1
|
Sanlier NT, Saçinti KG, Türkoğlu İ, Sanlier N. Some Polyphenolic Compounds as Potential Therapeutic Agents in Cervical Cancer: The Most Recent Advances and Future Prospects. Nutr Rev 2025; 83:880-896. [PMID: 39283708 DOI: 10.1093/nutrit/nuae126] [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] [Indexed: 04/12/2025] Open
Abstract
The leading causes of cancer include gradual changes in regulatory proteins, dysregulated cell-signaling pathways, dysfunction of apoptosis, and oxidative stress. Consuming polyphenols from food sources has been proven to have strong connections with ameliorating specific physiological biomarkers along with other elements concerning cancer. Recent studies have focused on polyphenols' molecular mechanisms of action and anticancer and chemopreventive properties and effects in the treatment of different types of cancer. Polyphenols participate in the regulation of numerous cellular mechanisms alongside signaling pathways through their effects on inflammation, cellular proliferation, apoptosis, and partially via epigenetic alterations in cervical cancer. A number of animal models and cell and human studies have indicated the use of polyphenols to be safe and tolerable. Thus, it would be fair to state that, with their advantages vis-à-vis lack of toxicity, cost, and access, and with the positive clinical results, polyphenols have a potential to make a difference in cancer treatment. The present review examined the chemical and physical properties, analogs, metabolites, and mechanisms of physiological activities of various polyphenols and how they may affect the incidence rate and management of cervical cancer. Therefore, this review constitutes a starting point to examine the potential applications for cervical cancer.
Collapse
Affiliation(s)
- Nazlı Tunca Sanlier
- Department of Obstetrics and Gynecology, Turkish Ministry of Health, Ankara City Hospital, Ankara 06800, Turkey
| | - Koray Görkem Saçinti
- Department of Obstetrics and Gynecology, Aksaray University Training and Research Hospital, Aksaray 68200, Turkey
- Division of Epidemiology, Department of Public Health, Hacettepe University Faculty of Medicine, Ankara 06100, Turkey
| | - İnci Türkoğlu
- Department of Nutrition and Dietetics, Hacettepe University School of Health Sciences, Ankara 06100, Turkey
| | - Nevin Sanlier
- Department of Nutrition and Dietetics, Ankara Medipol University School of Health Sciences, Ankara 06050, Turkey
| |
Collapse
|
2
|
Yarlagadda DL, Das S, Anand Vullendula SK, Manandhar S, Dengale SJ, Ranganath Pai KS, Bhat K. Computational-Based Polyphenol Therapy for Nonsmall Cell Lung Cancer: Naringin Coamorphous Systems for Solubility and Bioavailability Enhancement. Mol Pharm 2024; 21:3951-3966. [PMID: 39049477 PMCID: PMC11304383 DOI: 10.1021/acs.molpharmaceut.4c00303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024]
Abstract
In this research, we utilized molecular simulations to create co-amorphous materials (CAMs) of ceritinib (CRT) with the objective of improving its solubility and bioavailability. We identified naringin (NRG) as a suitable co-former for CRT CAMs based on binding energy and intermolecular interactions through computational modeling. We used the solvent evaporation method to produce CAMs of CRT and NRG, expecting to enhance both solubility and bioavailability simultaneously. The solid-state characterization using techniques like differential scanning calorimeter, X-ray powder diffraction, and Fourier-transform infrared spectroscopy affirmed the formation of a single amorphous phase and the presence of intermolecular interactions between CRT and NRG in the CAMs. These materials remained physically stable for up to six months under dry conditions at 40 °C. Moreover, the CAMs demonstrated significant improvements in the solubility and dissolution of CRT (specifically in the ratio CRT:NRG 1:2). This, in turn, led to an increase in cytotoxicity, apoptotic cells, and G0/G1 phase inhibition in A549 cells compared to CRT alone. Furthermore, CRT permeability is also improved twofold, as estimated by the everted gut sac method. The enhanced solubility of CAMs also positively affected the pharmacokinetic parameters. When compared to the physical mixture, the CAMs of CRT:NRG 2:1 exhibited a 2.1-fold increase in CRT exposure (AUC0-t) and a 2.4-fold increase in plasma concentration (Cmax).
Collapse
Affiliation(s)
- Dani Lakshman Yarlagadda
- Department
of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical
Sciences, Manipal Academy of Higher Education
(MAHE), Manipal, Karnataka 576104, India
| | - Subham Das
- Department
of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Sai Krishna Anand Vullendula
- Department
of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical
Sciences, Manipal Academy of Higher Education
(MAHE), Manipal, Karnataka 576104, India
| | - Suman Manandhar
- Department
of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Swapnil J. Dengale
- Department
of Pharmaceutical Analysis, National Institute
of Pharmaceutical Education and Research (NIPER), Guwahati, Changsari 781101, India
| | - K. Sreedhara Ranganath Pai
- Department
of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Krishnamurthy Bhat
- Department
of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical
Sciences, Manipal Academy of Higher Education
(MAHE), Manipal, Karnataka 576104, India
| |
Collapse
|
3
|
Zhou X, Wang H, Huang M, Chen J, Chen J, Cheng H, Ye X, Wang W, Liu D. Role of bitter contributors and bitter taste receptors: a comprehensive review of their sources, functions and future development. FOOD SCIENCE AND HUMAN WELLNESS 2024; 13:1806-1824. [DOI: 10.26599/fshw.2022.9250151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Effat H, Abosharaf HA, Radwan AM. Combined effects of naringin and doxorubicin on the JAK/STAT signaling pathway reduce the development and spread of breast cancer cells. Sci Rep 2024; 14:2824. [PMID: 38310190 PMCID: PMC10838285 DOI: 10.1038/s41598-024-53320-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
Breast cancer therapy options are limited due to its late diagnosis and poor prognosis. Doxorubicin is the fundamental therapy approach for this disease. Because chemotherapy has numerous adverse effects, the scope of the existing research was to appraise the synergetic effect of doxorubicin and naringin and explore the underlying mechanism. The cytotoxicity of doxorubicin and naringin on MCF-7 was monitored. Furthermore, the expression of STAT3 and JAK1 as well as the apoptotic and metastatic related genes (Bax, Bcl-2, Survivin, and VEGF) were conducted by immunoblotting assay and qRT-PCR. In addition, a wound healing test was utilized to appraise the migration and metastasis of MCF-7. Our results revealed that naringin and doxorubicin had a synergetic inhibitory influence on MCF-7 cells growth and migration. The synergetic action of doxorubicin and naringin effectively hindered the expression of STAT3, JAK1, Bcl-2, Survivin, and VEGF, with a boost in the level of Bax compared to cells treated with either doxorubicin or naringin. In conclusion, our findings imply that combining doxorubicin with naringin may be a favorable strategy for inhibiting the growth of breast cancer.
Collapse
Affiliation(s)
- Heba Effat
- Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institut, Cairo University, Cairo, Egypt.
| | - Hamed A Abosharaf
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Aliaa M Radwan
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
5
|
Yin Z, Hua X, Lu M. Integrated Network Pharmacology and Metabolomics to Dissect the Mechanisms of Naringin for Treating Cervical Cancer. Comb Chem High Throughput Screen 2024; 27:754-764. [PMID: 37143280 DOI: 10.2174/1386207326666230504124030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023]
Abstract
INTRODUCTION Cervical cancer is one of the malignant cancers with high mortality among women worldwide. Although vaccines and early detection have reduced cervical cancer mortality, it remains a malignancy with a high mortality rate in women. OBJECTIVES We aimed to develop a novel integrated strategy that combines metabolomics with network pharmacology to explore the therapeutic mechanisms of naringin in cervical cancer. The mechanism of naringin intervention in cervical cancer was initially clarified by metabolomics and network pharmacology. METHODS The method of LC-MS and network pharmacology for the detection and identification of potential biomarkers and the mechanisms of action of naringin was used. The metabolites were detected and identified based on ultra-high-performance liquid chromatography coupled with Quadrupole- Exactive Orbitrap MS (UHPLC-Q-Exactive Orbitrap MS) and followed by the network pharmacology analysis. RESULTS In network pharmacology, naringin played a synergetic role through regulatory shared pathways, such as steroid hormone biosynthesis, sphingolipid signaling pathway and arachidonic acid metabolism, etc. Besides, the metabolomics analysis showed that 20 differential metabolites and 10 metabolic pathways were mainly involved in the therapeutic effect of naringin on cervical cancer. The result showed that naringin treatment for cervical cancer mainly occurs through the following metabolic pathways: amino acid metabolism and arachidonic acid metabolism. CONCLUSION This work provided valuable information and a scientific basis for further studies of naringin in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Ziwei Yin
- Department of HBP Surgery II, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xuefeng Hua
- Department of HBP Surgery II, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Minqiang Lu
- Department of HBP Surgery II, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
6
|
Nishida T, Horita C, Imagawa M, Hibarino M, Tateno S, Kubo Y, Kawabe M, Morishita N, Endo S, Shiozaki K. Glucosyl hesperidin exhibits more potent anxiolytic activity than hesperidin accompanied by the attenuation of noradrenaline induction in a zebrafish model. Front Pharmacol 2023; 14:1213252. [PMID: 37663268 PMCID: PMC10470464 DOI: 10.3389/fphar.2023.1213252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Anxiety is a symptom of various mental disorders, including depression. Severe anxiety can significantly affect the quality of life. Hesperidin (Hes), a flavonoid found in the peel of citrus fruits, reportedly has various functional properties, one of which is its ability to relieve acute and chronic stress. However, Hes is insoluble in water, resulting in a low absorption rate in the body and low bioavailability. Glucosyl hesperidin (GHes) is produced by adding one glucose molecule to hesperidin. Its water solubility is significantly higher than that of Hes, which is expected to improve its absorption into the body and enhance its effects. However, its efficacy in alleviating anxiety has not yet been investigated. Therefore, in this study, the anxiolytic effects of GHes were examined in a zebrafish model of anxiety. Long-term administration of diets supplemented with GHes did not cause any toxicity in the zebrafish. In the novel tank test, zebrafish in the control condition exhibited an anxious behavior called freezing, which was significantly suppressed in GHes-fed zebrafish. In the black-white preference test, which also induces visual stress, GHes-fed zebrafish showed significantly increased swimming time in the white side area. Furthermore, in tactile (low water-level stress) and olfactory-mediated stress (alarm substance administration test) tests, GHes suppressed anxious behavior, and these effects were stronger than those of Hes. Increased noradrenaline levels in the brain generally cause freezing; however, in zebrafish treated with GHes, the amount of noradrenaline after stress was lower than that in the control group. Activation of c-fos/ERK/Th, which is upstream of the noradrenaline synthesis pathway, was also suppressed, while activation of the CREB/BDNF system, which is vital for neuroprotective effects, was significantly increased. These results indicate that GHes has a more potent anxiolytic effect than Hes in vivo, which may have potential applications in drug discovery and functional food development.
Collapse
Affiliation(s)
- Takumi Nishida
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Chihoko Horita
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Mikiya Imagawa
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Momoka Hibarino
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Sayaka Tateno
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Yurina Kubo
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Momoko Kawabe
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | | | - Shin Endo
- R&D Division, Hayashibara Co., Ltd., Okayama, Japan
| | - Kazuhiro Shiozaki
- Department of Food Life Sciences, Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
7
|
Quirino MWL, Albuquerque APB, De Souza MFD, Da Silva Filho AF, Martins MR, Da Rocha Pitta MG, Pereira MC, De Melo Rêgo MJB. alpha2,3 sialic acid processing enzymes expression in gastric cancer tissues reveals that ST3Gal3 but not Neu3 are associated with Lauren's classification, angiolymphatic invasion and histological grade. Eur J Histochem 2022; 66. [PMID: 36172711 PMCID: PMC9577379 DOI: 10.4081/ejh.2022.3330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 08/27/2022] [Indexed: 11/22/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide. Despite progress in the last decades, there are still no reliable biomarkers for the diagnosis of and prognosis for GC. Aberrant sialylation is a widespread critical event in the development of GC. Neuraminidases (Neu) and sialyltransferases (STs) regulate the ablation and addition of sialic acid during glycoconjugates biosynthesis, and they are a considerable source of biomarkers in various cancers. This study retrospectively characterized Neu3 and ST3Gal3 expression by immunohistochemistry in 71 paraffin-embedded GC tissue specimens and analyzed the relationship between their expression and the clinicopathological parameters. Neu3 expression was markedly increased in GC tissues compared with non-tumoral tissues (p<0.0001). Intratumoral ST3Gal3 staining was significantly associated with intestinal subtype (p=0.0042) and was negatively associated with angiolymphatic invasion (p=0.0002) and higher histological grade G3 (p=0.0066). Multivariate analysis revealed that ST3Gal3 positivity is able to predict Lauren's classification. No associations were found between Neu3 staining and clinical parameters. The in silico analysis of mRNA expression in GC validation cohorts corroborates the significant ST3Gal3 association with higher histological grade observed in our study. These findings suggest that ST3Gal3 expression may be an indicator for aggressiveness of primary GC.
Collapse
Affiliation(s)
- Michael W L Quirino
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | - Amanda P B Albuquerque
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | - Maria F D De Souza
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | - Antônio F Da Silva Filho
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | | | - Maira G Da Rocha Pitta
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | - Michelly C Pereira
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| | - Moacyr J B De Melo Rêgo
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for -Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE.
| |
Collapse
|
8
|
Song L, Xiong P, Zhang W, Hu H, Tang S, Jia B, Huang W. Mechanism of Citri Reticulatae Pericarpium as an Anticancer Agent from the Perspective of Flavonoids: A Review. Molecules 2022; 27:molecules27175622. [PMID: 36080397 PMCID: PMC9458152 DOI: 10.3390/molecules27175622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 12/24/2022] Open
Abstract
Citri Reticulatae Pericarpium (CRP), also known as “chenpi”, is the most common qi-regulating drug in traditional Chinese medicine. It is often used to treat cough and indigestion, but in recent years, it has been found to have multi-faceted anti-cancer effects. This article reviews the pharmacology of CRP and the mechanism of the action of flavonoids, the key components of CRP, against cancers including breast cancer, lung cancer, prostate cancer, hepatic carcinoma, gastric cancer, colorectal cancer, esophageal cancer, cervical cancer, bladder cancer and other cancers with a high diagnosis rate. Finally, the specific roles of CRP in important phenotypes such as cell proliferation, apoptosis, autophagy and migration–invasion in cancer were analyzed, and the possible prospects and deficiencies of CRP as an anticancer agent were evaluated.
Collapse
Affiliation(s)
- Li Song
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Peiyu Xiong
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Wei Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Hengchang Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Songqi Tang
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou 571199, China
| | - Bo Jia
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Wei Huang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
- Correspondence:
| |
Collapse
|
9
|
Molecular Recognition of Citroflavonoids Naringin and Naringenin at the Active Site of the HMG-CoA Reductase and DNA Topoisomerase Type II Enzymes of Candida spp. and Ustilago maydis. Indian J Microbiol 2022; 62:79-87. [PMID: 35068607 PMCID: PMC8758890 DOI: 10.1007/s12088-021-00980-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
Two agents from natural sources, citroflavonoids naringin and naringenin, can target enzymes in pathogenic yeasts responsible for hospital infections and crop failure. The aim of this study was to examine the molecular recognition site for naringin and naringenin on the HMGR and TOPOII enzymes of eleven Candida species and one phytopathogen, U. maydis, and evaluate yeast susceptibility to these flavonoids. The HMGR and TOPOII enzymes were analyzed in silico. The alignment of the two enzymes in the twelve pathogenic organisms with the corresponding enzyme of Homo sapiens revealed highly conserved amino acid sequences. Modeling studies of the enzymes indicated highly conserved structures. According to molecular docking simulations, both citroflavonoids recognized the amino acid residues of the active site of the enzymes. Binding energy values were higher for naringin (-10.75 and -9.38 kcal/mol, respectively) than simvastatin on HMGR (-9.9) and curcumin on TOPOII (-8.37). The appraisal of twenty-nine virtual mutations provided evidence of probable mechanisms of resistance (high binding energy) or susceptibility (low energy) to the drugs and emphasized the role of key residues. An in vitro susceptibility evaluation of the twelve yeasts demonstrated that the two flavonoids have similar or better MIC values than those reported for the reference compounds, obtaining the lowest with Candida dubliniensis (2.5 µg/ml) and U. maydis (5 µg/ml). Based on the present findings, naringin and naringenin could possibly be effective for treating diseases caused by pathogenic yeasts of the Candida species and U. maydis, presumably by inhibition of their HMGR and TOPOII enzymes. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s12088-021-00980-0.
Collapse
|
10
|
Antioxidant and Anticancer Activities and Protein Interaction of the Oxidovanadium(IV) Naringin Complex. INORGANICS 2022. [DOI: 10.3390/inorganics10010013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The complex of oxidovanadium(IV) with naringin (Narg) [VO(Narg)2] 8H2O (VONarg) was prepared according to the literature improving the synthetic procedure and physicochemical characterization. In addition, biological activities (cytotoxic, antioxidant, and BSA interaction) were determined. The metal coordinated through the 5-hydroxy and 4-carbonyl groups of rings A and C of naringin, respectively. The antioxidant activity of VONarg, determined in vitro, was higher than those of the flavonoid against superoxide and peroxyl reactive oxygen species (ROS) and DPPH radical. The cytotoxic properties were determined by a MTT assay on adenocarcinoma human alveolar basal epithelial cells (A549). VONarg exerted a 20% decrease in cancer cells viability at 24 h incubation, while naringin and oxidovanadium(IV) cation did not show cytotoxicity. Measurements with the normal HEK293 cell line showed that the inhibitory action of the complex is selective. VONarg generated intracellular reactive oxygen species (ROS), depletion of reduced glutathione and depolarization of mitochondrial membrane potential, typical for apoptotic pathway, producing cell death by oxidative stress mechanism. Moreover, naringin interacted with bovine serum albumin (BSA) through hydrophobic interactions in a spontaneous process, and VONarg showed greater affinity for the protein but can still be transported and delivered by it (Ka 104 L·mol−1 order).
Collapse
|
11
|
Plant-Derived Chinese Medicine Monomers on Ovarian Cancer via the Wnt/ β-Catenin Signaling Pathway: Review of Mechanisms and Prospects. JOURNAL OF ONCOLOGY 2021; 2021:6852867. [PMID: 34912456 PMCID: PMC8668291 DOI: 10.1155/2021/6852867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/03/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023]
Abstract
Ovarian cancer (OC) is a common malignant tumor of the female reproductive system and has a high morbidity and mortality rate. The progression and metastasis of OC are complex and involve multiple signaling pathways. The Wnt/β-catenin signaling pathway is closely related to OC, and therefore blocking the activation of the Wnt/β-catenin signaling directly or inhibiting related genes, and molecular targets is of great value in treating OC. Toxicities such as myelotoxicity, cardiotoxicity, genotoxicity, and vasospasm are the major side effects for common anticancer drugs and are well documented. There is, therefore, a need to develop new, effective, safer, and more affordable anticancer drugs from alternative sources. In recent years, plant-derived Chinese medicine monomers have drawn increasing attention due to their high safety, low toxicity, minimal side effects, and antitumor effects. Plant-derived Chinese medicine monomers are effective against multiple targets and can regulate the growth, proliferation, apoptosis, invasion, and migration of OC as well as reverse drug resistance by regulating the Wnt/β-catenin signaling pathway. In this review, we summarize and provide mechanisms and prospects for the use of plant-derived Chinese medicines for the prevention and treatment of OC.
Collapse
|
12
|
Badawy AA, Othman RQA, El-Magd MA. Effect of combined therapy with camel milk-derived exosomes, tamoxifen, and hesperidin on breast cancer. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00163-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Hosseinzadeh E, Hassanzadeh A, Marofi F, Alivand MR, Solali S. Flavonoid-Based Cancer Therapy: An Updated Review. Anticancer Agents Med Chem 2021; 20:1398-1414. [PMID: 32324520 DOI: 10.2174/1871520620666200423071759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/27/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022]
Abstract
As cancers are one of the most important causes of human morbidity and mortality worldwide, researchers try to discover novel compounds and therapeutic approaches to decrease survival of cancer cells, angiogenesis, proliferation and metastasis. In the last decade, use of special phytochemical compounds and flavonoids was reported to be an interesting and hopeful tactic in the field of cancer therapy. Flavonoids are natural polyphenols found in plant, fruits, vegetables, teas and medicinal herbs. Based on reports, over 10,000 flavonoids have been detected and categorized into several subclasses, including flavonols, anthocyanins, flavanones, flavones, isoflavones and chalcones. It seems that the anticancer effect of flavonoids is mainly due to their antioxidant and anti inflammatory activities and their potential to modulate molecular targets and signaling pathways involved in cell survival, proliferation, differentiation, migration, angiogenesis and hormone activities. The main aim of this review is to evaluate the relationship between flavonoids consumption and cancer risk, and discuss the anti-cancer effects of these natural compounds in human cancer cells. Hence, we tried to collect and revise important recent in vivo and in vitro researches about the most effective flavonoids and their main mechanisms of action in various types of cancer cells.
Collapse
Affiliation(s)
- Elham Hosseinzadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Ghanbari-Movahed M, Jackson G, Farzaei MH, Bishayee A. A Systematic Review of the Preventive and Therapeutic Effects of Naringin Against Human Malignancies. Front Pharmacol 2021; 12:639840. [PMID: 33854437 PMCID: PMC8039459 DOI: 10.3389/fphar.2021.639840] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Natural product-based cancer preventive and therapeutic entities, such as flavonoids and their derivatives, are shown to have a noticeable capability to suppress tumor formation and cancer cell growth. Naringin, a natural flavanone glycoside present in various plant species, has been indicated to modulate different signaling pathways and interact with numerous cell signaling molecules, which allows for an extensive variety of pharmacological actions, such as amelioration of inflammation, oxidative stress, metabolic syndromes, bone disorders, and cancer. The purpose of this systematic review is to present a critical and comprehensive assessment of the antitumor ability of naringin and associated molecular targets in various cancers. Methods: Studies were identified through systematic searches of Science Direct, PubMed, and Scopus as well as eligibility checks according to predefined selection criteria. Results: Eighty-seven studies were included in this systematic review. There was strong evidence for the association between treatment with naringin alone, or combined with other drugs and antitumor activity. Additionally, studies showed that naringin-metal complexes have greater anticancer effects compared to free naringin. It has been demonstrated that naringin employs multitargeted mechanisms to hamper cancer initiation, promotion, and progression through modulation of several dysregulated signaling cascades implicated in cell proliferation, autophagy, apoptosis, inflammation, angiogenesis, metastasis, and invasion. Conclusion: The results of our work show that naringin is a promising candidate for cancer prevention and treatment, and might offer substantial support for the clinical application of this phytocompound in the future. Nevertheless, further preclinical and clinical studies as well as drug delivery approaches are needed for designing novel formulations of naringin to realize the full potential of this flavonoid in cancer prevention and intervention.
Collapse
Affiliation(s)
- Maryam Ghanbari-Movahed
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Gloria Jackson
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
15
|
Zhang X, Shan S, Shi J, Li H, Li Z. Polyphenol from millet bran increases the sensitivity of colorectal cancer cells to oxaliplatin by blocking the ganglioside GM3 catabolism. Food Funct 2021; 12:291-301. [DOI: 10.1039/d0fo02232b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The study implies that bound polyphenol from millet bran dramatically prevents ganglioside GM3 catabolism followed by the suppression of P-gp, which eventually reverse drug-resistance in colorectal cancer cells to oxaliplatin.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Institute of Biotechnology
- Shanxi University
- Taiyuan
- China
| | - Shuhua Shan
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Institute of Biotechnology
- Shanxi University
- Taiyuan
- China
| | - Jiangying Shi
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Institute of Biotechnology
- Shanxi University
- Taiyuan
- China
| | - Hanqing Li
- School of Life Science
- Shanxi University
- Taiyuan
- China
| | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Institute of Biotechnology
- Shanxi University
- Taiyuan
- China
| |
Collapse
|
16
|
Kong F, Ding Z, Zhang K, Duan W, Qin Y, Su Z, Bi Y. Optimization of extraction flavonoids from Exocarpium Citri Grandis and evaluation its hypoglycemic and hypolipidemic activities. JOURNAL OF ETHNOPHARMACOLOGY 2020; 262:113178. [PMID: 32736047 DOI: 10.1016/j.jep.2020.113178] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/13/2020] [Accepted: 07/08/2020] [Indexed: 05/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Exocarpium Citri Grandis (ECG; Huajuhong in Chinese) is a precious traditional Chinese medicine with a history of hundreds of years in China. It has been demonstrated to possess numerous pharmacological properties, including antitussive, expectorant, anti-inflammatory, hypoglycemic, and hypolipidemic. However, no in-depth report exists on the hypoglycemic and hypolipidemic properties of ECG. AIM OF THE STUDY This study aimed to evaluate the hypoglycemic and hypolipidemic properties of ECG flavonoids extract in vitro and in vivo so as to lay the foundation for further researches in this field. MATERIALS AND METHODS Total flavonoids (TF) and naringin were separately extracted from ECG, and the components of TF were identified by HPLC-MS. The antioxidant capacities of TF and naringin were determined by 2,2,1-diphenyl-1-picrylhydrazyl (DPPH)-free radical scavenging tests, and digestive enzymes activity inhibition assays in vitro in order to evaluate their hypoglycemic properties. Furthermore, diabetic mice experiments were performed to assess the hypoglycemic and hypolipidemic properties of TF and naringin in vivo. RESULTS Five compounds were identified from TF, including naringin, rhoifolin, poncirin, bergaptol, and naringenin. The half maximal inhibitory concentration (IC50) of TF and naringin to DPPH-free radicals were 0.269 and 1.946 mg/mL, respectively. TF and naringin demonstrated a certain inhibitory effect on α-glucosidase and a weaker inhibitory effect on α-amylase. The results of animal experiments showed that TF and naringin had no significant effect on the blood glucose levels, but they could lead to significant (p < 0.05 or p < 0.01) increase in the serum insulin level and high-density lipoprotein cholesterol (HDL-C) levels with concomitant reduction in the total cholesterol (TC), total triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C) levels in diabetic mice. In addition, TF and naringin could reduce the liver index of diabetic mice (p < 0.01) and reduce the kidney index at low doses (p < 0.05). CONCLUSIONS Our study revealed that the hypoglycemic and hypolipidemic activities of TF is mainly derived from naringin, and other active ingredients in TF also have the effects of alleviating oxidative stress, inhibiting digestive enzyme activity and reducing blood lipids. Our results thus provide a scientific basis for the application of ECG in antidiabetic treatment.
Collapse
Affiliation(s)
- Fansheng Kong
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhendong Ding
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Kai Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Weijie Duan
- Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, 650021, China
| | - Yaru Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhipeng Su
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yongguang Bi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| |
Collapse
|
17
|
Memariani Z, Abbas SQ, Ul Hassan SS, Ahmadi A, Chabra A. Naringin and naringenin as anticancer agents and adjuvants in cancer combination therapy: Efficacy and molecular mechanisms of action, a comprehensive narrative review. Pharmacol Res 2020; 171:105264. [PMID: 33166734 DOI: 10.1016/j.phrs.2020.105264] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/10/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Although the rates of many cancers are controlled in Western countries, those of some cancers, such as lung, breast, and colorectal cancer are currently increasing in many low- and middle-income countries due to increases in risk factors caused by development and societal problems. Additionally, endogenous factors, such as inherited mutations, steroid hormones, insulin, and insulin-like growth factor systems, inflammation, oxidative stress, and exogenous factors (including tobacco, alcohol, infectious agents, and radiation), are believed to compromise cell functions and lead to carcinogenesis. Chemotherapy, surgery, radiation therapy, hormone therapy, and targeted therapies are some examples of the approaches used for cancer treatment. However, various short- and long-term side effects can also considerably impact patient prognosis based on clinical factors associated with treatments. Recently, increasing numbers of studies have been conducted to identify novel therapeutic agents from natural products, among which plant-derived bioactive compounds have been increasingly studied. Naringin (NG) and its aglycone naringenin (NGE) are abundantly present in citrus fruits, such as grapefruits and oranges. Their anti-carcinogenic activities have been shown to be exerted through several cell signal transduction pathways. Recently, different pharmacological strategies based on combination therapy, involving NG and NGE with the current anti-cancer agents have shown prodigious synergistic effects when compared to monotherapy. Besides, NG and NGE have been reported to overcome multidrug resistance, resulting from different defensive mechanisms in cancer, which is one of the major obstacles of clinical treatment. Thus, we comprehensively reviewed the inhibitory effects of NG and NGE on several types of cancers through different signal transduction pathways, the roles on sensitizing with the current anticancer medicines, and the efficacy of the cancer combination therapy.
Collapse
Affiliation(s)
- Zahra Memariani
- Traditional Medicine and History of Medical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Syed Qamar Abbas
- Department of Pharmacy, Sarhad University of Science and Technology, Peshawar, Pakistan.
| | - Syed Shams Ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Aroona Chabra
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
18
|
Lee N, Spears ME, Carlisle AE, Kim D. Endogenous toxic metabolites and implications in cancer therapy. Oncogene 2020; 39:5709-5720. [PMID: 32709924 PMCID: PMC7452860 DOI: 10.1038/s41388-020-01395-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/16/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022]
Abstract
It is well recognized that many metabolic enzymes play essential roles in cancer cells in producing building blocks such as nucleotides, which are required in greater amounts due to their increased proliferation. On the other hand, the significance of enzymes in preventing the accumulation of their substrates is less recognized. Here, we outline the evidence and underlying mechanisms for how many metabolites normally produced in cells are highly toxic, such as metabolites containing reactive groups (e.g., methylglyoxal, 4-hydroxynonenal, and glutaconyl-CoA), or metabolites that act as competitive analogs against other metabolites (e.g., deoxyuridine triphosphate and l-2-hydroxyglutarate). Thus, if a metabolic pathway contains a toxic intermediate, then we may be able to induce accumulation and poison a cancer cell by targeting the downstream enzyme. Furthermore, this poisoning may be cancer cell selective if this pathway is overactive in a cancer cell relative to a nontransformed cell. We describe this concept as illustrated in selenocysteine metabolism and other pathways and discuss future directions in exploiting toxic metabolites to kill cancer cells.
Collapse
Affiliation(s)
- Namgyu Lee
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Meghan E Spears
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Anne E Carlisle
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
19
|
Wang D, Fu Z, Xing Y, Tan Y, Han L, Yu H, Wang T. Rapid identification of chemical composition and metabolites of Pingxiao Capsule
in vivo
using molecular networking and untargeted data‐dependent tandem mass spectrometry. Biomed Chromatogr 2020; 34:e4882. [DOI: 10.1002/bmc.4882] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/26/2020] [Accepted: 05/08/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Dong Wang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
- National Clinical Research Center of CancerTianjin Medical University Cancer Institute and Hospital Tianjin People's Republic of China
| | - Zhifei Fu
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Yanchao Xing
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Yao Tan
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Lifeng Han
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Haiyang Yu
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| |
Collapse
|
20
|
Honda A, Chigwechokha PK, Takase R, Hayasaka O, Fujimura K, Kotani T, Komatsu M, Shiozaki K. Novel Nile tilapia Neu1 sialidases: Molecular cloning and biochemical characterization of the sialidases Neu1a and Neu1b. Gene 2020; 742:144538. [DOI: 10.1016/j.gene.2020.144538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/06/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
|
21
|
Koolaji N, Shammugasamy B, Schindeler A, Dong Q, Dehghani F, Valtchev P. Citrus Peel Flavonoids as Potential Cancer Prevention Agents. Curr Dev Nutr 2020; 4:nzaa025. [PMID: 32391511 PMCID: PMC7199889 DOI: 10.1093/cdn/nzaa025] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/11/2019] [Accepted: 02/24/2020] [Indexed: 12/18/2022] Open
Abstract
Citrus fruit and in particular flavonoid compounds from citrus peel have been identified as agents with utility in the treatment of cancer. This review provides a background and overview regarding the compounds found within citrus peel with putative anticancer potential as well as the associated in vitro and in vivo studies. Historical studies have identified a number of cellular processes that can be modulated by citrus peel flavonoids including cell proliferation, cell cycle regulation, apoptosis, metastasis, and angiogenesis. More recently, molecular studies have started to elucidate the underlying cell signaling pathways that are responsible for the flavonoids' mechanism of action. These growing data support further research into the chemopreventative potential of citrus peel extracts, and purified flavonoids in particular. This critical review highlights new research in the field and synthesizes the pathways modulated by flavonoids and other polyphenolic compounds into a generalized schema.
Collapse
Affiliation(s)
- Nooshin Koolaji
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
- Center for Advanced Food Enginomics, University of Sydney, Sydney, Australia
| | - Balakrishnan Shammugasamy
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
- Center for Advanced Food Enginomics, University of Sydney, Sydney, Australia
| | - Aaron Schindeler
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
- Center for Advanced Food Enginomics, University of Sydney, Sydney, Australia
- Bioengineering & Molecular Medicine, The Children's Hospital at Westmead, Sydney, Australia
| | - Qihan Dong
- School of Science and Health, Western Sydney University, Sydney, Australia
- Greg Brown Laboratory, Central Clinical School and Charles Perkins Centre, University of Sydney, Sydney, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
- Center for Advanced Food Enginomics, University of Sydney, Sydney, Australia
| | - Peter Valtchev
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
- Center for Advanced Food Enginomics, University of Sydney, Sydney, Australia
| |
Collapse
|
22
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020. [DOI: 10.3389/fphar.2020.00451
expr 967555229 + 995954239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
|
23
|
Cao S, Han Y, Li Q, Chen Y, Zhu D, Su Z, Guo H. Mapping Pharmacological Network of Multi-Targeting Litchi Ingredients in Cancer Therapeutics. Front Pharmacol 2020; 11:451. [PMID: 32390834 PMCID: PMC7193898 DOI: 10.3389/fphar.2020.00451] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Considerable pharmacological studies have demonstrated that the extracts and ingredients from different parts (seeds, peels, pulps, and flowers) of Litchi exhibited anticancer effects by affecting the proliferation, apoptosis, autophagy, metastasis, chemotherapy and radiotherapy sensitivity, stemness, metabolism, angiogenesis, and immunity via multiple targeting. However, there is no systematical analysis on the interaction network of “multiple ingredients-multiple targets-multiple pathways” anticancer effects of Litchi. In this study, we summarized the confirmed anticancer ingredients and molecular targets of Litchi based on published articles and applied network pharmacology approach to explore the complex mechanisms underlying these effects from a perspective of system biology. The top ingredients, top targets, and top pathways of each anticancer function were identified using network pharmacology approach. Further intersecting analyses showed that Epigallocatechin gallate (EGCG), Gallic acid, Kaempferol, Luteolin, and Betulinic acid were the top ingredients which might be the key ingredients exerting anticancer function of Litchi, while BAX, BCL2, CASP3, and AKT1 were the top targets which might be the main targets underling the anticancer mechanisms of these top ingredients. These results provided references for further understanding and exploration of Litchi as therapeutics in cancer as well as the application of “Component Formula” based on Litchi’s effective ingredients.
Collapse
Affiliation(s)
- Sisi Cao
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yaoyao Han
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Qiaofeng Li
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China.,School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Yanjiang Chen
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
| | - Dan Zhu
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Hongwei Guo
- College of Pharmacy, Guangxi Medical University, Nanning, China.,Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
Cheng H, Jiang X, Zhang Q, Ma J, Cheng R, Yong H, Shi H, Zhou X, Ge L, Gao G. Naringin inhibits colorectal cancer cell growth by repressing the PI3K/AKT/mTOR signaling pathway. Exp Ther Med 2020; 19:3798-3804. [PMID: 32346444 PMCID: PMC7185071 DOI: 10.3892/etm.2020.8649] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, the incidence of colorectal cancer (CRC) has increased and research into new treatment methods for CRC has become a hot topic. Naringin has an inhibitory effect on the PI3k/AKT/mTOR signaling pathway in various tumor cell types and the effect of naringin is closely related to the occurrence and proliferation of tumor cells. The aim of this present study was to investigate whether naringin could inhibit the proliferation of CRC cells by inhibiting the PI3K/AKT/mTOR signaling pathway. This could provide a more mechanism-based treatment for CRC. MTT assays were used to detect the proliferation of CRC cells treated with various concentrations of naringin. The degree of apoptosis and the expression of apoptosis-related proteins (Bcl-2 and Bax) in CRC cells stimulated by naringin was detected using flow cytometry and western blot assays, respectively. The expression levels of PI3K/AKT/mTOR-related proteins [PI3K, AKT, mTOR, phosphorylated (p)-PI3K, p-AKT and p-mTOR] after naringin stimulation in CRC cells were detected using western blot assays. Naringin inhibited the proliferation of CRC cells in a dose-dependent manner. Naringin promoted the apoptosis of CRC cells and inhibited the activation of the PI3K/AKT/mTOR signaling pathway in a dose-dependent manner. The results demonstrated that naringin may be a promising therapeutic agent for the treatment of CRC, which may inhibit the proliferation of CRC cells and induce apoptosis by inhibiting the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hongyun Cheng
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Xue Jiang
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Qian Zhang
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Jun Ma
- Department of Oncology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Ronghui Cheng
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Hongmei Yong
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Huichang Shi
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Xueyi Zhou
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Liyue Ge
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Guangyi Gao
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| |
Collapse
|
25
|
Synergetic effects of whey protein isolate and naringin on physical and oxidative stability of oil-in-water emulsions. Food Hydrocoll 2020. [DOI: 10.1016/j.foodhyd.2019.105517] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
26
|
Zhou X, Zhai Y, Liu C, Yang G, Guo J, Li G, Sun C, Qi X, Li X, Guan F. Sialidase NEU1 suppresses progression of human bladder cancer cells by inhibiting fibronectin-integrin α5β1 interaction and Akt signaling pathway. Cell Commun Signal 2020; 18:44. [PMID: 32164705 PMCID: PMC7066847 DOI: 10.1186/s12964-019-0500-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/16/2019] [Indexed: 01/17/2023] Open
Abstract
Background Sialic acids are widely distributed in animal tissues, and aberrantly expressed in a variety of cancer types. High expression of sialic acid contributes to tumor aggressiveness by promoting cell proliferation, migration, angiogenesis, and metastasis. Sialidases are responsible for removal of sialic acids from glycoproteins and glycolipids. Methods N-glycomics of bladder cancer cells were detected by MALDI-TOF mass spectrometry. Sialic acid modification in bladder cancer tissue was determined by lectin blot. The down-regulation of NEU1 in bladder cancer cells was determined by high resolution liquid chromatography mass spectrometry (HR LC-MS). The effects of sialidase NEU1 expression on proliferation and apoptosis of human bladder cancer cells were examined by western blot, RT-PCR, confocal imaging and flow cytometry. Moreover, the function of sialic acids on fibronectin-integrin α5β1 interaction were assayed by immunoprecipitation and ELISA. The importance of NEU1 in tumor formation in vivo was performed using BALB/c-nu mice. Expression of NEU1 in primary human bladder cancer tissue samples was estimated using bladder cancer tissue microarray. Results (1) Downregulation of NEU1 was primarily responsible for aberrant expression of sialic acids in bladder cancer cells. (2) Decreased NEU1 expression was correlated with bladder cancer progression. (3) NEU1 overexpression enhanced apoptosis and reduced proliferation of bladder cancer cells. (4) NEU1 disrupted FN-integrin α5β1 interaction and deactivated the Akt signaling pathway. (5) NEU1 significantly suppressed in vivo tumor formation in BALB/c-nu mice. Conclusions Our data showed that NEU1 inhibited cancer cell proliferation, induced apoptosis, and suppressed tumor formation both in vitro and in vivo, by disrupting interaction of FN and integrin β1 and inhibiting the Akt signaling pathway. Our observations indicate that NEU1 is an important modulator of the malignant properties of bladder cancer cells, and is a potential therapeutic target for prognosis and treatment of bladder cancer. Video Abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Yanhong Zhai
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Changmei Liu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Ganglong Yang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Jia Guo
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Guang Li
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Chengwen Sun
- Department of Urology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiang Li
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China. .,Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Science, Northwest University, Xi'an, China.
| |
Collapse
|
27
|
Chae HS, Xu R, Won JY, Chin YW, Yim H. Molecular Targets of Genistein and Its Related Flavonoids to Exert Anticancer Effects. Int J Mol Sci 2019; 20:E2420. [PMID: 31100782 PMCID: PMC6566427 DOI: 10.3390/ijms20102420] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 02/08/2023] Open
Abstract
Increased health awareness among the public has highlighted the health benefits of dietary supplements including flavonoids. As flavonoids target several critical factors to exert a variety of biological effects, studies to identify their target-specific effects have been conducted. Herein, we discuss the basic structures of flavonoids and their anticancer activities in relation to the specific biological targets acted upon by these flavonoids. Flavonoids target several signaling pathways involved in apoptosis, cell cycle arrest, mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/AKT kinase, and metastasis. Polo-like kinase 1 (PLK1) has been recognized as a valuable target in cancer treatment due to the prognostic implication of PLK1 in cancer patients and its clinical relevance between the overexpression of PLK1 and the reduced survival rates of several carcinoma patients. Recent studies suggest that several flavonoids, including genistein directly inhibit PLK1 inhibitory activity. Later, we focus on the anticancer effects of genistein through inhibition of PLK1.
Collapse
Affiliation(s)
- Hee-Sung Chae
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi-do 10326, Korea.
| | - Rong Xu
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea.
| | - Jae-Yeon Won
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea.
| | - Young-Won Chin
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi-do 10326, Korea.
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea.
| |
Collapse
|
28
|
Lin F, Zhu Y, Hu G. Naringin promotes cellular chemokine synthesis and potentiates mesenchymal stromal cell migration via the Ras signaling pathway. Exp Ther Med 2018; 16:3504-3510. [PMID: 30233702 PMCID: PMC6143896 DOI: 10.3892/etm.2018.6634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
Directional migration of mesenchymal stem cells (MSCs) is known to serve roles in bone fracture healing. Naringin is a traditional medicine used in China to treat bone injury and has been confirmed to act as a chemoattractant to MSCs. In the present study, the secretion of chemokines and stimulation of relevant signaling pathways by naringin were detected to determine the molecular mechanism of naringin-induced MSC migration. In these experiments, Quantibody® arrays were used to detect chemokines secreted by MSCs with or without the addition of naringin. The results revealed differential naringin-induced chemokine secretion of C-X-C motif chemokine (CXCL)5, CXCL6 and C-C motif chemokine 20. Furthermore, the Ras signaling pathway was markedly activated in the naringin-treated groups, suggesting that naringin may enhance the migrational ability of MSCs via Ras activation. Furthermore, naringin was able to promote the secretion of various chemokines derived from MSCs, which would, in turn, increase the mobility of MSCs. The aim of the present study was to provide novel candidate agents for clinical orthopedics and theoretical basis for the future improvement of adjunctive medication for bone fracture healing.
Collapse
Affiliation(s)
- Feng Lin
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Yuan Zhu
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Gangfeng Hu
- Department of Orthopedics, Xiaoshan First People's Hospital, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
29
|
Khamis AAA, Ali EMM, El-Moneim MAA, Abd-Alhaseeb MM, El-Magd MA, Salim EI. Hesperidin, piperine and bee venom synergistically potentiate the anticancer effect of tamoxifen against breast cancer cells. Biomed Pharmacother 2018; 105:1335-1343. [PMID: 30021371 DOI: 10.1016/j.biopha.2018.06.105] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023] Open
Abstract
Despite advances in cancer treatment, breast cancer remains one of the main life threatening diseases in women. Most anti-breast cancer drugs cause severe health complications and multidrug resistance. Although, some natural products, such as hesperidin (Hes), piperine (Pip) and bee venom (BV), showed anti-breast cancer effect when used separately, their combined effect together or with the anti-cancer drug tamoxifen (Tam) has not yet been studied. Herein, we hypothesized that these three natural products could potentiate the therapeutic effect of Tam when used together. First, we studied the cytotoxic effect of Hes, Pip, and BV on MCF7 and T47D cells using MTT assay and found reasonable IC50 comparable to that of Tam. Second, we checked the effect of all combinations (n = 67 for each cell line, prepared as non-constant ratio from fractions of IC50 of the four compounds) and found enhanced anti-proliferative effects on MCF7 and T47D and synergistic effect, revealed by combination index (CI) values below one. Next, the best 5 combinations with lowest Tam doses and CI but with highest cell death were selected for further molecular analysis in comparison to single-drug treatment. All single- and combined-treated groups showed a significant increase in apoptosis (indicated by upregulated mRNA level of the pro-apoptotic marker Bax and downregulated mRNA level of the anti-apoptotic marker Bcl2) and a significant decrease in mRNA level of the two breast cancer related receptors EGFR and ERα, with the best effect in combined groups especially that contained the 4 compounds, as compared to vehicle-treated group. Moreover, Pip, BV and all combinations, except Tam + Hes group, arrested MCF7 and T47D in G2/M phase of cell cycle, while Tam and/or Hes caused G0/G1 phase arrest. These results indicate that Hes, Pip and BV synergistically enhance the anti-cancer effect of Tam and could be used as safe adjuvant/vehicle to Tam in treatment of breast cancer after further confirmatory in vivo investigations.
Collapse
Affiliation(s)
- Abeer A A Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Ehab M M Ali
- Biochemistry Division, Chemistry Department, Faculty of Science, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Mohamed A Abd El-Moneim
- Biochemistry Department, Faculty of Dentistry, Sinai University, Al-Arish, North Sinai, Egypt.
| | - Mohammad M Abd-Alhaseeb
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Mohammed Abu El-Magd
- Anatomy Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt.
| | - Elsayed I Salim
- Zoology Department, Faculty of Science, Tanta University, Tanta
| |
Collapse
|
30
|
Unique nuclear localization of Nile tilapia ( Oreochromis niloticus ) Neu4 sialidase is regulated by nuclear transport receptor importin α/β. Biochimie 2018; 149:92-104. [DOI: 10.1016/j.biochi.2018.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/04/2018] [Indexed: 02/02/2023]
|
31
|
Křivohlavá R, Grobárová V, Neuhöferová E, Fišerová A, Benson V. Interaction of colon cancer cells with glycoconjugates triggers complex changes in gene expression, glucose transporters and cell invasion. Mol Med Rep 2018; 17:5508-5517. [PMID: 29393416 DOI: 10.3892/mmr.2018.8490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/15/2017] [Indexed: 11/06/2022] Open
Abstract
Glycan metabolism balance is critical for cell prosperity, and macromolecule glycosylation is essential for cell communication, signaling and survival. Thus, glycotherapy may be a potential cancer treatment. The aim of the present study was to determine whether combined synthetic glycoconjugates (GCs) induce changes in gene expression that alter the survival of colon cancer cells. The current study evaluated the effect of the GCs N‑acetyl‑D‑glucosamine modified polyamidoamine dendrimer and calix[4]arene scaffold on cancer cell proliferation, apoptosis, invasion and sensitivity to immune cell‑mediated killing. Using reverse transcription‑quantitative polymerase chain reaction, the expression of genes involved in the aforementioned processes was measured. It was determined that GCs reduce the expression of the glucosaminyltransferases Mgat3 and Mgat5 responsible for surface glycosylation and employed components of the Wnt signaling pathway Wnt2B and Wnt9B. In addition, the calix[4]arene‑based GC reduced cell colony formation; this was accompanied by the downregulation of the metalloproteinase Mmp3. By contrast, the dendrimer‑based GC affected the expression of the glucose transporter components Sglt1 and Egfr1. Therefore, to the best of our knowledge, the present study is the first to reveal that N‑acetyl‑D‑glucosamine‑dendrimer/calix[4]arene GCs alter mRNA expression in a comprehensive way, resulting in the reduced malignant phenotype of the colon cancer cell line HT‑29.
Collapse
Affiliation(s)
- Romana Křivohlavá
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Valika Grobárová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Eva Neuhöferová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Anna Fišerová
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| | - Veronika Benson
- Laboratory of Molecular Biology and Immunology, Department of Immunology, Institute of Microbiology, Czech Academy of Sciences, 14220 Prague 4, Czech Republic
| |
Collapse
|
32
|
Groux-Degroote S, Rodríguez-Walker M, Dewald JH, Daniotti JL, Delannoy P. Gangliosides in Cancer Cell Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:197-227. [DOI: 10.1016/bs.pmbts.2017.10.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
33
|
Xie C, Yagai T, Luo Y, Liang X, Chen T, Wang Q, Sun D, Zhao J, Ramakrishnan SK, Sun L, Jiang C, Xue X, Tian Y, Krausz KW, Patterson AD, Shah YM, Wu Y, Jiang C, Gonzalez FJ. Activation of intestinal hypoxia-inducible factor 2α during obesity contributes to hepatic steatosis. Nat Med 2017; 23:1298-1308. [PMID: 29035368 PMCID: PMC6410352 DOI: 10.1038/nm.4412] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease is becoming the most common chronic liver disease in Western countries, and limited therapeutic options are available. Here we uncovered a role for intestinal hypoxia-inducible factor (HIF) in hepatic steatosis. Human-intestine biopsies from individuals with or without obesity revealed that intestinal HIF-2α signaling was positively correlated with body-mass index and hepatic toxicity. The causality of this correlation was verified in mice with an intestine-specific disruption of Hif2a, in which high-fat-diet-induced hepatic steatosis and obesity were substantially lower as compared to control mice. PT2385, a HIF-2α-specific inhibitor, had preventive and therapeutic effects on metabolic disorders that were dependent on intestine HIF-2α. Intestine HIF-2α inhibition markedly reduced intestine and serum ceramide levels. Mechanistically, intestine HIF-2α regulates ceramide metabolism mainly from the salvage pathway, by positively regulating the expression of Neu3, the gene encoding neuraminidase 3. These results suggest that intestinal HIF-2α could be a viable target for hepatic steatosis therapy.
Collapse
Affiliation(s)
- Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tomoki Yagai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xianyi Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Tao Chen
- Department of Internal Medicine, Key Laboratory of Environment and Genes Related to Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qiong Wang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Dongxue Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jie Zhao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sadeesh K Ramakrishnan
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lulu Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chunmei Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiang Xue
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Yatrik M Shah
- Departments of Molecular & Integrative Physiology, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yue Wu
- Department of Internal Medicine, Key Laboratory of Environment and Genes Related to Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- These authors jointly directed this work
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- These authors jointly directed this work
| |
Collapse
|
34
|
Shiozaki K, Yoshikawa M, Kiguchiya S, Ikeda A, Kamada Y, Chigwechokha PK, Komatsu M. Matrix metalloproteinase-7 inhibitory activity of lipid extract from dwarf gulper shark (Centrophorus atromarginatus) through down-regulation of gene transcription. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
35
|
Shinyoshi S, Kamada Y, Matsusaki K, Chigwechokha PK, Tepparin S, Araki K, Komatsu M, Shiozaki K. Naringenin suppresses Edwardsiella tarda infection in GAKS cells by NanA sialidase inhibition. FISH & SHELLFISH IMMUNOLOGY 2017; 61:86-92. [PMID: 27988309 DOI: 10.1016/j.fsi.2016.12.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/11/2016] [Accepted: 12/13/2016] [Indexed: 06/06/2023]
Abstract
Edwardsiella tarda (E. tarda) is a gram-negative bacterium, which causes Edwardsiellosis in aquaculture. Previous studies indicate that E. tarda NanA sialidase plays crucial roles in infection through the desialylation of glycoproteins in fish cells. On the other hand, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid, classic sialidase inhibitor, negatively regulates E. tarda infection of goldfish scale GAKS cells. Here, to development the suppression model of E. tarda infection for aquaculture application, the possibility of NanA inhibitory activities in citrus phytochemicals was evaluated as citrus extracts have widely been used as a supplement in fish diets for the improvement of meat quality. Some flavanones such as naringenin, hesperetin, hesperidin and naringin showed sialidase inhibitory activity toward recombinant NanA in vitro. Among them, naringenin showed the most potent inhibitory activity and its inhibitory pattern was non-competitive. Naringenin significantly suppressed E. tarda infection in GAKS cells at 200 and 400 μM without bactericidal effect on E. tarda. On the other hand, naringin, glycosylation form of naringenin, showed slight suppression of E. tarda infection toward GAKS cells, suggesting the glycosides on flavanone could be important for NanA inhibition. Fluorescence microscopy analysis verified that number of invading E. tarda in GAKS cells was declined by naringenin treatment. The present study exhibited the possibility of naringenin as an effective ingredient in fish diet for the inhibition of E. tarda infection.
Collapse
Affiliation(s)
| | - Yuko Kamada
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Koki Matsusaki
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan
| | - Petros Kingstone Chigwechokha
- Department of Fisheries, Mzuzu University, Mzuzu, Malawi; The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | | | - Kyosuke Araki
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Masaharu Komatsu
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Kazuhiro Shiozaki
- Faculty of Fisheries, Kagoshima University, Kagoshima, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
36
|
Cirmi S, Ferlazzo N, Lombardo GE, Maugeri A, Calapai G, Gangemi S, Navarra M. Chemopreventive Agents and Inhibitors of Cancer Hallmarks: May Citrus Offer New Perspectives? Nutrients 2016; 8:E698. [PMID: 27827912 PMCID: PMC5133085 DOI: 10.3390/nu8110698] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 12/12/2022] Open
Abstract
Fruits and vegetables have long been recognized as potentially important in the prevention of cancer risk. Thus, scientific interest in nutrition and cancer has grown over time, as shown by increasing number of experimental studies about the relationship between diet and cancer development. This review attempts to provide an insight into the anti-cancer effects of Citrus fruits, with a focus on their bioactive compounds, elucidating the main cellular and molecular mechanisms through which they may protect against cancer. Scientific literature was selected for this review with the aim of collecting the relevant experimental evidence for the anti-cancer effects of Citrus fruits and their flavonoids. The findings discussed in this review strongly support their potential as anti-cancer agents, and may represent a scientific basis to develop nutraceuticals, food supplements, or complementary and alternative drugs in a context of a multi-target pharmacological strategy in the oncology.
Collapse
Affiliation(s)
- Santa Cirmi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98168, Italy.
| | - Nadia Ferlazzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98168, Italy.
| | - Giovanni E Lombardo
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro I-88100, Italy.
| | - Alessandro Maugeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98168, Italy.
| | - Gioacchino Calapai
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina I-98125, Italy.
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, University of Messina, Messina I-98125, Italy.
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council (CNR), Pozzuoli I-80078, Italy.
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina I-98168, Italy.
| |
Collapse
|