1
|
Cui G, Fan Y, Yang Y, Ma Y, Deng H, Wang P, Zhu Y, Li J, Wei J, Zhang Y. Discovery of N-Trifluoromethylated Noscapines as Novel and Potent Agents for the Treatment of Glioblastoma. J Med Chem 2024. [PMID: 39688535 DOI: 10.1021/acs.jmedchem.4c01786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The search for new and effective chemotherapeutic agents for the treatment of glioblastoma (GBM) represents an unmet need in drug discovery. Herein, a class of novel N-trifluoromethylated noscapines has been disclosed. Among them, 9'-bromo-N-trifluoromethyl noscapine 15c displayed superior in vitro anti-GBM potency. Unexpectedly, in contrast with the general N-trifluoromethyl amines, these compounds exhibited good hydrolytic stability and further investigation of this distinct stability revealed a novel strategy for the structure modification of tetrahydroisoquinoline alkaloids, where N-methyl could be bioisosterically replaced with trifluoromethyl. Furthermore, 15c showed excellent BBB permeability and good in vivo anti-GBM activity and could efficiently suppress the migration of GBM cells, while no apparent toxicity was observed, thus representing an attractive lead for further drug discovery. Further mechanistic studies revealed that 15c exhibited an ability to induce G2/M-phase arrest in GBM cells associated with the disruption of tubulin polymerization, which is consistent with the mechanism of action of noscapine.
Collapse
Affiliation(s)
- Guangwei Cui
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yuhang Fan
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yue Yang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yiwen Ma
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Haiyang Deng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Pan Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yuxin Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jinlian Wei
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yongqiang Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
2
|
Tornese R, Montefusco A, Placì R, Semeraro T, Durante M, De Caroli M, Calabrese G, Caprifico AE, Lenucci MS. Antiangiogenic Potential of Pomegranate Extracts. PLANTS (BASEL, SWITZERLAND) 2024; 13:3350. [PMID: 39683144 DOI: 10.3390/plants13233350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Pomegranate (Punica granatum L.) has long been recognised for its rich antioxidant profile and potential health benefits. Recent research has expanded its therapeutic potential to include antiangiogenic properties, which are crucial for inhibiting the growth of tumours and other pathological conditions involving aberrant blood vessel formation. This review consolidates current findings on the antiangiogenic effects of pomegranate extracts. We explore the impact of pomegranate polyphenols, including ellagic acid, punicalagin, anthocyanins, punicic acid and bioactive polysaccharides on key angiogenesis-related pathways and endothelial cell function. Emphasis is placed on the effects of these extracts as phytocomplexes rather than isolated compounds. Additionally, we discuss the use of pomegranate by-products, such as peels and seeds, in the preparation of extracts within a green chemistry and circular economy framework, highlighting their value in enhancing extract efficacy and sustainability. By primarily reviewing in vitro and in vivo preclinical studies, we assess how these extracts modulate angiogenesis across various disease models and explore their potential as adjunctive therapies for cancer and other angiogenesis-driven disorders. This review also identifies existing knowledge gaps and proposes future research directions to fully elucidate the clinical utility of pomegranate extracts in therapeutic applications.
Collapse
Affiliation(s)
- Riccardo Tornese
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Prov.le Lecce Monteroni, 73100 Lecce, Italy
| | - Anna Montefusco
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Prov.le Lecce Monteroni, 73100 Lecce, Italy
| | - Rocco Placì
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Prov.le Lecce Monteroni, 73100 Lecce, Italy
| | - Teodoro Semeraro
- Research Institute on Terrestrial Ecosystems (IRET-URT Lecce), National Research Council of Italy (CNR), Campus Ecotekne, 73100 Lecce, Italy
| | - Miriana Durante
- Istituto di Scienze delle Produzioni Alimentari (ISPA)-CNR, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy
| | - Monica De Caroli
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Prov.le Lecce Monteroni, 73100 Lecce, Italy
- NBCF National Biodiversity Future Center, 90133 Palermo, Italy
| | - Gianpiero Calabrese
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK
| | - Anna Eleonora Caprifico
- School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Marcello Salvatore Lenucci
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Prov.le Lecce Monteroni, 73100 Lecce, Italy
| |
Collapse
|
3
|
Wang J, Sun M, Yu J, Wang J, Cui Q. Pomegranate seeds: a comprehensive review of traditional uses, chemical composition, and pharmacological properties. Front Pharmacol 2024; 15:1401826. [PMID: 39055489 PMCID: PMC11269133 DOI: 10.3389/fphar.2024.1401826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Pomegranate seeds (PS) are the dried seeds derived from pomegranate fruit, accounting for approximately 20% of the fruit's total weight, and are a by-product of pomegranate juice extraction. These seeds hold significance in traditional medicine among Uyghurs and Tibetan cultures, featuring diverse clinical applications within traditional Chinese medicine. These applications include management of gastric coldness and acidity, abdominal distension, liver and gallbladder fever, and pediatric enteritis. PS demonstrates properties such as stomach tonicity, qi regulation, analgesia, and anti-inflammatory effects. Extensive research underscores the richness of PS in various phytochemical compounds and metabolites, notably unsaturated fatty acids (particularly linolenic acid and linoleic acid), phenolic compounds tocopherols, proteins, and volatile oils. Notably, among these bioactive compounds, punicic acid (PA), found within PS, demonstrates potential in the prevention and treatment of cancers, diabetes, obesity, and other ailments. Despite extensive literature on pomegranate as a botanical entity, a comprehensive review focusing specifically on the chemical composition and pharmacological effects of PS remains elusive. Therefore, this review aimed to consolidate knowledge regarding the medicinal properties of PS, summarizing its chemical composition, traditional uses, and pharmacological effects in treating various diseases, thereby laying a foundation for the advancement and application of PS in the field of pharmacology.
Collapse
Affiliation(s)
- Jian Wang
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, China
| | - Mengjie Sun
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, China
| | - Jian Yu
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, China
| | - Jinglong Wang
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, China
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Liu Y, Hamid N, Manzoor R, Zhang BF, Liao YL, Wang JX, Pei DS. PPARβ/δ-ANGPTL4 axis mediates the promotion of mono-2-ethylhexyl phthalic acid on MYCN-amplified neuroblastoma development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168949. [PMID: 38042186 DOI: 10.1016/j.scitotenv.2023.168949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/25/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Abstract
Di-2-ethylhexyl phthalic acid (DEHP) is one of the most widely used plasticizers in the industry, which can improve the flexibility and durability of plastics. It is prone to migrate from various daily plastic products through wear and leaching into the surrounding environment and decompose into the more toxic metabolite mono-2-ethylhexyl phthalic acid (MEHP) after entering the human body. However, the impacts and mechanisms of MEHP on neuroblastoma are unclear. We exposed MYCN-amplified neuroblastoma SK-N-BE(2)C cells to an environmentally related concentration of MEHP and found that MEHP increased the proliferation and migration ability of tumor cells. The peroxisome proliferator-activated receptor (PPAR) β/δ pathway was identified as a pivotal signaling pathway in neuroblastoma, mediating the effects of MEHP through transcriptional sequencing analysis. Because MEHP can bind to the PPARβ/δ protein and initiate the expression of the downstream gene angiopoietin-like 4 (ANGPTL4), the PPARβ/δ-specific agonist GW501516 and antagonist GSK3787, the recombinant human ANGPTL4 protein, and the knockdown of gene expression confirmed the regulation of the PPARβ/δ-ANGPTL4 axis on the malignant phenotype of neuroblastoma. Based on the critical role of PPARβ/δ and ANGPTL4 in the metabolic process, a non-targeted metabolomics analysis revealed that MEHP altered multiple metabolic pathways, particularly lipid metabolites involving fatty acyls, glycerophospholipids, and sterol lipids, which may also be potential factors promoting tumor progression. We have demonstrated for the first time that MEHP can target binding to PPARβ/δ and affect the progression of neuroblastoma by activating the PPARβ/δ-ANGPTL4 axis. This mechanism confirms the health risks of plasticizers as tumor promoters and provides new data support for targeted prevention and treatment of neuroblastoma.
Collapse
Affiliation(s)
- Yiyun Liu
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Naima Hamid
- Faculty of Science and Marine Environment, Ocean Pollution and Ecotoxicology (OPEC) Research Group, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Rakia Manzoor
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Bao-Fu Zhang
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yan-Ling Liao
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jin-Xia Wang
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
5
|
Van Dingenen L, Segers C, Wouters S, Mysara M, Leys N, Kumar-Singh S, Malhotra-Kumar S, Van Houdt R. Dissecting the role of the gut microbiome and fecal microbiota transplantation in radio- and immunotherapy treatment of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1298264. [PMID: 38035338 PMCID: PMC10687483 DOI: 10.3389/fcimb.2023.1298264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and poses a major burden on the human health worldwide. At the moment, treatment of CRC consists of surgery in combination with (neo)adjuvant chemotherapy and/or radiotherapy. More recently, immune checkpoint blockers (ICBs) have also been approved for CRC treatment. In addition, recent studies have shown that radiotherapy and ICBs act synergistically, with radiotherapy stimulating the immune system that is activated by ICBs. However, both treatments are also associated with severe toxicity and efficacy issues, which can lead to temporary or permanent discontinuation of these treatment programs. There's growing evidence pointing to the gut microbiome playing a role in these issues. Some microorganisms seem to contribute to radiotherapy-associated toxicity and hinder ICB efficacy, while others seem to reduce radiotherapy-associated toxicity or enhance ICB efficacy. Consequently, fecal microbiota transplantation (FMT) has been applied to reduce radio- and immunotherapy-related toxicity and enhance their efficacies. Here, we have reviewed the currently available preclinical and clinical data in CRC treatment, with a focus on how the gut microbiome influences radio- and immunotherapy toxicity and efficacy and if these treatments could benefit from FMT.
Collapse
Affiliation(s)
- Lena Van Dingenen
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Charlotte Segers
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shari Wouters
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Mohamed Mysara
- Bioinformatics Group, Center for Informatics Science, School of Information Technology and Computer Science, Nile University, Giza, Egypt
| | - Natalie Leys
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Rob Van Houdt
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| |
Collapse
|
6
|
Grape, Pomegranate, Olive, and Tomato By-Products Fed to Dairy Ruminants Improve Milk Fatty Acid Profile without Depressing Milk Production. Foods 2023; 12:foods12040865. [PMID: 36832939 PMCID: PMC9957115 DOI: 10.3390/foods12040865] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The continuous increase in the cost of feeds and the need to improve the sustainability of animal production require the identification of alternative feeds, such as those derived from the agro-industrial sector, that can be effectively used for animal nutrition. Since these by-products (BP) are sources of bioactive substances, especially polyphenols, they may play an important role as a new resource for improving the nutritional value of animal-derived products, being effective in the modulation of the biohydrogenation process in the rumen, and, hence, in the composition of milk fatty acids (FA). The main objective of this work was to evaluate if the inclusion of BP in the diets of dairy ruminants, as a partial replacement of concentrates, could improve the nutritional quality of dairy products without having negative effects on animal production traits. To meet this goal, we summarized the effects of widespread agro-industrial by-products such as grape pomace or grape marc, pomegranate, olive cake, and tomato pomace on milk production, milk composition, and FA profile in dairy cows, sheep, and goats. The results evidenced that substitution of part of the ratio ingredients, mainly concentrates, in general, does not affect milk production and its main components, but at the highest tested doses, it can depress the yield within the range of 10-12%. However, the general positive effect on milk FA profile was evident by using almost all BP at different tested doses. The inclusion of these BP in the ration, from 5% up to 40% of dry matter (DM), did not depress milk yield, fat, or protein production, demonstrating positive features in terms of both economic and environmental sustainability and the reduction of human-animal competition for food. The general improvement of the nutritional quality of milk fat related to the inclusion of these BP in dairy ruminant diets is an important advantage for the commercial promotion of dairy products resulting from the recycling of agro-industrial by-products.
Collapse
|
7
|
Progress in targeting PTEN/PI3K/Akt axis in glioblastoma therapy: Revisiting molecular interactions. Biomed Pharmacother 2023; 158:114204. [PMID: 36916430 DOI: 10.1016/j.biopha.2022.114204] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is one of the most malignant cancers of central nervous system and due to its sensitive location, surgical resection has high risk and therefore, chemotherapy and radiotherapy are utilized for its treatment. However, chemoresistance and radio-resistance are other problems in GBM treatment. Hence, new therapies based on genes are recommended for treatment of GBM. PTEN is a tumor-suppressor operator in cancer that inhibits PI3K/Akt/mTOR axis in diminishing growth, metastasis and drug resistance. In the current review, the function of PTEN/PI3K/Akt axis in GBM progression is evaluated. Mutation or depletion of PTEN leads to increase in GBM progression. Low expression level of PTEN mediates poor prognosis in GBM and by increasing proliferation and invasion, promotes malignancy of tumor cells. Moreover, loss of PTEN signaling can result in therapy resistance in GBM. Activation of PTEN signaling impairs GBM metabolism via glycolysis inhibition. In contrast to PTEN, PI3K/Akt signaling has oncogenic function and during tumor progression, expression level of PI3K/Akt enhances. PI3K/Akt signaling shows positive association with oncogenic pathways and its expression similar to PTEN signaling, is regulated by non-coding RNAs. PTEN upregulation and PI3K/Akt signaling inhibition by anti-cancer agents can be beneficial in interfering GBM progression. This review emphasizes on the signaling networks related to PTEN/PI3K/Akt and provides new insights for targeting this axis in effective GBM treatment.
Collapse
|
8
|
Hu T, Chen X, Lu S, Zeng H, Guo L, Han Y. Biological Role and Mechanism of Lipid Metabolism Reprogramming Related Gene ECHS1 in Cancer. Technol Cancer Res Treat 2022; 21:15330338221140655. [PMID: 36567598 PMCID: PMC9806408 DOI: 10.1177/15330338221140655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cancer is a major threat to human health today. Although the existing anticancer treatments have effectively improved the prognosis of some patients, there are still other patients who cannot benefit from these well-established strategies. Reprogramming of lipid metabolism is one of the typical features of cancers. Recent studies have revealed that key enzymes involved in lipid metabolism may be effective anticancer therapeutic targets, but the development of therapeutic lipid metabolism targets is still insufficient. ECHS1 (enoyl-CoA hydratase, short chain 1) is a key enzyme mediating the hydration process of mitochondrial fatty acid β-oxidation and has been observed to be abnormally expressed in a variety of cancers. Therefore, with ECHS1 and cancer as the main keywords, we searched the relevant studies of ECHS1 in the field of cancer in Pubmed, summarized the research status and functions of ECHS1 in different cancer contexts, and explored its potential regulatory mechanisms, with a view to finding new therapeutic targets for anti-metabolic therapy. By reviewing and summarizing the retrieved literatures, we found that ECHS1 regulates malignant biological behaviors such as cell proliferation, metastasis, apoptosis, autophagy, and drug resistance by remodeling lipid metabolism and regulating intercellular oncogenic signaling pathways. Not only that, ECHS1 exhibits early diagnostic and prognostic value in clear cell renal cell carcinoma, and small-molecule inhibitors that regulate ECHS1 also show therapeutic significance in preclinical studies. Taken together, we propose that ECHS1 has the potential to serve as a therapeutic target of lipid metabolism.
Collapse
Affiliation(s)
- Teng Hu
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Xiaojing Chen
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Simin Lu
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Hao Zeng
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Lu Guo
- Department of Ophthalmology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest
Medical University, Luzhou, Sichuan, China,Yunwei Han, Department of Oncology, The
Affiliated Hospital of Southwest Medical University, Taiping Street, No. 25,
Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
9
|
Rakhshandeh H, Baradaran Rahimi V, Habibi Z, Sirousi Z, Askari VR. Punica granatum seed oil detracts peritoneal adhesion: Perusing antioxidant, anti-inflammatory, antifibrotic, and antiangiogenic impacts. Physiol Rep 2022; 10:e15545. [PMID: 36541264 PMCID: PMC9768730 DOI: 10.14814/phy2.15545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023] Open
Abstract
Peritoneal adhesion is a significant problem following gastrointestinal surgeries, accompanied by a significant economic burden and morbidity for patients. Punica granatum seed oil (PSO) possesses antioxidative, anti-inflammatory, and anticancer effects. Thus, we aimed to evaluate the antiperitoneal adhesive properties of PSO in rats. Forty-eight Wistar rats (200-250 g) were randomly and equally divided into six groups: sham group, control group; peritoneal adhesion without any treatment, vehicle group; peritoneal adhesion with saline + Tween-80.5% treatment, and experimental groups; peritoneal adhesion with 0.5%, 1.5%, and 4.5% v/v PSO treatment. In addition, peritoneal adhesion was examined macroscopically along with evaluating the oxidative stress (malondialdehyde [MDA], nitric oxide [NO], and glutathione [GSH]) inflammatory (interleukin [IL]-6, IL-1β, and tumor necrosis factor-α [TNF-α]), fibrotic (transforming growth factor-β [TGF-β]), and angiogenic (vascular endothelial growth factor [VEGF]) factors. Our results revealed that the levels of adhesion scores, MDA, NO, IL-6, TNF-α, IL-1β, TGF-β, and VEGF, were propagated in the vehicle group while the GSH level was alleviated (p < 0.001). In contrast, premedication with PSO, especially at the lowest concentration, notably lessened the levels of adhesion scores, MDA, NO, IL-6, TNF-α, IL-1β, TGF-β, and VEGF as well as GSH in comparison to the vehicle group following the peritoneal adhesion induction (p < 0.001-0.05). As a result, PSO may prevent peritoneal adhesion through its antioxidant, anti-inflammatory, antifibrotic, and antiangiogenic properties. Therefore, PSO could be considered a beneficial candidate for the treatment of postoperative peritoneal adhesion.
Collapse
Affiliation(s)
- Hassan Rakhshandeh
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical SciencesMashhadIran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Zahra Habibi
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical SciencesMashhadIran
| | - Zahra Sirousi
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical SciencesMashhadIran
| | - Vahid Reza Askari
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
- Applied Biomedical Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
10
|
Yang P, Yang X, Liu H, He G, Yao Q, Yan J. Isolation of punicic acid from pomegranate seed oil by modified freeze crystallization and response surface methodology. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.17154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Ping Yang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy Chengdu University Chengdu China
- Chengdu Brilliant Pharmaceutical Co., Ltd. Chengdu China
| | - Xiulan Yang
- School of Food and Bioengineering Chengdu University Chengdu China
| | - Hui Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy Chengdu University Chengdu China
| | - Gang He
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy Chengdu University Chengdu China
| | - Qian Yao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy Chengdu University Chengdu China
| | - Jun Yan
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy Chengdu University Chengdu China
| |
Collapse
|
11
|
Lee HS, Lee IH, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. Unveiling the Mechanism of the Traditional Korean Medicinal Formula FDY003 on Glioblastoma Through a Computational Network Pharmacology Approach. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221126311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is the most common type of primary malignant tumor that develops in the brain, with 0.21 million new cases per year globally and a median survival period of less than 2 years after diagnosis. Traditional Korean medicines have been increasingly suggested as effective and safe therapeutic strategies for GBM. However, their pharmacological effects and mechanistic characteristics remain to be studied. In this study, we employed a computational network pharmacological approach to determine the effects and mechanisms of the traditional Korean medicinal formula FDY003 on GBM. We found that FDY003 treatment decreased the viability of human GBM cells and increased their response to chemotherapeutics. We identified 10 potential active pharmacological compounds of FDY003 and 67 potential GBM-related target genes and proteins. The GBM-related targets of FDY003 were signaling components of various crucial GBM-associated pathways, such as PI3K-Akt, focal adhesion, MAPK, HIF-1, FoxO, Ras, and TNF. These pathways are functional regulators for the determination of cell growth and proliferation, survival and death, and cell division cycle of GBM cells. Together, the overall analyses contribute to the pharmacological basis for the anti-GBM roles of FDY003 and its systematic mechanisms.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | - In-Hee Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | | | - Minho Jung
- Forest Hospital, Seoul, Republic of Korea
| | | | | | - Dae-Yeon Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| |
Collapse
|
12
|
Huang J, Zheng X, Kang W, Hao H, Mao Y, Zhang H, Chen Y, Tan Y, He Y, Zhao W, Yin Y. Metagenomic and metabolomic analyses reveal synergistic effects of fecal microbiota transplantation and anti-PD-1 therapy on treating colorectal cancer. Front Immunol 2022; 13:874922. [PMID: 35911731 PMCID: PMC9336524 DOI: 10.3389/fimmu.2022.874922] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Anti-PD-1 immunotherapy has saved numerous lives of cancer patients; however, it only exerts efficacy in 10-15% of patients with colorectal cancer. Fecal microbiota transplantation (FMT) is a potential approach to improving the efficacy of anti-PD-1 therapy, whereas the detailed mechanisms and the applicability of this combination therapy remain unclear. In this study, we evaluated the synergistic effect of FMT with anti-PD-1 in curing colorectal tumor-bearing mice using a multi-omics approach. Mice treated with the combination therapy showed superior survival rate and tumor control, compared to the mice received anti-PD-1 therapy or FMT alone. Metagenomic analysis showed that composition of gut microbiota in tumor-bearing mice treated with anti-PD-1 therapy was remarkably altered through receiving FMT. Particularly, Bacteroides genus, including FMT-increased B. thetaiotaomicron, B. fragilis, and FMT-decreased B. ovatus might contribute to the enhanced efficacy of anti-PD-1 therapy. Furthermore, metabolomic analysis upon mouse plasma revealed several potential metabolites that upregulated after FMT, including punicic acid and aspirin, might promote the response to anti-PD-1 therapy via their immunomodulatory functions. This work broadens our understanding of the mechanism by which FMT improves the efficacy of anti-PD-1 therapy, which may contribute to the development of novel microbiota-based anti-cancer therapies.
Collapse
Affiliation(s)
- Jiayuan Huang
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Xing Zheng
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Wanying Kang
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Huaijie Hao
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Yudan Mao
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Hua Zhang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yuan Chen
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Yan Tan
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
| | - Yulong He
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Wenjing Zhao
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
- *Correspondence: Yiming Yin, ; Wenjing Zhao,
| | - Yiming Yin
- Department of Research and Development, Shenzhen Xbiome Biotech Co. Ltd., Shenzhen, China
- *Correspondence: Yiming Yin, ; Wenjing Zhao,
| |
Collapse
|
13
|
Thakur A, Faujdar C, Sharma R, Sharma S, Malik B, Nepali K, Liou JP. Glioblastoma: Current Status, Emerging Targets, and Recent Advances. J Med Chem 2022; 65:8596-8685. [PMID: 35786935 PMCID: PMC9297300 DOI: 10.1021/acs.jmedchem.1c01946] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Glioblastoma (GBM) is a highly malignant
brain tumor characterized
by a heterogeneous population of genetically unstable and highly infiltrative
cells that are resistant to chemotherapy. Although substantial efforts
have been invested in the field of anti-GBM drug discovery in the
past decade, success has primarily been confined to the preclinical
level, and clinical studies have often been hampered due to efficacy-,
selectivity-, or physicochemical property-related issues. Thus, expansion
of the list of molecular targets coupled with a pragmatic design of
new small-molecule inhibitors with central nervous system (CNS)-penetrating
ability is required to steer the wheels of anti-GBM drug discovery
endeavors. This Perspective presents various aspects of drug discovery
(challenges in GBM drug discovery and delivery, therapeutic targets,
and agents under clinical investigation). The comprehensively covered
sections include the recent medicinal chemistry campaigns embarked
upon to validate the potential of numerous enzymes/proteins/receptors
as therapeutic targets in GBM.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Chetna Faujdar
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida 201307, India
| | - Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Basant Malik
- Department of Sterile Product Development, Research and Development-Unit 2, Jubiliant Generics Ltd., Noida 201301, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wuxing Street, Taipei 11031, Taiwan
| |
Collapse
|
14
|
Zhang F, Xu H, Xia R, Yu P, Li Y, Yu X, Sui D. Pseudo-ginsenoside Rh2 Induces Protective Autophagy in Hepatocellular Carcinoma HepG2 Cells. Recent Pat Anticancer Drug Discov 2021; 16:521-532. [PMID: 34109916 DOI: 10.2174/1574892816666210607100239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/02/2021] [Accepted: 03/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Pseudo-ginsenoside-Rh2 (pseudo-G-Rh2), a novel derivative of ginsenoside Rh2, is reported to exert a pro-apoptotic effect on various malignancies. However, whether this anti-cancer action of pseudo-G-Rh2 involves autophagy remains to be determined and explored. OBJECTIVES Investigation of pseudo-G-Rh2-induced apoptosis and autophagy and the underlying mechanism. METHODS In the present study, the MTT assay was used for evaluating cell viability and the lactate dehydrogenase (LDH) assay was performed to assess cell toxicity. Autophagy evaluation was performed using monodansylcadaverine (MDC) staining and transmission electron microscopy (TEM). The levels of autophagy-associated and apoptosis-associated proteins were determined using Western blotting. The Annexin V FITC/propidium iodide (PI) assay was used to assess apoptosis. RESULTS The Annexin V FITC/PI assay revealed that the percentage of apoptotic cells in HepG2 cells at concentrations 0, 20, 40, and 60 μM was 3.75%±1.37%, 5.70%±1.04%, 12.30%±2.10%, and 34.26%±4.73%, respectively. Pseudo-G-Rh2 was observed to significantly increase the expressions of BAX, cleaved-caspase-3, and cleaved-caspase-9, while it decreased the Bcl-2 expression. MDC and TEM analyses revealed that pseudo-G-Rh2 at concentrations 20, 40, and 60 μM significantly facilitated the accumulation of autophagosomes and autolysosomes within the HepG2 cells. Moreover, pseudo-G-Rh2 significantly increased the expressions of LC3 Ⅱ/LC3 Ⅰ, and Beclin-1 and decreased the expression of p62. The Annexin V FITC/PI assay also revealed that in comparison to the pseudo-G-Rh2 group, the concurrent treatment with pseudo-G-Rh2 and an autophagy inhibitor (CQ or 3-MA) significantly induced distinct apoptosis. In addition, pseudo-G-Rh2 activated AMPK and inhibited the PI3K/Akt/mTOR pathway in a concentration-dependent manner. Pseudo-G-Rh2 is similar to the current patents, which enhanced its anti-cancer activity by combining with autophagy inhibitors. CONCLUSION Pseudo-G-Rh2 could induce protective autophagy in HepG2 cells, at least in part, via AMPK and the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Fuyuan Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Rui Xia
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Ping Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuangeng Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, Jilin Province, China
| |
Collapse
|
15
|
Fuentes-Fayos AC, Vázquez-Borrego MC, Jiménez-Vacas JM, Bejarano L, Pedraza-Arévalo S, L-López F, Blanco-Acevedo C, Sánchez-Sánchez R, Reyes O, Ventura S, Solivera J, Breunig JJ, Blasco MA, Gahete MD, Castaño JP, Luque RM. Splicing machinery dysregulation drives glioblastoma development/aggressiveness: oncogenic role of SRSF3. Brain 2020; 143:3273-3293. [PMID: 33141183 PMCID: PMC7904102 DOI: 10.1093/brain/awaa273] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/17/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastomas remain the deadliest brain tumour, with a dismal ∼12–16-month survival from diagnosis. Therefore, identification of new diagnostic, prognostic and therapeutic tools to tackle glioblastomas is urgently needed. Emerging evidence indicates that the cellular machinery controlling the splicing process (spliceosome) is altered in tumours, leading to oncogenic splicing events associated with tumour progression and aggressiveness. Here, we identify for the first time a profound dysregulation in the expression of relevant spliceosome components and splicing factors (at mRNA and protein levels) in well characterized cohorts of human high-grade astrocytomas, mostly glioblastomas, compared to healthy brain control samples, being SRSF3, RBM22, PTBP1 and RBM3 able to perfectly discriminate between tumours and control samples, and between proneural-like or mesenchymal-like tumours versus control samples from different mouse models with gliomas. Results were confirmed in four additional and independent human cohorts. Silencing of SRSF3, RBM22, PTBP1 and RBM3 decreased aggressiveness parameters in vitro (e.g. proliferation, migration, tumorsphere-formation, etc.) and induced apoptosis, especially SRSF3. Remarkably, SRSF3 was correlated with patient survival and relevant tumour markers, and its silencing in vivo drastically decreased tumour development and progression, likely through a molecular/cellular mechanism involving PDGFRB and associated oncogenic signalling pathways (PI3K-AKT/ERK), which may also involve the distinct alteration of alternative splicing events of specific transcription factors controlling PDGFRB (i.e. TP73). Altogether, our results demonstrate a drastic splicing machinery-associated molecular dysregulation in glioblastomas, which could potentially be considered as a source of novel diagnostic and prognostic biomarkers as well as therapeutic targets for glioblastomas. Remarkably, SRSF3 is directly associated with glioblastoma development, progression, aggressiveness and patient survival and represents a novel potential therapeutic target to tackle this devastating pathology.
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Mari C Vázquez-Borrego
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Leire Bejarano
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Sergio Pedraza-Arévalo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Fernando L-López
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,Pathology Service, Reina Sofia University Hospital, 14004 Cordoba, Spain
| | - Oscar Reyes
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Computer Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Sebastián Ventura
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Computer Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Juan Solivera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - María A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain.,Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain.,Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain.,Agrifood Campus of International Excellence (ceiA3), 14004 Cordoba, Spain
| |
Collapse
|
16
|
A Network Pharmacology Approach to Explore the Mechanisms of Shugan Jianpi Formula in Liver Fibrosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4780383. [PMID: 32617108 PMCID: PMC7306883 DOI: 10.1155/2020/4780383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/28/2020] [Accepted: 05/26/2020] [Indexed: 12/30/2022]
Abstract
Purpose We explored the mechanism of Shugan Jianpi Formula (SGJPF) and its effective components for the treatment of liver fibrosis (LF). Materials and Methods We collected the active ingredients in SGJPF through the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform and screened the effective components by absorption, distribution, metabolism, and excretion. Herb-associated target proteins were predicted and screened based on the Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine and Search Tool for Interactions of Chemicals databases. LF-associated target proteins were predicted and screened based on the Online Mendelian Inheritance in Man® Database and Comparative Toxicogenomics Database. Common genes with LF and herbs were selected, and Cytoscape 3.5.1 software was used to construct an herb pathway and component-LF common target network. The Search Tool for the Retrieval of Interacting Genes/Proteins was used to build a protein-protein interaction, and quantitative PCR was used to verify the related target genes. Finally, clusterProfiler was applied for the analysis of Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways. Results The pharmacological network contained 252 active compounds (e.g., Astragaloside A, saikosaponin, linoleic acid, and Poria acid A), 84 common target genes, and 94 significant signaling pathways. Among them, interleukin 6 (IL-6), tumor protein 53 p53 (TP53), prostaglandin-endoperoxide synthase 2 (PTGS2), AKT1, IL-1β, and the nucleotide-binding and oligomerization domain-like receptor and Janus kinase-signal transducer and activator of transcription signaling pathways were selected as the critical target gene and critical signal pathway, respectively. Conclusion The mechanisms of SGJPF in protecting against LF include the regulation of multiple targets such as IL-6, TP53, PTGS2, and AKT1. These target proteins affect LF through various signal transduction pathways.
Collapse
|
17
|
Wei D, Zhang G, Zhu Z, Zheng Y, Yan F, Pan C, Wang Z, Li X, Wang F, Meng P, Zheng W, Yan Z, Zhai D, Lu Z, Yuan J. Nobiletin Inhibits Cell Viability via the SRC/AKT/STAT3/YY1AP1 Pathway in Human Renal Carcinoma Cells. Front Pharmacol 2019; 10:690. [PMID: 31354472 PMCID: PMC6635658 DOI: 10.3389/fphar.2019.00690] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022] Open
Abstract
Nobiletin is a polymethoxy flavonoid isolated from Citrus depressa and Citrus reticulata. It has been reported that nobiletin can suppress tumors. We primarily explored the antitumor effects of nobiletin and the associated potential mechanisms in ACHN and Caki-2 renal carcinoma cells. A CCK-8 assay and cloning experiments were used to assess cell viability, and a transwell assay and scratch test were used to assess metastatic ability. The cell cycle was analyzed by flow cytometry, whereas apoptosis was analyzed using flow cytometry and a terminal dexynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. Protein expression was examined by Western blot and immunofluorescence. Renal cancer cells were subcutaneously transplanted into nude mice for in vivo studies. The data showed that nobiletin administration significantly dose- and time-dependently suppressed renal cancer cell proliferation; moreover, nobiletin treatment induced cell cycle arrest in the G0/G1 phase and promoted apoptosis. Immunofluorescence analysis indicated that nobiletin decreased the nuclear localization of signal transducer and activator of transcription 3 (STAT3) and YY1-associated protein 1 (YY1AP1). Western blot showed that the levels of phosphorylated SRC, phosphorylated AKT serine/threonine kinase (AKT), and phosphorylated STAT3 were decreased, whereas that of phosphorylated YY1AP1 was increased. The results further showed that application of insulin-like growth factor 1 (IGF1) was able to reverse the nobiletin-induced changes in the levels of phosphorylated AKT, phosphorylated STAT3, and phosphorylated YY1AP1, and could also reverse the antitumor effects of nobiletin. The results of in vivo experiments showed that, compared to the control, tumor volume and weight were both reduced following nobiletin treatment. In conclusion, our study demonstrated that nobiletin can inhibit renal carcinoma cell viability and provides a novel therapeutic approach for the treatment of kidney cancer.
Collapse
Affiliation(s)
- Di Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Geng Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu Zheng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Yan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chongxian Pan
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Zhiyong Wang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xian Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fuli Wang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Meng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wanxiang Zheng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhao Yan
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongsheng Zhai
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, China
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, China
| | - Jianlin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|