF inhibitors that are currently commercially available in clinical applications, severe adverse effects have been associated with these treatments. There is still a need to develop novel VEGF-based therapeutics against these VEGF-related diseases. Here, we established a series of VEGF-based computational docking analyses and cell models, such as a wound healing assay in HaCaT cells and an evaluation of NF-κB performance in macrophages, to screen a large library of flavonoid-type phytochemicals. Three flavonoids, namely, farrerol, ononin and (-)-epicatechin, were shown to express binding affinities to VEGF protein and inhibit VEGF-mediated biological activities. The investigation evidently suggested that the three flavonoids above could be considered potential anti-VEGF agents for the following drug development against VEGF-mediated diseases.
Collapse
Affiliation(s)
- Shengying Lin
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Roy Wai-Lun Tang
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yutong Ye
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Chenxi Xia
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jiahui Wu
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ran Duan
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ka-Wing Leung
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Tina Ting-Xia Dong
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Karl Wah-Keung Tsim
- Center for Chinese Medicine, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; (S.L.); (R.W.-L.T.); (Y.Y.); (C.X.); (J.W.); (R.D.); (K.-W.L.); (T.T.-X.D.)
- State Key Laboratory of Molecular Neuroscience, Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
2
|
Liu S, Ai Z, Hu Y, Ren G, Zhang J, Tang P, Zou H, Li X, Wang Y, Nan B, Wang Y. Ginseng glucosyl oleanolate inhibit cervical cancer cell proliferation and angiogenesis via PI3K/AKT/HIF-1α pathway. NPJ Sci Food 2024; 8:105. [PMID: 39702347 PMCID: PMC11659631 DOI: 10.1038/s41538-024-00341-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
Ginseng (Panax ginseng C.A. Meyer) is widely used in several functional foods at present. Ginsenosides, is the most crucial bioactive constituents in ginseng whose antitumor activity have been widely reported. In this study, the effect of ginseng glucosyl oleanolate (GGO) produced from ginsenoside Ro through enzymatic transformation, on cervical cancer was evaluated in vitro and in vivo. GGO significantly inhibited the viability and colony forming ability of HeLa cells, and blocked the cell cycle in G0/G1 phase, which showed its ability to inhibit the proliferation of HeLa cells. GGO exhibited anti-angiogenesis effect in HUVECs, chick chorioallantoic membrane (CAM) and Matrigel plugs model. These effects were related to interference with the paracrine axis of VEGF/VEGFR2 and blockage of the downstream PI3K/AKT/HIF-1α signaling pathway of the autocrine axis. The dual inhibitory effects of GGO were also exhibited in immunocompromised mice undergoing heterograft and suppressed tumor growth without any side effects. These findings provide a theoretical basis for further development of GGO as a functional food with anti-tumor properties.
Collapse
Affiliation(s)
- Sitong Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Zhiyi Ai
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yue Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Guangquan Ren
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Junshun Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Ping Tang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Hongyang Zou
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Xia Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yu Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Bo Nan
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yuhua Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China.
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China.
- National Processing Laboratory for Soybean Industry and Technology, Changchun, China.
- National Engineering Research Center for Wheat and Cord Deep Processing, Changchun, China.
| |
Collapse
|
3
|
Nowacka A, Śniegocka M, Smuczyński W, Liss S, Ziółkowska E, Bożiłow D, Śniegocki M, Wiciński M. The Potential Application of Resveratrol and Its Derivatives in Central Nervous System Tumors. Int J Mol Sci 2024; 25:13338. [PMID: 39769099 PMCID: PMC11728356 DOI: 10.3390/ijms252413338] [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: 11/23/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Resveratrol, a naturally occurring polyphenolic compound found in various plants, has been extensively studied for its broad spectrum of beneficial biological effects. These encompass its potent antioxidant properties, anti-inflammatory activities, anti-aging capabilities, cardioprotective functions, and neuroprotective potential. The diverse biological actions of resveratrol extend beyond these well-established properties. It also exerts a significant impact on metabolic processes and bioavailability, and critically, it demonstrates the ability to effectively traverse the blood-brain barrier. This capacity to penetrate the central nervous system renders resveratrol a promising therapeutic agent for the management of central nervous system malignancies, as it has been shown to inhibit tumor cell proliferation, induce apoptosis, and modulate key signaling cascades, such as PI3K/Akt, JAK/STAT, and NF-kB. The multifaceted nature of resveratrol's biological effects, including its influence on diverse physiological processes, underscores its potential as a valuable therapeutic option for the treatment of central nervous system tumors.
Collapse
Affiliation(s)
- Agnieszka Nowacka
- Department of Neurosurgery, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Martyna Śniegocka
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14-16, 00161 Rome, Italy
| | - Wojciech Smuczyński
- Department of Physiotherapy, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Techników 3, 85-801 Bydgoszcz, Poland
| | - Sara Liss
- Department of Pharmacology and Therapeutics, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-090 Bydgoszcz, Poland
| | - Ewa Ziółkowska
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dominika Bożiłow
- Anaesthesiology and Intensive Care Clinical Ward, The 10th Military Research Hospital and Polyclinic, ul. Powstańców Warszawy 5, 85-681 Bydgoszcz, Poland
| | - Maciej Śniegocki
- Department of Neurosurgery, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Michał Wiciński
- Department of Pharmacology and Therapeutics, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-090 Bydgoszcz, Poland
| |
Collapse
|
4
|
Azevedo T, Ferreira T, Peña‐Corona SI, Cortes H, Silva‐Reis R, da Costa RMG, Faustino‐Rocha AI, Oliveira PA, Calina D, Cardoso SM, Büsselberg D, Leyva‐Gómez G, Sharifi‐Rad J, Cho WC. Natural products‐based antiangiogenic agents: New frontiers in cancer therapy. FOOD FRONTIERS 2024; 5:2423-2466. [DOI: 10.1002/fft2.466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAngiogenesis, vital for tumor growth and metastasis, is a promising target in cancer therapy. Natural compounds offer potential as antiangiogenic agents with reduced toxicity. This review provides a comprehensive overview of natural product‐based antiangiogenic therapies, focusing on molecular mechanisms and therapeutic potential. A systematic search identified relevant articles from 2019 to 2023. Various natural compounds, including polyphenols, terpenes, alkaloids, cannabinoids, omega‐3 fatty acids, polysaccharides, proteins, and carotenoids, were investigated for their antiangiogenic properties. Challenges such as dose standardization, routes of administration, and potential side effects remain. Further studies, including in‐depth animal models and human epidemiological studies, must elucidate clinical efficacy and safety. Synergistic effects with current antiangiogenic therapies, such as bevacizumab and tyrosine kinase inhibitors, should be explored. Additionally, the potential hormone‐dependent effects of compounds like genistein highlight the need for safety evaluation. In conclusion, natural products hold promise as adjunctive therapies to conventional antineoplastic drugs in modulating angiogenesis in cancer. However, robust clinical trials are needed to validate preclinical findings and ensure safety and efficacy.
Collapse
Affiliation(s)
- Tiago Azevedo
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Tiago Ferreira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Ciudad de México Mexico
| | - Rita Silva‐Reis
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | - Rui M. Gil da Costa
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC) Porto Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering University of Porto Porto Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering University of Porto Porto Portugal
- Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology Federal University of Maranhão (UFMA), UFMA University Hospital (HUUFMA) São Luís Brazil
| | - Ana I. Faustino‐Rocha
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology University of Évora Evora Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy University of Medicine and Pharmacy of Craiova Craiova Romania
| | - Susana M. Cardoso
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | | | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Javad Sharifi‐Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo Veracruz Mexico
- Department of Medicine, College of Medicine Korea University Seoul Republic of Korea
- Facultad de Medicina Universidad del Azuay Cuenca Ecuador
| | - William C. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Kowloon Hong Kong
| |
Collapse
|
5
|
Shi Y, Shen C, Zhang W. Efficacy and safety of a topical skincare regimen containing CE Ferulic serum and resveratrol BE serum following ablative fractional CO 2 laser treatment: A prospective, randomized, split-face, controlled trial. J Eur Acad Dermatol Venereol 2024; 38 Suppl 6:17-25. [PMID: 38828801 DOI: 10.1111/jdv.20070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/14/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Ablative fractional CO2 laser is widely used to address various skin problems, but the treatment often leads to adverse effects such as erythema, dyspigmentation, and extended recovery periods, negatively impacting patients' quality of life. OBJECTIVES This study aimed to evaluate the efficacy and safety of a topical skincare regimen containing both CE Ferulic serum and Resveratrol BE night serum following fractional CO2 laser treatment in Chinese population. METHODS In this randomized, investigator-blinded, split-face, controlled trial, individuals aged 18-65 undergoing ablative CO2 laser treatment were randomly assigned to apply CE Ferulic plus resveratrol BE serum (CEF-RBE) to either side of face and normal saline (NS) to the other, for 14 consecutive days. The primary endpoint was erythema index (EI) on day 14, with key secondary endpoints including scabbing detachment time, percentage changes in EI and melanin index (MI), skin hydration, transepidermal water loss, skin sebum content, oedema, and overall subject satisfaction. RESULTS The study included 51 patients, of whom 29 (56.9%) were female, with a mean (SD) age of 29.8 (5.39) years. On day 14, the CEF-RBE side exhibited significantly lower EI than the NS side (308.9 vs. 325.3, p = 0.034). The median (IQR) time (days) for complete scabbing detachment at the CEF-RBE side was 6.0 (5.0-8.0) compared to 6.5 (5.0-9.0) at NS side (p = 0.018). Additionally, the CEF-RBE side showed a 7.4% decrease in MI from baseline to day 14, while the NS side experienced a 0.2% increase (Δ = -7.6%, p = 0.044). Throughout the 14-day follow-up, the CEF-RBE side consistently displayed higher skin hydration than the NS side. CONCLUSIONS The study highlighted the benefits of incorporating CEF-RBE following laser treatment in reducing erythema and hyperpigmentation, promoting wound healing, and maintaining skin hydration, although limitations such as contamination and adherence issues should be considered.
Collapse
Affiliation(s)
- Yu Shi
- Department of Medical Cosmetology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chen Shen
- Department of Medical Cosmetology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Zhang
- Department of Medical Cosmetology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
6
|
Liu ZB, Fan XY, Wang CW, Ye X, Wu CJ. Potentially active compounds that improve PAD through angiogenesis: A review. Biomed Pharmacother 2023; 168:115634. [PMID: 37879211 DOI: 10.1016/j.biopha.2023.115634] [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: 08/08/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Peripheral arterial disease (PAD) has been historically neglected, which has resulted in a lack of effective drugs in clinical practice. However, with the increasing prevalence of diseases like atherosclerosis and diabetes, the incidence of PAD is rising and cannot be ignored. Researchers are exploring the potential of promoting angiogenesis through exogenous compounds to improve PAD. This paper focuses on the therapeutic effect of natural products (Salidroside, Astragaloside IV, etc.) and synthetic compounds (Cilostazol, Dapagliflozin, etc.). Specifically, it examines how they can promote autocrine secretion of vascular endothelial cells, enhance cell paracrine interactions, and regulate endothelial progenitor cell function. The activation of these effects may be closely related to PI3K, AMPK, and other pathways. Overall, these exogenous compounds have promising therapeutic potential for PAD. This study aims to summarize the potential active compounds, provide a variety of options for the search for drugs for the treatment of PAD, and bring light to the treatment of patients.
Collapse
Affiliation(s)
- Zi-Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin-Yun Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen-Wei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xun Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chun-Jie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu Univesity of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
7
|
Gao AX, Xia TC, Lin LS, Dong TT, Tsim KW. The neurotrophic activities of brain-derived neurotrophic factor are potentiated by binding with apigenin, a common flavone in vegetables, in stimulating the receptor signaling. CNS Neurosci Ther 2023; 29:2787-2799. [PMID: 37101380 PMCID: PMC10493664 DOI: 10.1111/cns.14230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023] Open
Abstract
AIMS We aimed to identify the neurotrophic activities of apigenin (4',5,7-trihydroxyflavone) via its coordination with brain-derived neurotrophic factor (BNDF) and an elevated signaling of tyrosine kinase receptor B (Trk B receptor). METHODS The direct binding of apigenin to BDNF was validated by ultrafiltration and biacore assay. Neurogenesis, triggered by apigenin and/or BDNF, was determined in cultured SH-SY5Y cells and rat cortical neurons. The amyloid-beta (Aβ)25-35 -induced cellular stress was revealed by propidium iodide staining, mitochondrial membrane potential, bioenergetic analysis, and formation of reactive oxygen species levels. Activation of Trk B signaling was tested by western blotting. RESULTS Apigenin and BDNF synergistically maintained the cell viability and promoted neurite outgrowth of cultured neurons. In addition, the BDNF-induced neurogenesis of cultured neurons was markedly potentiated by applied apigenin, including the induced expressions of neurofilaments, PSD-95 and synaptotagmin. Moreover, the synergy of apigenin and BDNF alleviated the (Aβ)25-35 -induced cytotoxicity and mitochondrial dysfunction. The synergy could be accounted by phosphorylation of Trk B receptor, and which was fully blocked by a Trk inhibitor K252a. CONCLUSION Apigenin potentiates the neurotrophic activities of BDNF through direct binding, which may serve as a possible treatment for its curative efficiency in neurodegenerative diseases and depression.
Collapse
Affiliation(s)
- Alex Xiong Gao
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Tracy Chen‐Xi Xia
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Lish Sheng‐Ying Lin
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Tina Ting‐Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Karl Wah‐Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| |
Collapse
|
8
|
Guo GX, Wu KY, Zhang XY, Lai FX, Tsim KWK, Qin QW, Hu WH. The extract of Curcumae Longae Rhizoma suppresses angiogenesis via VEGF-induced PI3K/Akt-eNOS-NO pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116299. [PMID: 36842721 DOI: 10.1016/j.jep.2023.116299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcumae Longae Rhizoma (CLR) is a safe natural herbal medicine, and which has been widely used for centuries as functional food and health products, but its effects on angiogenesis and related underlying mechanism remain unclear. AIM OF THE STUDY The abnormal angiogenesis is closely related with various diseases, and therefore the precise control of angiogenesis is of great importance. The well-known angiogenic factor, vascular endothelial growth factor (VEGF), mediates angiogenesis and induces multiple signalling pathways via binding to VEGF receptor (VEGFR). The attenuation of VEGF-triggered angiogenic-related signalling pathways may relieve various diseases through suppression of angiogenesis. Here, we aimed to elucidate that CLR extract could exert striking anti-angiogenic activities both in vitro and in vivo. MATERIALS AND METHODS The viability of human umbilical vascular endothelial cell (HUVEC) was examined by LDH and MTT assays. Migrative and invasive ability of the endothelial cells were independently evaluated by wound healing and transwell assays. The activities of CLR extract on in vitro angiogenesis was tested by tube formation assay. In vivo vascularization was determined by using zebrafish embryo model in the present of CLR extract. Western blotting was applied to determine the phosphorylated levels of VEGFR2, PI3K, AKT and eNOS. Besides, the levels of nitric oxide (NO) and reactive oxygen species (ROS) were separately evaluated by Griess assay and 2'7'-dichlorofluorescein diacetate reaction. In addition, the cell migrative ability of cancer cell was estimated by using cultured human colon carcinoma cells (HT-29 cell line), and immunofluorescence assay was applied to evaluate the effect of CLR extract on nuclear translocation of NF-κB p65 subunit in the VEGF-treated HT-29 cultures. RESULTS CLR extract significantly suppressed a series of VEGF-mediated angiogenic responses, including endothelial cell proliferation, migration, invasion, and tube formation. Moreover, CLR extract reduced in vivo sub-intestinal vessel formation in zebrafish embryo model. Mechanistically, the extract of CLR attenuated the VEGF-triggered signalling, as demonstrated by decreased level of phosphorylated VEGFR2 and subsequently inactivated its downstream regulators, e.g. phospho-PI3K, phospho-AKT and phospho-eNOS. The production of NO and formation of ROS were markedly inhibited in HUVECs. Furthermore, CLR extract suppressed cell migration and NF-κB translocation in cultured HT-29 cells. CONCLUSIONS These preclinical findings demonstrate that the extract of CLR remarkably attenuates angiogenesis and which has great potential as a natural drug candidate with excellent anti-angiogenic activity.
Collapse
Affiliation(s)
- Guo-Xia Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Ke-Yue Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Xiao-Yong Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| | - Fu-Xiang Lai
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Karl Wah-Keung Tsim
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Qi-Wei Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| | - Wei-Hui Hu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| |
Collapse
|
9
|
Li X, Zhou J, Wang X, Li C, Ma Z, Wan Q, Peng F. New advances in the research of clinical treatment and novel anticancer agents in tumor angiogenesis. Biomed Pharmacother 2023; 163:114806. [PMID: 37163782 DOI: 10.1016/j.biopha.2023.114806] [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: 02/10/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
In 1971, Folkman proposed that tumors could be limited to very small sizes by blocking angiogenesis. Angiogenesis is the generation of new blood vessels from pre-existing vessels, considered to be one of the important processes in tumor growth and metastasis. Angiogenesis is a complex process regulated by various factors and involves many secreted factors and signaling pathways. Angiogenesis is important in the transport of oxygen and nutrients to the tumor during tumor development. Therefore, inhibition of angiogenesis has become an important strategy in the clinical management of many solid tumors. Combination therapies of angiogenesis inhibitors with radiotherapy and chemotherapy are often used in clinical practice. In this article, we will review common targets against angiogenesis, the most common and up-to-date anti-angiogenic drugs and clinical treatments in recent years, including active ingredients from chemical and herbal medicines.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jianbo Zhou
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Wang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunxi Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zifan Ma
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qiaoling Wan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
El-Sheikh M, Mesalam A, Khalil AAK, Idrees M, Ahn MJ, Mesalam AA, Kong IK. Downregulation of PI3K/AKT/mTOR Pathway in Juglone-Treated Bovine Oocytes. Antioxidants (Basel) 2023; 12:antiox12010114. [PMID: 36670976 PMCID: PMC9854430 DOI: 10.3390/antiox12010114] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
We have previously reported that juglone, a natural compound found in Juglandaceae with a wide range of biological activities, can reduces the developmental competence of bovine oocytes. In the current study, we investigated the possible mechanisms behind the toxicity of juglone and the relationship with PI3K/AKT/mTOR signaling during the in vitro maturation (IVM) of oocytes. Results show that oocyte exposure to juglone was associated with a significant decrease in filamentous actin (F-actin) accumulation. The RT-qPCR showed downregulation of the meiosis progression indicator GSK-3A, oocyte development marker BMP15, mitochondria fusion controlling MFN1, oxidative stress-related OGG1, and histone methylation-related EZH1, EZH2, SUZ12, G9a, and SUV39H2 genes in juglone-treated oocytes. In addition, glycolysis- (PFK1 and GLUT1), ATP synthesis- (ATPase8 and ATP5F1B), and OXPHOS-specific markers (SDHA and SDHD), as well as the oocyte survival regulators (SOD2, VEGF, and MAPK1) significantly decreased upon juglone treatment. Moreover, lower expression of PI3K, AKT, and mTOR was observed at the transcriptional and/or translational level(s). The autophagy markers LC3B and beclin-1 as well as the DNA damage-specific marker 8-OxoG displayed overexpression in juglone-exposed oocytes. Taken together, our results show that administration of juglone during the IVM can reduce the quality and developmental health of bovine oocytes through downregulation of the PI3K/AKT/mTOR pathway and its downstream signaling cascades.
Collapse
Affiliation(s)
- Marwa El-Sheikh
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre (NRC), Dokki, Cairo 12622, Egypt
| | - Ayman Mesalam
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Atif Ali Khan Khalil
- Department of Pharmacognosy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Lahore 54000, Pakistan
| | - Muhammad Idrees
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Mi-Jeong Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ahmed Atef Mesalam
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre (NRC), Dokki, Cairo 12622, Egypt
- Correspondence: (A.A.M.); (I.-K.K.)
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Republic of Korea
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
- The King Kong Corp. Ltd., Gyeongsang National University, Jinju 52828, Republic of Korea
- Correspondence: (A.A.M.); (I.-K.K.)
| |
Collapse
|
11
|
R. M. Metawea O, Teleb M, Haiba NS, Elzoghby AO, Khafaga AF, Noreldin AE, Khattab SN, Khalil HH. Folic acid-poly(N-isopropylacrylamide-maltodextrin) nanohydrogels a novel thermo-/pH-responsive polymer for resveratrol breast cancer targeted therapy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Wang P, Shang R, Ma Y, Wang D, Zhao W, Chen F, Hu X, Zhao X. Targeting microbiota-host interactions with resveratrol on cancer: Effects and potential mechanisms of action. Crit Rev Food Sci Nutr 2022; 64:311-333. [PMID: 35917112 DOI: 10.1080/10408398.2022.2106180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Resveratrol (RSV) is a natural polyphenolic compound detected in grapes, berries, and red wine. The anticancer activities of RSV have been observed in vivo and in vitro studies. However, the pharmacology mechanism of RSV is confusing due to its low bioavailability. According to studies of the metabolic characteristics of RSV, the gut intestine is a crucial site of its health benefits. Dietary RSV exhibits a profound effect on the gut microbiota structure and metabolic function. In addition, emerging evidence demonstrates a protective effect of RSV metabolites against carcinogenesis. Therefore, to better understand the anticancer mechanisms of dietary RSV, it is vital to evaluate the role of RSV-microbiota-host interactions in cancer therapy. In this review, we summarized significant findings on the anticancer activities of RSV based on epidemiological, experimental and clinical studies involved in investigating the metabolic characteristics and the traditional anticancer mechanisms of RSV. Special attention is given to the putative mechanisms involving microbiota-host interactions, such as the modulation of gut microecology and the anticancer effects of RSV metabolites. The changes in microbiota-host interactions after RSV supplementation play vital roles in cancer prevention and thus offering a new perspective on nutritional interventions to treat cancer.
Collapse
Affiliation(s)
- Pan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Runze Shang
- Department of General Surgery, Affiliated Haixia Hospital of Huaqiao University (The 910 Hospital), Quanzhou, Fujian, China
| | - Yue Ma
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Dan Wang
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Wenting Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Fang Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaosong Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xiaoyan Zhao
- Institute of Agri-food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
13
|
Hu WH, Zhang XY, Leung KW, Duan R, Dong TX(T, Qin QW, Tsim KWK. Resveratrol, an Inhibitor Binding to VEGF, Restores the Pathology of Abnormal Angiogenesis in Retinopathy of Prematurity (ROP) in Mice: Application by Intravitreal and Topical Instillation. Int J Mol Sci 2022; 23:ijms23126455. [PMID: 35742898 PMCID: PMC9223486 DOI: 10.3390/ijms23126455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/06/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a severe eye disease leading to blindness. Abnormal vessel formation is the pathological hallmark of neovascular ROP. In forming vessels, vascular endothelial growth factor (VEGF) is an important stimulator. The current anti-ROP therapy has focused on bevacizumab, a monoclonal antibody against VEGF, and pazopanib, a tyrosine kinase inhibitor on the VEGF receptor (VEGFR). Several lines of evidence have proposed that natural compounds may be more effective and safer for anti-VEGF function. Resveratrol, a common natural compound, binds to VEGF and blocks its interaction with VEGFR, thereafter suppressing angiogenesis. Here, we evaluate the efficacy of intravitreal injection, or topical instillation (eye drops), of resveratrol into the eyes of mice suffering from oxygen-induced retinopathy, i.e., developing ROP. The treatment of resveratrol significantly relieved the degree of vascular distortion, permeability and hyperplasia; the efficacy could be revealed by both methods of resveratrol application. In parallel, the treatments of resveratrol inhibited the retinal expressions of VEGF, VEGFR and CD31. Moreover, the applied resveratrol significantly relieved the damage caused by oxygen radicals through upregulating the level of superoxide dismutase (SOD) and downregulating the level of malondialdehyde (MDA) in the retina. Taken together, the potential therapeutic benefit of resveratrol in pro-angiogenic diseases, including retinopathy, can be considered.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (X.-Y.Z.); (T.-X.D.); (Q.-W.Q.)
| | - Xiao-Yong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (X.-Y.Z.); (T.-X.D.); (Q.-W.Q.)
| | - Ka-Wing Leung
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518063, China; (K.-W.L.); (R.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518063, China; (K.-W.L.); (R.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ting-Xia (Tina) Dong
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (X.-Y.Z.); (T.-X.D.); (Q.-W.Q.)
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518063, China; (K.-W.L.); (R.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qi-Wei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (X.-Y.Z.); (T.-X.D.); (Q.-W.Q.)
| | - Karl Wah-Keung Tsim
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (X.-Y.Z.); (T.-X.D.); (Q.-W.Q.)
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518063, China; (K.-W.L.); (R.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Correspondence: ; Tel.: +852-2358-7332; Fax: +852-2358-1559
| |
Collapse
|
14
|
Guo MS, Gao X, Hu W, Wang X, Dong TT, Tsim KWK. Scutellarin potentiates the skin regenerative function of Self Growth Colony, an optimized platelet-rich plasma extract, in cultured keratinocytes through VEGF receptor and MAPK signalling. J Cosmet Dermatol 2022; 21:4836-4845. [PMID: 35080332 DOI: 10.1111/jocd.14800] [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: 09/28/2021] [Revised: 01/04/2022] [Accepted: 01/17/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Migration of keratinocyte plays an essential role in wound healing. The proprietary platelet-rich plasma from human blood, named as Self-Growth Colony (SGC), functions in stimulating migration of wounded keratinocytes. And the growth factors, including VEGF, being enriched in SGC could account for this function. Scutellarin, an active phytochemical from root of Scutellaria barbata D. Don, has been proposed to have various pharmacological functions; however, the activity in epidermal skin cells is yet to be explored. Here, the role of scutellarin in potentiating the functionality of SGC to promote the regeneration of wounded keratinocyte was probed. METHODS Molecular docking and ultrafiltration-based LC-MS were performed to verify the binding between scutellarin and VEGF, which potentiated the VEGF-mediated functions. Scratch assay, performed on cultured keratinocytes, was to analyse the treatments of SGC and scutellarin in the process of wound healing. Western blot analysis was to confirm the involvement of signalling cascades in observed effects. RESULTS We have identified the binding of scutellarin with VEGF. The binding accounted for the potentiation role of scutellarin in skin regeneration, as triggered by SGC. The co-treatment of scutellarin and SGC onto scratched keratinocyte cultures was able to enhance the process of wound healing, i.e. scutellarin showed a potentiating effect to SGC. In addition, the potentiation of scutellarin was shown to be mediated by phosphorylation of VEGF receptor 2 (VEGFR2) and mitogen-activated protein kinase (MAPK) signalling. CONCLUSION These findings support the application of scutellarin as an enhancing agent in potentiating the SGC-mediated wound healing.
Collapse
Affiliation(s)
- Maggie Suisui Guo
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Xiong Gao
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Weihui Hu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Xiaoyang Wang
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Tina Tingxia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Karl Wah Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, Shenzhen Research Institute, Shenzhen, 518000, China.,Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| |
Collapse
|
15
|
García-Caballero M, Torres-Vargas JA, Marrero AD, Martínez-Poveda B, Medina MÁ, Quesada AR. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022; 14:pharmaceutics14020256. [PMID: 35213989 PMCID: PMC8875200 DOI: 10.3390/pharmaceutics14020256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
The number of cancer cases worldwide keeps growing unstoppably, despite the undeniable advances achieved by basic research and clinical practice. Urologic tumors, including some as prevalent as prostate, bladder or kidney tumors, are no exceptions to this rule. Moreover, the fact that many of these tumors are detected in early stages lengthens the duration of their treatment, with a significant increase in health care costs. In this scenario, prevention offers the most cost-effective long-term strategy for the global control of these diseases. Although specialized diets are not the only way to decrease the chances to develop cancer, epidemiological evidence support the role of certain plant-derived foods in the prevention of urologic cancer. In many cases, these plants are rich in antiangiogenic phytochemicals, which could be responsible for their protective or angiopreventive properties. Angiogenesis inhibition may contribute to slow down the progression of the tumor at very different stages and, for this reason, angiopreventive strategies could be implemented at different levels of chemoprevention, depending on the targeted population. In this review, epidemiological evidence supporting the role of certain plant-derived foods in urologic cancer prevention are presented, with particular emphasis on their content in bioactive phytochemicals that could be used in the angioprevention of cancer.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - José Antonio Torres-Vargas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Ana Dácil Marrero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
| | - Beatriz Martínez-Poveda
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28019 Madrid, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
| | - Ana R. Quesada
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Malaga, Andalucía Tech, E-29071 Malaga, Spain; (M.G.-C.); (J.A.T.-V.); (A.D.M.); (B.M.-P.); (M.Á.M.)
- IBIMA (Biomedical Research Institute of Malaga), E-29071 Malaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Malaga, Spain
- Correspondence:
| |
Collapse
|
16
|
Fan D, Liu C, Guo Z, Huang K, Peng M, Li N, Luo H, Wang T, Cen Z, Cai W, Gu L, Chen S, Li Z. Resveratrol Promotes Angiogenesis in a FoxO1-Dependent Manner in Hind Limb Ischemia in Mice. Molecules 2021; 26:molecules26247528. [PMID: 34946610 PMCID: PMC8707225 DOI: 10.3390/molecules26247528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022] Open
Abstract
Critical limb ischemia (CLI) is a severe form of peripheral artery diseases (PAD) and seriously endangers the health of people. Therapeutic angiogenesis represents an important treatment strategy for CLI; various methods have been applied to enhance collateral circulation. However, the current development drug therapy to promote angiogenesis is limited. Resveratrol (RSV), a polyphenol compound extracted from plants, has various properties such as anti-oxidative, anti-inflammatory and anti-cancer effects. Whether RSV exerts protective effects on CLI remains elusive. In the current study, we demonstrated that oral intake of RSV significantly improved hind limb ischemia in mice, and increased the expression of phosphorylated Forkhead box class-O1 (FoxO1). RSV treatment in human umbilical vein endothelial cells (HUVECs) could increase the phosphorylation of FoxO1 and its cytoplasmic re-localization to promote angiogenesis. Then we manipulated FoxO1 in HUVECs to further verify that the effect of RSV on angiogenesis is in a FoxO1-dependent manner. Furthermore, we performed metabolomics to screen the metabolic pathways altered upon RSV intervention. We found that the pathways of pyrimidine metabolism, purine metabolism, as well as alanine, aspartate and glutamate metabolism, were highly correlated with the beneficial effects of RSV on the ischemic muscle. This study provides a novel direction for the medical therapy to CLI.
Collapse
Affiliation(s)
- Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (D.F.); (C.L.); (K.H.); (N.L.)
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (D.F.); (C.L.); (K.H.); (N.L.)
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Zeling Guo
- Department of Otolaryngology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (D.F.); (C.L.); (K.H.); (N.L.)
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Meixiu Peng
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Na Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (D.F.); (C.L.); (K.H.); (N.L.)
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Hengli Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (H.L.); (T.W.); (Z.C.)
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (H.L.); (T.W.); (Z.C.)
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Zhipeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (H.L.); (T.W.); (Z.C.)
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568, USA;
| | - Lei Gu
- Max Planck Institute for Heart and Lung Research and Cardiopulmonary Institute (CPI), 61231 Bad Nauheim, Germany;
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (H.L.); (T.W.); (Z.C.)
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- Correspondence: (S.C.); (Z.L.)
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (D.F.); (C.L.); (K.H.); (N.L.)
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
- Correspondence: (S.C.); (Z.L.)
| |
Collapse
|
17
|
A Perspective on Withania somnifera Modulating Antitumor Immunity in Targeting Prostate Cancer. J Immunol Res 2021; 2021:9483433. [PMID: 34485538 PMCID: PMC8413038 DOI: 10.1155/2021/9483433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/07/2021] [Indexed: 01/07/2023] Open
Abstract
Medicinal plants serve as a lead source of bioactive compounds and have been an integral part of day-to-day life in treating various disease conditions since ancient times. Withaferin A (WFA), a bioactive ingredient of Withania somnifera, has been used for health and medicinal purposes for its adaptogenic, anti-inflammatory, and anticancer properties long before the published literature came into existence. Nearly 25% of pharmaceutical drugs are derived from medicinal plants, classified as dietary supplements. The bioactive compounds in these supplements may serve as chemotherapeutic substances competent to inhibit or reverse the process of carcinogenesis. The role of WFA is appreciated to polarize tumor-suppressive Th1-type immune response inducing natural killer cell activity and may provide an opportunity to manipulate the tumor microenvironment at an early stage to inhibit tumor progression. This article signifies the cumulative information about the role of WFA in modulating antitumor immunity and its potential in targeting prostate cancer.
Collapse
|
18
|
Gong G, Zheng Y, Kong X, Wen Z. Anti-angiogenesis Function of Ononin via Suppressing the MEK/Erk Signaling Pathway. JOURNAL OF NATURAL PRODUCTS 2021; 84:1755-1762. [PMID: 34029083 DOI: 10.1021/acs.jnatprod.1c00008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Angiogenesis is a complicated pathological process and plays an important role in modulating tumor development. Flavonoids, sharing the basic functional group with estrogen, have been utilized as chemopreventive agents to inhibit endothelial cell angiogenesis and also suppress tumor cell proliferation. Ononin, also referred to as formononetin-7-O-β-d-glucoside, is one of the bioactive chemicals found within many functional food or plants. The anticancer functions of ononin have been reported both in vitro and in vivo. However, the anti-angiogenetic properties of ononin have not been reported. The possible efficacies of ononin against angiogenesis was verified in cultured endothelial cells. Ononin suppressed vascular endothelial growth factor (VEGF)-induced HUVEC migration, invasion. and tube formation activity after 48 h. The apoptosis rate and specific markers, i.e., Bax/Bc-2 ratio, cleaved caspase 3/9 (Cl-caspase 3/9), and cytochrome c (Cyto c), were enhanced in the ononin-treated group. On the other hand, the protein expressions levels of hypoxia-inducible factor 1α (HIF-1α), mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK), and vascular endothelial growth factor receptor 2 (VEGFR2) were restricted after ononin treatment for 2 days in VEGF-pretreated endothelial cells. In summary, ononin acts as a candidate for angiogenetic-related disease prevention and treatment.
Collapse
Affiliation(s)
- Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China
| | - Yuzhong Zheng
- School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Xiangpeng Kong
- Institute of Pharmaceutical & Food Engineering, Chinese Medicine Master Studio of Wang shimin, Shanxi University of Chinese Medicine, Shanxi 030619, China
| | - Zhen Wen
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
19
|
Resveratrol and endothelial function: A literature review. Pharmacol Res 2021; 170:105725. [PMID: 34119624 DOI: 10.1016/j.phrs.2021.105725] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022]
Abstract
Endothelial dysfunction is a major contributing factor to diseases such as atherosclerosis, diabetes mellitus, obesity, hypertension, acute lung injury, preeclampsia, among others. Resveratrol (RSV) is a naturally occurring bioactive polyphenol found in grapes and red wine. According to experimental studies, RSV modulates several events involved in endothelial dysfunction such as impaired vasorelaxation, eNOS uncoupling, leukocyte adhesion, endothelial senescence, and endothelial mesenchymal transition. The endothelial protective effects of RSV are found to be mediated by numerous molecular targets (e.g. Silent Information Regulator 1 (SIRT1), 5' AMP-activated protein kinase (AMPK), endothelial nitric oxide synthase (eNOS), nuclear factor-erythroid-derived 2-related factor-2 (Nrf2), peroxisome proliferator-activated receptor (PPAR), Krüppel-like factor-2 (KLF2), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB)). Herein, we present an updated review addressing pharmacological effects and molecular targets of RSV in maintaining endothelial function, and the potential of this phytochemical for endothelial dysfunction-associated disorders.
Collapse
|
20
|
Hu WH, Dai DK, Zheng BZY, Duan R, Chan GKL, Dong TTX, Qin QW, Tsim KWK. The binding of kaempferol-3-O-rutinoside to vascular endothelial growth factor potentiates anti-inflammatory efficiencies in lipopolysaccharide-treated mouse macrophage RAW264.7 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153400. [PMID: 33157413 DOI: 10.1016/j.phymed.2020.153400] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/08/2020] [Accepted: 10/25/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Vascular Endothelial Growth Factors (VEGFs) are a group of growth factor in regulating development and maintenance of blood capillary. The VEGF family members include VEGF-A, placenta growth factor (PGF), VEGF-B, VEGF-C and VEGF-D. VEGF receptor activation leads to multiple complex signaling pathways, particularly in inducing angiogenesis. Besides, VEGF is produced by macrophages and T cells, which is playing roles in inflammation. In macrophages, VEGF receptor-3 (VEGFR-3) and its ligand VEGF-C are known to attenuate the release of pro-inflammatory cytokines. METHODS Immunoprecipitation and molecular docking assays showed the binding interaction of kaempferol-3-O-rutinoside and VEGF-C. Western blotting and qRT-PCR methods were applied to explore the potentiating effect of kaempferol-3-O-rutinoside in VEGF-C-mediated expressions of proteins and genes in endothelial cells and LPS-induced macrophages. Enzyme-linked immunosorbent assay (ELISA) was employed to reveal the release of proinflammatory cytokines in LPS-induced macrophages. Immunofluorescence assay was performed to determine the effect of kaempferol-3-O-rutinoside in regulating nuclear translocation of NF-κB p65 subunit in the VEGF-C-treated cultures. In addition, Transwell® motility assay was applied to detect the ability of cell migration after drug treatment in LPS-induced macrophages. RESULTS We identified kaempferol-3-O-rutinoside, a flavonoid commonly found in vegetable and fruit, was able to act on cultured macrophages in inhibiting inflammatory response, and the inhibition was mediated by its specific binding to VEGF-C. The kaempferol-3-O-rutinoside-bound VEGF-C showed high potency to trigger the receptor activation. In LPS-treated cultured macrophages, applied kaempferol-3-O-rutinoside potentiated inhibitory effects of exogenous applied VEGF-C on the secretions of pro-inflammatory cytokines, i.e. IL-6 and TNF-α, as well as expressions of nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). This inhibition was in parallel to transcription and translocation of NF-κB. Moreover, the binding of kaempferol-3-O-rutinoside with VEGF-C suppressed the LPS-induced migration of macrophage. CONCLUSION Taken together, our results suggested the pharmacological roles of kaempferol-3-O-rutinoside in VEGF-C-mediated anti-inflammation.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Diana Kun Dai
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Brody Zhong-Yu Zheng
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Gallant Kar-Lun Chan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Tina Ting-Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qi-Wei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Karl Wah-Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
21
|
Caradonna F, Cruciata I, Luparello C. Nutrigenetics, nutrigenomics and phenotypic outcomes of dietary low-dose alcohol consumption in the suppression and induction of cancer development: evidence from in vitro studies. Crit Rev Food Sci Nutr 2020; 62:2122-2139. [PMID: 33287559 DOI: 10.1080/10408398.2020.1850416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is known that the intake of alcoholic beverages may impair genetic and epigenetic regulatory events with consequent crucial effects on cell phenotypes and that its association with selected genotypes can lead to a different risk of cancer in the population. The aim of this review is to pick up selected studies on this topic and recapitulate some of the biochemical and nutrigenetic/nutrigenomic aspects involved in the impact of dietary low-dose alcohol consumption on the switching-on or -off of tumorigenic pathways. These include i) the existence of predisposing or protective human genotypes and the relationship between dietary compounds and alcohol in the promotion or inhibition of carcinogenesis; ii) the effects of other components of alcoholic drinks in the modulation of the expression of oncogenes and oncosuppressors, the autophagic flux and the onset of apoptosis, with examples of their cytospecificity; and iii) the role of alcoholic beverage consumption within particular dietary regimens, including the Mediterranean diet. Taking all the data into account, several alcohol-associated bioactive dietary compounds appear capable to modulate peculiar intracellular pathways predisposing to or protecting from cancer. Advances in the nutrigenetic, nutrigenomic and nutriepigenetic knowledge complementing the biochemical and molecular approaches will help in unveiling their impact on health outcome.
Collapse
Affiliation(s)
- Fabio Caradonna
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Ilenia Cruciata
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Claudio Luparello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| |
Collapse
|
22
|
Adnan M, Rasul A, Hussain G, Shah MA, Zahoor MK, Anwar H, Sarfraz I, Riaz A, Manzoor M, Adem Ş, Selamoglu Z. Ginkgetin: A natural biflavone with versatile pharmacological activities. Food Chem Toxicol 2020; 145:111642. [PMID: 32783998 DOI: 10.1016/j.fct.2020.111642] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/12/2020] [Accepted: 07/18/2020] [Indexed: 12/13/2022]
Abstract
Natural products, being richly endowed with curative powers, have become spotlight for biomedical and pharmaceutical research to develop novel therapeutics during recent years. Ginkgetin (GK), a natural non-toxic biflavone, has been shown to exhibit anti-cancer, anti-inflammatory, anti-microbial, anti-adipogenic, and neuroprotective activities. GK combats cancer progression by arresting cell cycle, inducing apoptosis, stimulating autophagy, and targeting many deregulated signaling pathways such as JAK/STAT and MAPKs. GKhalts inflammation mediators like interleukins, iNOS, COX-2, PGE2, NF-κB, and acts as an inhibitor of PLA2. GK shows strong neuroprotection against oxidative stress-promoted cell death, inhibits cerebral micro-hemorrhage, decreases neurologic deficits, and halts apoptosis of neurons. GK also acts as anti-fungal, anti-viral, anti-bacterial, leishmanicidal and anti-plasmodial agent. GK shows substantial preventive or therapeutic effects in in vivo models of many diseases including atherosclerosis, cancer, neurodegenerative, hepatic, influenza, and inflammatory diseases. Based on various computational, in vitro and in vivo evidences, this article demonstrates the potential of ginkgetin for development of therapeutics against various diseases. Although GK has been systematically studied from pharmacological point of view, a vast field of pharmacokinetics, pre-clinical and clinical studies is still open for the researchers to fully validate its potential for the treatment of various diseases.
Collapse
Affiliation(s)
- Muhammad Adnan
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan.
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan.
| | - Muhammad Kashif Zahoor
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Maleeha Manzoor
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Şevki Adem
- Department of Chemistry, Faculty of Sciences, Çankırı Karatekin University, Uluyazı Campus Çankırı, Turkey
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde Campus, 51240, Turkey
| |
Collapse
|
23
|
Fonseca-Santos B, Chorilli M. The uses of resveratrol for neurological diseases treatment and insights for nanotechnology based-drug delivery systems. Int J Pharm 2020; 589:119832. [PMID: 32877730 DOI: 10.1016/j.ijpharm.2020.119832] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Neurological disorders have been growing in recent years and are highly prevalent globally. Resveratrol (RES) is a natural product from plant sources such as grape skins. This compound has shown biological activity in many diseases, in particular, those that act on the central nervous system. The mechanism of action and the key points in neurological disorders were described and show the targeted mechanism of action. Due to the insolubility of this compound; the use of nanotechnology-based systems has been proposed for the incorporation of RES and RES-loaded nanocarriers have been designed for intranasal administration, oral or parenteral routes to deliver it to the brain. In general, these nanosystems have shown to be effective in many studies, pharmacological and pharmacokinetic assays, as well as some cell studies. The outcomes show that RES has been reported in human clinical trials for some neurological diseases, although no studies were performed in humans using nanocarriers, animal and/or cellular models have been reported to show good results regarding therapeutics on neurological diseases. Thus, the use of this nutraceutical has shown true for neurological diseases and its loading into nanocarriers displaying good results on the stability, delivery and targeting to the brain.
Collapse
Affiliation(s)
- Bruno Fonseca-Santos
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Department of Drugs and Medicines, Araraquara, São Paulo 14801-903, Brazil
| | - Marlus Chorilli
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Department of Drugs and Medicines, Araraquara, São Paulo 14801-903, Brazil.
| |
Collapse
|
24
|
Hu WH, Dai DK, Zheng BZY, Duan R, Dong TTX, Qin QW, Tsim KWK. Piceatannol, a Natural Analog of Resveratrol, Exerts Anti-angiogenic Efficiencies by Blockage of Vascular Endothelial Growth Factor Binding to Its Receptor. Molecules 2020; 25:molecules25173769. [PMID: 32824997 PMCID: PMC7504081 DOI: 10.3390/molecules25173769] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022] Open
Abstract
Piceatannol is also named as trans-3,4,3′,5′-tetrahydroxy-stilbene, which is a natural analog of resveratrol and a polyphenol existing in red wine, grape and sugar cane. Piceatannol has been proved to possess activities of immunomodulatory, anti-inflammatory, antiproliferative and anticancer. However, the effect of piceatannol on VEGF-mediated angiogenesis is not known. Here, the inhibitory effects of piceatannol on VEGF-induced angiogenesis were tested both in vitro and in vivo models of angiogenesis. In human umbilical vein endothelial cells (HUVECs), piceatannol markedly reduced the VEGF-induced cell proliferation, migration, invasion, as well as tube formation without affecting cell viability. Furthermore, piceatannol significantly inhibited the formation of subintestinal vessel in zebrafish embryos in vivo. In addition, we identified the underlying mechanism of piceatannol in triggering the anti-angiogenic functions. Piceatannol was proposed to bind with VEGF, thus attenuating VEGF in activating VEGF receptor and blocking VEGF-mediated downstream signaling, including expressions of phosphorylated eNOS, Erk and Akt. Furthermore, piceatannol visibly suppressed ROS formation, as triggered by VEGF. Moreover, we further determined the outcome of piceatannol binding to VEGF in cancer cells: piceatannol significantly suppressed VEGF-induced colon cancer proliferation and migration. Thus, these lines of evidence supported the conclusion that piceatannol could down regulate the VEGF-mediated angiogenic functions with no cytotoxicity via decreasing the amount of VEGF binding to its receptors, thus affecting the related downstream signaling. Piceatannol may be developed into therapeutic agents or health products to reduce the high incidence of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (Q.-W.Q.)
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Diana Kun Dai
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Brody Zhong-Yu Zheng
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Tina Ting-Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
| | - Qi-Wei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; (W.-H.H.); (Q.-W.Q.)
| | - Karl Wah-Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (D.K.D.); (B.Z.-Y.Z.); (R.D.); (T.T.-X.D.)
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-2358-7332; Fax: +852-2358-1559
| |
Collapse
|
25
|
Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020; 49:5008-5057. [PMID: 32538379 PMCID: PMC7418030 DOI: 10.1039/c8cs01021h] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis plays a critical role within the human body, from the early stages of life (i.e., embryonic development) to life-threatening diseases (e.g., cancer, heart attack, stroke, wound healing). Many pharmaceutical companies have expended huge efforts on both stimulation and inhibition of angiogenesis. During the last decade, the nanotechnology revolution has made a great impact in medicine, and regulatory approvals are starting to be achieved for nanomedicines to treat a wide range of diseases. Angiogenesis therapies involve the inhibition of angiogenesis in oncology and ophthalmology, and stimulation of angiogenesis in wound healing and tissue engineering. This review aims to summarize nanotechnology-based strategies that have been explored in the broad area of angiogenesis. Lipid-based, carbon-based and polymeric nanoparticles, and a wide range of inorganic and metallic nanoparticles are covered in detail. Theranostic and imaging approaches can be facilitated by nanoparticles. Many preparations have been reported to have a bimodal effect where they stimulate angiogenesis at low dose and inhibit it at higher doses.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, 917794-8564 Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 101 29 Torino, Italy
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Hu WH, Wang HY, Xia YT, Dai DK, Xiong QP, Dong TTX, Duan R, Chan GKL, Qin QW, Tsim KWK. Kaempferol, a Major Flavonoid in Ginkgo Folium, Potentiates Angiogenic Functions in Cultured Endothelial Cells by Binding to Vascular Endothelial Growth Factor. Front Pharmacol 2020; 11:526. [PMID: 32410995 PMCID: PMC7198864 DOI: 10.3389/fphar.2020.00526] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/03/2020] [Indexed: 12/24/2022] Open
Abstract
Kaempferol is a major flavonoid in Ginkgo Folium and other edible plants, which is being proposed here to have roles in angiogenesis. Angiogenesis is important in both physiological and pathological development. Here, kaempferol was shown to bind with vascular endothelial growth factor (VEGF), probably in the heparin binding domain of VEGF: this binding potentiated the angiogenic functions of VEGF in various culture models. Kaempferol potentiated the VEGF-induced cell motility in human umbilical vein endothelial cells (HUVECs), as well as the sub-intestinal vessel sprouting in zebrafish embryos and formation of microvascular in rat aortic ring. In cultured HUVECs, application of kaempferol strongly potentiated the VEGF-induced phosphorylations of VEGFR2, endothelial nitric oxide synthase (eNOS) and extracellular signal-regulated kinase (Erk) in time-dependent and concentration-dependent manners, and in parallel the VEGF-mediated expressions of matrix metalloproteinases (MMPs), MMP-2 and MMP-9, were significantly enhanced. In addition, the potentiation effect of kaempferol was revealed in VEGF-induced migration of skin cell and monocyte. Taken together, our results suggested the pharmacological roles of kaempferol in potentiating VEGF-mediated functions should be considered.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong.,Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Huai-You Wang
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Yi-Teng Xia
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Diana Kun Dai
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Qing-Ping Xiong
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong.,Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Jiangsu, China
| | - Tina Ting-Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Gallant Kar-Lun Chan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Qi-Wei Qin
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Karl Wah-Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, The Hong Kong University of Science and Technology, Nanshan, Shenzhen, China.,Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| |
Collapse
|
27
|
Xiong Q, Li H, Zhou L, Liang J, Zhang Z, Han Y, Jing Y, Hu Y, Shi Y, Xu T, Qian G, Yuan J. A sulfated polysaccharide from the edible flesh of Cipangopaludina chinensis inhibits angiogenesis to enhance atherosclerotic plaque stability. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103800] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
28
|
Hu WH, Chan GKL, Duan R, Wang HY, Kong XP, Dong TTX, Tsim KWK. Synergy of Ginkgetin and Resveratrol in Suppressing VEGF-Induced Angiogenesis: A Therapy in Treating Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11121828. [PMID: 31757048 PMCID: PMC6966653 DOI: 10.3390/cancers11121828] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
Ginkgetin, a biflavone from Ginkgo biloba leaf, and resveratrol, a polyphenol found in grape and wine, are two phytochemicals being identified for its binding to vascular endothelial growth factor (VEGF): the binding, therefore, resulted in the alteration of the physiological roles of VEGF-mediated angiogenesis. The bindings of ginkgetin and resveratrol were proposed on different sites of VEGF, but both of them suppressed the angiogenic properties of VEGF. The suppressive activities of ginkgetin and resveratrol in VEGF-mediated angiogenesis were supported by several lines of evidence including (i) inhibiting the formation of sub-intestinal vessel in zebrafish embryos and microvascular sprouting in rat aortic ring; and (ii) suppressing the phosphorylations of VEGFR2, Akt, eNOS, and Erk as well as expressions of matrix metalloproteinases (MMPs), MMP-2, and MMP-9 in human umbilical vein endothelial cells (HUVECs). Here, we showed the synergy of ginkgetin and resveratrol in suppressing the VEGF-induced endothelial cell proliferation, migration, invasion, and tube formation. The synergy of ginkgetin and resveratrol was further illustrated in HT-29 colon cancer xenograft nude mice. Ginkgetin and resveratrol, when applied together, exerted a synergistic anti-tumor effect of 5-fluorouracil with decreasing microvessel density of tumors. In parallel, the combination of ginkgetin and resveratrol synergistically relieved the 5-fluorouracil-induced inflammatory response by suppressing expressions of COX-2 and inflammatory cytokines. Thus, the anti-angiogenic roles of ginkgetin and/or resveratrol could provide effective therapeutic strategy in cancer, similar to that of Avastin, in suppressing the VEGF-mediated angiogenesis during cancer development.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Gallant Kar-Lun Chan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Huai-You Wang
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Xiang-Peng Kong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Tina Ting-Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Karl Wah-Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-2358-7332; Fax: +852-2358-1552
| |
Collapse
|
29
|
Zambrano A, Molt M, Uribe E, Salas M. Glut 1 in Cancer Cells and the Inhibitory Action of Resveratrol as A Potential Therapeutic Strategy. Int J Mol Sci 2019; 20:ijms20133374. [PMID: 31324056 PMCID: PMC6651361 DOI: 10.3390/ijms20133374] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022] Open
Abstract
An important hallmark in cancer cells is the increase in glucose uptake. GLUT1 is an important target in cancer treatment because cancer cells upregulate GLUT1, a membrane protein that facilitates the basal uptake of glucose in most cell types, to ensure the flux of sugar into metabolic pathways. The dysregulation of GLUT1 is associated with numerous disorders, including cancer and metabolic diseases. There are natural products emerging as a source for inhibitors of glucose uptake, and resveratrol is a molecule of natural origin with many properties that acts as antioxidant and antiproliferative in malignant cells. In the present review, we discuss how GLUT1 is involved in the general scheme of cancer cell metabolism, the mechanism of glucose transport, and the importance of GLUT1 structure to understand the inhibition process. Then, we review the current state-of-the-art of resveratrol and other natural products as GLUT1 inhibitors, focusing on those directed at treating different types of cancer. Targeting GLUT1 activity is a promising strategy for the development of drugs aimed at treating neoplastic growth.
Collapse
Affiliation(s)
- Angara Zambrano
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Concepción, Concepción 4070386, Chile
| | - Matías Molt
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile
| | - Elena Uribe
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Concepción, Concepción 4070386, Chile
| | - Mónica Salas
- Instituto de Bioquimica y Microbiologia, Universidad Austral de Chile, Valdivia 0000000, Chile.
| |
Collapse
|
30
|
Han Y, Jo H, Cho JH, Dhanasekaran DN, Song YS. Resveratrol as a Tumor-Suppressive Nutraceutical Modulating Tumor Microenvironment and Malignant Behaviors of Cancer. Int J Mol Sci 2019; 20:925. [PMID: 30791624 PMCID: PMC6412705 DOI: 10.3390/ijms20040925] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor-suppressive effects of resveratrol have been shown in various types of cancer. However, regulation of tumor microenvironment by resveratrol is still unclear. Recent findings suggest resveratrol can potentiate its tumor-suppressive effect through modulation of the signaling pathways of cellular components (fibroblasts, macrophages and T cells). Also, studies have shown that resveratrol can suppress malignant phenotypes of cancer cells acquired in response to stresses of the tumor microenvironment, such as hypoxia, oxidative stress and inflammation. We discuss the effects of resveratrol on cancer cells in stress environment of tumors as well as interactions between cancer cells and non-cancer cells in this review.
Collapse
Affiliation(s)
- Youngjin Han
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - HyunA Jo
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Jae Hyun Cho
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma city, OK 73012, USA.
| | - Yong Sang Song
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|