1
|
Han P, Wei S, Wang H, Cai Y. Licochalcone A decreases cancer cell proliferation and enhances ferroptosis in acute myeloid leukemia through suppressing the IGF2BP3/MDM2 cascade. Ann Hematol 2024:10.1007/s00277-024-06003-4. [PMID: 39264435 DOI: 10.1007/s00277-024-06003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
Licochalcone A (Lico A), a naturally bioactive flavonoid, has shown antitumor activity in several types of cancers. However, few studies have focused on its effect on acute myeloid leukemia (AML). Cell viability and colony formation potential were detected by CCK-8 assay and colony formation assay, respectively. Cell cycle distribution and apoptosis were assessed by flow cytometry. Ferroptosis was assessed by measuring reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA), and glutathione (GSH). Protein expression levels were determined by immunoblotting and immunohistochemistry (IHC), and mRNA expression was detected by real-time qPCR. The m6A modification of MDM2 mRNA was verified by methylated RNA immunoprecipitation (MeRIP) assay, and the interaction of IGF2BP3 and MDM2 mRNA was analyzed by RIP assay. Actinomycin D was used to evaluate mRNA stability. The efficacy of Lico A in vivo was examined by a murine xenograft model. Lico A suppressed cell proliferation and induced ferroptosis in MOLM-13 and U-937 in vitro, and slowed the growth of xenograft tumors in vivo. IGF2BP3 was highly expressed in human AML specimens and cells, and Lico A suppressed IGF2BP3 expression in AML cells. Lico A exerted the anti-proliferative and pro-ferroptosis effects by downregulating IGF2BP3. Moreover, IGF2BP3 enhanced the stability and expression of MDM2 mRNA through an m6A-dependent manner. Downregulation of IGF2BP3 impeded AML cell proliferation and enhanced ferroptosis via repressing MDM2. Furthermore, Lico A could affect the MDM2/p53 pathway by downregulating IGF2BP3 expression. Lico A exerts the anti-proliferative and pro-ferroptosis activity in AML cells by affecting the IGF2BP3/MDM2/p53 pathway, providing new evidence for Lico A as a promising agent for the treatment of AML.
Collapse
Affiliation(s)
- Pingping Han
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China.
| | - Suhua Wei
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Huaiyu Wang
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yun Cai
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| |
Collapse
|
2
|
Han TH, Lee J, Harmalkar DS, Kang H, Jin G, Park MK, Kim M, Yang HA, Kim J, Kwon SJ, Han TS, Choi Y, Won M, Ban HS, Lee K. Stilbenoid derivatives as potent inhibitors of HIF-1α-centric cancer metabolism under hypoxia. Biomed Pharmacother 2024; 176:116838. [PMID: 38820970 DOI: 10.1016/j.biopha.2024.116838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Hypoxia-inducible factor (HIF)-1α is a crucial transcription factor associated with cancer metabolism and is regarded as a potent anticancer therapeutic strategy within the hypoxic microenvironment of cancer. In this study, stilbenoid derivatives were designed, synthesized, and assessed for their capacity to inhibit HIF-1α-associated cancer metabolism and evaluated for inhibition of cancer cell viability and HIF activation. Through the structure-activity relationship studies, compound 28e was identified as the most potent derivative. Specifically, under the hypoxic condition, 28e reduced the accumulation of HIF-1α protein and the expression of its target genes related to glucose metabolism without affecting the expression of HIF-1α mRNA. Furthermore, 28e inhibited glucose uptake, glycolytic metabolism, and mitochondrial respiration, decreasing cellular ATP production under hypoxic conditions. In addition, 28e displayed significant anti-tumor effects and effectively suppressed the accumulation of HIF-1α protein in tumor tissue in vivo xenograft model. These findings suggest that our stilbenoid derivatives exert their anticancer effects by targeting HIF-1α-centered cancer metabolism under hypoxic conditions.
Collapse
Affiliation(s)
- Tae-Hee Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Joohan Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Dipesh S Harmalkar
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; Department of Chemistry, Government College of Arts, Science and Commerce, Sanquelim, Goa 403505, India
| | - Hyeseul Kang
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Guanghai Jin
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Min Kyung Park
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Minkyoung Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Hyun-A Yang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jinsu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Su Jeong Kwon
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Tae-Su Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Yongseok Choi
- Department of Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Misun Won
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon 34141, Republic of Korea
| | - Hyun Seung Ban
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| | - Kyeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea.
| |
Collapse
|
3
|
Yang HA, Han TH, Haam K, Lee KS, Kim J, Han TS, Lee MS, Ban HS. Prodigiosin regulates cancer metabolism through interaction with GLUT1. Nat Prod Res 2024:1-8. [PMID: 38913075 DOI: 10.1080/14786419.2024.2367241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
In contrast to normal cells, cancer cells predominantly utilise glycolysis for ATP generation under aerobic conditions, facilitating proliferation and metastasis. Targeting glycolysis is effective for cancer treatment. Prodigiosin (PDG) is a natural compound with various bioactivities, including anticancer effects. However, the precise action mechanisms and molecular targets of PDG, which has demonstrated efficacy in regulating glucose metabolism in cancer cells, remain elusive. Here, we aimed to investigate the anti-cancer activity of PDG and mechanism in cancer metabolism. PDG regulated cancer metabolism by suppressing intracellular ATP production rate and levels. It inhibited glycolysis and mitochondrial oxidative phosphorylation, impeding ATP production dependent on both glycolysis and mitochondrial respiration. Moreover, it inhibited cellular glucose uptake by directly interacting with glucose transporter 1 without affecting its mRNA or protein levels in HCT116 cells. We provide insights into the anti-cancer effects of PDG mediated via cancer metabolism regulation, suggesting its therapeutic potential for cancer.
Collapse
Affiliation(s)
- Hyun-A Yang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Tae-Hee Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Keeok Haam
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kyung-Soo Lee
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jinsu Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Tae-Su Han
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Moo-Seung Lee
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hyun Seung Ban
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
4
|
Li Y, Zhao M, Lin Y, Jiang X, Jin L, Ye P, Lu Y, Pei R, Jiang L. Licochalcone A induces mitochondria-dependent apoptosis and interacts with venetoclax in acute myeloid leukemia. Eur J Pharmacol 2024; 968:176418. [PMID: 38350590 DOI: 10.1016/j.ejphar.2024.176418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
The management of patients with acute myeloid leukemia (AML) remains a challenge because of the complexity and heterogeneity of this malignancy. Despite the recent approval of several novel targeted drugs, resistance seems inevitable, and clinical outcomes are still suboptimal. Increasing evidence supports the use of natural plants as an important source of anti-leukemic therapeutics. Licochalcone A (LCA) is an active flavonoid isolated from the roots of licorice, Glycyrrhiza uralensis Fisch., possessing extensive anti-tumor activities. However, its effects on AML and the underlying mechanisms remain unknown. Here, we showed that LCA decreased the viability of established human AML cell lines in a dose- and time-dependent manner. LCA significantly induced mitochondrial apoptotic cell death, accompanied by the downregulation of MCL-1, upregulation of BIM, truncation of BID, and cleavage of PARP. A prominent decline in the phosphorylation of multiple critical molecules, including AKT, glycogen synthase kinase-3β (GSK3β), ERK, and P38 was observed upon LCA treatment, indicating PI3K and MAPK signals were suppressed. Both transcription and translation of c-Myc were also inhibited by LCA. In addition, LCA enhanced the cytotoxicity of the BCL-2 inhibitor venetoclax. Furthermore, the anti-survival and pro-apoptotic effects were confirmed in primary blasts from 10 patients with de novo AML. Thus, our results expand the applications of LCA, which can be regarded as a valuable agent in treating AML.
Collapse
Affiliation(s)
- Youhong Li
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Mengting Zhao
- Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Ye Lin
- Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Xia Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Lili Jin
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Peipei Ye
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Ying Lu
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China
| | - Renzhi Pei
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Institute of Hematology, Ningbo University, Ningbo, China.
| | - Lei Jiang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, China; Department of Pathology and Pathogenic Biology, and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
5
|
Almatary AM, El Husseiny WM, Selim KB, Eisa HMH. Nitroimidazole derivatives potentiated against tumor hypoxia: Design, synthesis, antitumor activity, molecular docking study, and QSAR study. Drug Dev Res 2024; 85:e22126. [PMID: 37915124 DOI: 10.1002/ddr.22126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/01/2023] [Accepted: 10/15/2023] [Indexed: 11/03/2023]
Abstract
A hypoxic environment occurs predominantly in tumors. During the growth phase of a tumor, it grows until it exceeds its blood supply, leaving regions of the tumor in which the oxygen pressure is dramatically low. They are virtually absent in normal tissues, thus creating perfect conditions for selective bioreductive therapy of tumors. To this aim, a novel series of cytotoxic radiosensitizer agents were synthesized by linking the nitroimidazole scaffold with oxadiazole or triazole rings. The majority of the compounds exhibited moderate to excellent antiproliferative activities toward HCT116 cell line under normoxic and hypoxic conditions. The structure-activity relationship study revealed that compounds containing the free thiol group either in the oxadiazoles 11a,b or the triazoles 21a,b-23a,b demonstrated the strongest antiproliferative activity, which proves that the free thiol group plays a crucial role in the antiproliferative activity of our compounds under both normoxic (half-maximal inhibitory concentration [IC50 ] = 12.50-24.39 µM) and hypoxic conditions (IC50 = 4.69-11.56 µM). Radiosensitizing assay of the four most active cytotoxic compounds 11b and 21-23b assured the capability of the compounds to enhance the sensitivity of the tumor cells to the DNA damaging activity of γ-radiation (IC50 = 2.23-5.18 µM). To further investigate if the cytotoxicity of our most active compounds was due to a specific signaling pathway, the online software SwissTargetPrediction was exploited and a molecular docking study was done that proposed cyclin-dependent kinase 2 (CDK2) enzyme to be the most promising target. The CDK2 inhibitory assay assured this assumption as five out of six compounds demonstrated a comparable inhibitory activity with roscovitine, among which compound 21b showed threefold more potent inhibitory activity in comparison with the reference compound. A further biological evaluation proved compound 21b to have an apoptotic activity and cell cycle arrest activity at the G1 and S phases. During the AutoQSAR analysis, the model demonstrated excellent regression between the predicted and experimental activity with r2 = 0.86. Subsequently, we used the model to predict the activity of the test set compounds that came with r2 = 0.95.
Collapse
Affiliation(s)
- Aya M Almatary
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Walaa M El Husseiny
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Khalid B Selim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Hassan M H Eisa
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
6
|
Park W, Han JH, Wei S, Yang ES, Cheon SY, Bae SJ, Ryu D, Chung HS, Ha KT. Natural Product-Based Glycolysis Inhibitors as a Therapeutic Strategy for Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:807. [PMID: 38255882 PMCID: PMC10815680 DOI: 10.3390/ijms25020807] [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: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Targeted therapy against the epidermal growth factor receptor (EGFR) is a promising treatment approach for NSCLC. However, resistance to EGFR tyrosine kinase inhibitors (TKIs) remains a major challenge in its clinical management. EGFR mutation elevates the expression of hypoxia-inducible factor-1 alpha to upregulate the production of glycolytic enzymes, increasing glycolysis and tumor resistance. The inhibition of glycolysis can be a potential strategy for overcoming EGFR-TKI resistance and enhancing the effectiveness of EGFR-TKIs. In this review, we specifically explored the effectiveness of pyruvate dehydrogenase kinase inhibitors and lactate dehydrogenase A inhibitors in combating EGFR-TKI resistance. The aim was to summarize the effects of these natural products in preclinical NSCLC models to provide a comprehensive understanding of the potential therapeutic effects. The study findings suggest that natural products can be promising inhibitors of glycolytic enzymes for the treatment of EGFR-TKI-resistant NSCLC. Further investigations through preclinical and clinical studies are required to validate the efficacy of natural product-based glycolytic inhibitors as innovative therapeutic modalities for NSCLC.
Collapse
Affiliation(s)
- Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Shibo Wei
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| | - Sung-Jin Bae
- Department of Molecular Biology and Immunology, Kosin University College of Medicine, Busan 49267, Republic of Korea;
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea;
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan 50612, Republic of Korea; (E.-S.Y.); (S.-Y.C.)
| |
Collapse
|
7
|
Hba S, Ghaddar S, Wahnou H, Pinon A, El Kebbaj R, Pouget C, Sol V, Liagre B, Oudghiri M, Limami Y. Natural Chalcones and Derivatives in Colon Cancer: Pre-Clinical Challenges and the Promise of Chalcone-Based Nanoparticles. Pharmaceutics 2023; 15:2718. [PMID: 38140059 PMCID: PMC10748144 DOI: 10.3390/pharmaceutics15122718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Colon cancer poses a complex and substantial global health challenge, necessitating innovative therapeutic approaches. Chalcones, a versatile class of compounds with diverse pharmacological properties, have emerged as promising candidates for addressing colon cancer. Their ability to modulate pivotal signaling pathways in the development and progression of colon cancer makes them invaluable as targeted therapeutics. Nevertheless, it is crucial to recognize that although chalcones exhibit promise, further pre-clinical studies are required to validate their efficacy and safety. The journey toward effective colon cancer treatment is multifaceted, involving considerations such as optimizing the sequencing of therapeutic agents, comprehending the resistance mechanisms, and exploring combination therapies incorporating chalcones. Furthermore, the integration of nanoparticle-based drug delivery systems presents a novel avenue for enhancing the effectiveness of chalcones in colon cancer treatment. This review delves into the mechanisms of action of natural chalcones and some derivatives. It highlights the challenges associated with their use in pre-clinical studies, while also underscoring the advantages of employing chalcone-based nanoparticles for the treatment of colon cancer.
Collapse
Affiliation(s)
- Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693 Maarif, Casablanca 20100, Morocco; (S.H.); (H.W.); (M.O.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Suzan Ghaddar
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693 Maarif, Casablanca 20100, Morocco; (S.H.); (H.W.); (M.O.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Riad El Kebbaj
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| | - Christelle Pouget
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Vincent Sol
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (S.G.); (A.P.); (C.P.); (V.S.)
| | - Mounia Oudghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693 Maarif, Casablanca 20100, Morocco; (S.H.); (H.W.); (M.O.)
| | - Youness Limami
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693 Maarif, Casablanca 20100, Morocco; (S.H.); (H.W.); (M.O.)
- Laboratory of Health Sciences and Technologies, Higher Institute of Health Sciences, Hassan First University of Settat, Settat 26000, Morocco;
| |
Collapse
|
8
|
Wei F, Nian Q, Zhao M, Wen Y, Yang Y, Wang J, He Z, Chen X, Yin X, Wang J, Ma X, Chen Y, Feng P, Zeng J. Natural products and mitochondrial allies in colorectal cancer therapy. Biomed Pharmacother 2023; 167:115473. [PMID: 37713992 DOI: 10.1016/j.biopha.2023.115473] [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: 06/28/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023] Open
Abstract
Colorectal cancer (CRC) is a globally prevalent malignancy with a high potential for metastasis. Existing cancer treatments have limitations, including drug resistance and adverse effects. Researchers are striving to develop effective therapies to address these challenges. Impressively, contemporary research has discovered that many natural products derived from foods, plants, insects, and marine invertebrates can suppress the progression, metastasis, and invasion of CRC. In this review, we conducted a comprehensive search of the CNKI, PubMed, Embase, and Web of Science databases from inception to April 2023 to evaluate the efficacy of natural products targeting mitochondria to fight against CRC. Mitochondria are intracellular energy factories involved in cell differentiation, signal transduction, cell cycle regulation, apoptosis, and tumorigenesis. The identified natural products have been classified and summarized based on their mechanisms of action. These findings indicate that natural products can induce apoptosis in colorectal cancer cells by inhibiting the mitochondrial respiratory chain, ROS elevation, disruption of mitochondrial membrane potential, the release of pro-apoptotic factors, modulation of the Bcl-2 protein family to facilitate cytochrome c release, induction of apoptotic vesicle activity by activating the caspase protein family, and selective targeting of mitochondrial division. Furthermore, diverse apoptotic signaling pathways targeting mitochondria, such as the MAPK, p53, STAT3, JNK and AKT pathway, have been triggered by natural products. Natural products such as diosgenin, allopurinol, and clausenidin have demonstrated low toxicity, high efficacy, and multi-targeted properties. Mitochondria-targeting natural products have great potential for overcoming the challenges of CRC therapy.
Collapse
Affiliation(s)
- Feng Wei
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Qing Nian
- Department of Blood Transfusion, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Maoyuan Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Yang
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Jundong Wang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Zhelin He
- Endoscopy center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiaoyan Chen
- Endoscopy center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiang Yin
- Endoscopy center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Jian Wang
- Endoscopy center, Guang'an Hospital of Traditional Chinese Medicine, Guang'an 638000, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Peimin Feng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
9
|
Liu B, Qu X, Wang J, Xu L, Zhang L, Xu B, Su J, Bian X. LINC00365 functions as a tumor suppressor by inhibiting HIF-1α-mediated glucose metabolism reprogramming in breast cancer. Exp Cell Res 2023; 425:113514. [PMID: 36804531 DOI: 10.1016/j.yexcr.2023.113514] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Long non-coding RNAs (lncRNAs) play an important role in regulating several physiological processes and have been implicated in several pathologies including cancer. LncRNAs have been found to regulate key cellular pathways involved in cancer development, and their aberrant expression plays critical roles in the onset or progression of disease. The role of lncRNAs in breast cancer (BC) has become a hot topic of research in recent years. We previously showed that LINC00365 inhibits BC survival. In the current study, based on the important role of energy metabolism and HIF-1α for tumor cell proliferation, we investigated the role and mechanism of the LINC00365/HIF-1α axis in affecting tumor growth through glycolysis using the breast cancer cell lines MCF-7 and HCC-1937. We found that LINC00365 inhibited BC cell proliferation. Furthermore, LINC00365 overexpression suppressed aerobic glycolysis in BC cells. RNA-sequencing identified hypoxia-inducible factor-1α (HIF-1α), which has been linked with glycolysis and upregulates glycolysis-related genes, as a potential target gene of LINC00365. Accordingly, we found that LINC00365 overexpression resulted in decreased expression of key glycolytic enzymes such as downstream hexokinase 2 (HK2), recombinant pyruvate kinase isozymes M2 (PKM2) and lactate dehydrogenase A (LDHA). Our results suggest that targeting LINC00365 may reverse the glucose metabolism pattern of BC and effectively inhibit BC survival both in vitro and in vivo.
Collapse
Affiliation(s)
- Buhan Liu
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Labor-atory of Surgical Translational Medicine, Changchun, China; Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xianzhi Qu
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Jian Wang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Long Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lichao Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Bo Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuehai Bian
- Department of Thyroid Surgery, China-Japan Union Hospital of Jilin University, Jilin Provincial Key Labor-atory of Surgical Translational Medicine, Changchun, China.
| |
Collapse
|
10
|
Michalkova R, Kello M, Cizmarikova M, Bardelcikova A, Mirossay L, Mojzis J. Chalcones and Gastrointestinal Cancers: Experimental Evidence. Int J Mol Sci 2023; 24:ijms24065964. [PMID: 36983038 PMCID: PMC10059739 DOI: 10.3390/ijms24065964] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal (CRC) and gastric cancers (GC) are the most common digestive tract cancers with a high incidence rate worldwide. The current treatment including surgery, chemotherapy or radiotherapy has several limitations such as drug toxicity, cancer recurrence or drug resistance and thus it is a great challenge to discover an effective and safe therapy for CRC and GC. In the last decade, numerous phytochemicals and their synthetic analogs have attracted attention due to their anticancer effect and low organ toxicity. Chalcones, plant-derived polyphenols, received marked attention due to their biological activities as well as for relatively easy structural manipulation and synthesis of new chalcone derivatives. In this study, we discuss the mechanisms by which chalcones in both in vitro and in vivo conditions suppress cancer cell proliferation or cancer formation.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martina Cizmarikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Annamaria Bardelcikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
11
|
Deng N, Qiao M, Li Y, Liang F, Li J, Liu Y. Anticancer effects of licochalcones: A review of the mechanisms. Front Pharmacol 2023; 14:1074506. [PMID: 36755942 PMCID: PMC9900005 DOI: 10.3389/fphar.2023.1074506] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Cancer is a disease with a high fatality rate representing a serious threat to human health. Researchers have tried to identify effective anticancer drugs. Licorice is a widely used traditional Chinese medicine with various pharmacological properties, and licorice-derived flavonoids include licochalcones like licochalcone A, licochalcone B, licochalcone C, licochalcone D, licochalcone E, and licochalcone H. By regulating the expression in multiple signaling pathways such as the EGFR/ERK, PI3K/Akt/mTOR, p38/JNK, JAK2/STAT3, MEK/ERK, Wnt/β-catenin, and MKK4/JNK pathways, and their downstream proteins, licochalcones can activate the mitochondrial apoptosis pathway and death receptor pathway, promote autophagy-related protein expression, inhibit the expression of cell cycle proteins and angiogenesis factors, regulate autophagy and apoptosis, and inhibit the proliferation, migration, and invasion of cancer cells. Among the licochalcones, the largest number of studies examined licochalcone A, far more than other licochalcones. Licochalcone A not only has prominent anticancer effects but also can be used to inhibit the efflux of antineoplastic drugs from cancer cells. Moreover, derivatives of licochalcone A exhibit strong antitumor effects. Currently, most results of the anticancer effects of licochalcones are derived from cell experiments. Thus, more clinical studies are needed to confirm the antineoplastic effects of licochalcones.
Collapse
Affiliation(s)
- Nan Deng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingming Qiao
- Chongqing Institute for Food and Drug Control, Chongqing, China
| | - Ying Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fengyan Liang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanfeng Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Yanfeng Liu,
| |
Collapse
|
12
|
Zhao Y, Chard Dunmall LS, Cheng Z, Wang Y, Si L. Natural products targeting glycolysis in cancer. Front Pharmacol 2022; 13:1036502. [PMID: 36386122 PMCID: PMC9663463 DOI: 10.3389/fphar.2022.1036502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 09/05/2023] Open
Abstract
Many energy metabolism pathways exist in cancer, including glycolysis, amino acid metabolism, fatty acid oxidation, and mitochondrial respiration. Tumor cells mainly generate energy through glycolysis to maintain growth and biosynthesis of tumor cells under aerobic conditions. Natural products regulate many steps in glycolysis and targeting glycolysis using natural products is a promising approach to cancer treatment. In this review, we exemplify the relationship between glycolysis and tumors, demonstrate the natural products that have been discovered to target glycolysis for cancer treatment and clarify the mechanisms involved in their actions. Natural products, such as resveratrol mostly found in red grape skin, licochalcone A derived from root of Glycyrrhiza inflate, and brusatol found in Brucea javanica and Brucea mollis, largely derived from plant or animal material, can affect glycolysis pathways in cancer by targeting glycolytic enzymes and related proteins, oncogenes, and numerous glycolytic signal proteins. Knowledge of how natural products regulate aerobic glycolysis will help illuminate the mechanisms by which these products can be used as therapeutics to inhibit cancer cell growth and regulate cellular metabolism. Systematic Review Registration: https://pubmed.ncbi.nlm.nih.gov/, https://clinicaltrials.gov/, http://lib.zzu.edu.cn/.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lingling Si
- National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Phytochemicals as Regulators of Tumor Glycolysis and Hypoxia Signaling Pathways: Evidence from In Vitro Studies. Pharmaceuticals (Basel) 2022; 15:ph15070808. [PMID: 35890106 PMCID: PMC9315613 DOI: 10.3390/ph15070808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
The full understanding of the complex nature of cancer still faces many challenges, as cancers arise not as a result of a single target disruption but rather involving successive genetic and epigenetic alterations leading to multiple altered metabolic pathways. In this light, the need for a multitargeted, safe and effective therapy becomes essential. Substantial experimental evidence upholds the potential of plant-derived compounds to interfere in several important pathways, such as tumor glycolysis and the upstream regulating mechanisms of hypoxia. Herein, we present a comprehensive overview of the natural compounds which demonstrated, in vitro studies, an effective anticancer activity by affecting key regulators of the glycolytic pathway such as glucose transporters, hexokinases, phosphofructokinase, pyruvate kinase or lactate dehydrogenase. Moreover, we assessed how phytochemicals could interfere in HIF-1 synthesis, stabilization, accumulation, and transactivation, emphasizing PI3K/Akt/mTOR and MAPK/ERK pathways as important signaling cascades in HIF-1 activation. Special consideration was given to cell culture-based metabolomics as one of the most sensitive, accurate, and comprising approaches for understanding the response of cancer cell metabolome to phytochemicals.
Collapse
|
14
|
Effect of Licochalcone-A Combined with Rab23 Gene on Proliferation of Glioma U251 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9299442. [PMID: 35497928 PMCID: PMC9054455 DOI: 10.1155/2022/9299442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/04/2022] [Indexed: 11/18/2022]
Abstract
This research aimed to explore the effect of Licochalcone-A (LCA) combined with Rab23 gene on the proliferation, migration, and invasion of glioma U251 cells through the Wnt/β-catenin signaling pathway. The glioma U251 cell line was taken as the research object, and the Rab23 overexpression plasmid was constructed. According to the treatment method, U251 cells were rolled into blank control group (BC), Rab23 overexpression plasmid transfection group (Rab23), 25 μmol·L−1 LCA treatment group (LCA), and Rab23 overexpression plasmid transfection combined with 25 μmol·L−1 LCA treatment group (Rab23 + LCA). Subsequently, the ability of cell proliferation, migration, and invasion of each group was detected by methyl thiazolyl tetrazolium (MTT) assay, scratch healing test, and Transwell cell invasion test, respectively. Western blot was implemented to detect the expression differences of cell proliferation antigen Ki-67, apoptosis-related proteins Bcl-2 and Bax, and Wnt/β-catenin pathway-related proteins β-catenin, glycogen synthase kinase-3 (GSK3β), Axin2, and c-myc. The results showed the successful construction of Rab23 overexpression and stable transfection U251 cell line. After grouping and treatments, the cell proliferation, migration, and invasion ability of the Rab23 group, LCA group, and Rab23 + LCA group was substantially reduced relative to BC group (P < 0.05). In addition, the cell proliferation, migration, and invasion ability of Rab23 + LCA group decreased relatively more significantly. The expression levels of Ki-67, Bcl-2, β-catenin, and c-myc in the Rab23, LCA, and Rab23 + LCA groups were greatly lower versus those of BC group. Moreover, the protein expression levels of Bax, GSK3β, and Axin2 were considerably increased (P < 0.05), while the expression of protein in Rab23 + LCA group increased notably. These findings indicate that LCA combined with Rab23 gene can inhibit the proliferation, migration, and invasion of glioma U251 cells through the Wnt/β-catenin signaling and can promote cell apoptosis.
Collapse
|
15
|
Han TH, Park MK, Nakamura H, Ban HS. Capsaicin inhibits HIF-1α accumulation through suppression of mitochondrial respiration in lung cancer cells. Biomed Pharmacother 2021; 146:112500. [PMID: 34891118 DOI: 10.1016/j.biopha.2021.112500] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia inducible factor (HIF)-1α is an important transcription factor regulating cancer metabolism in hypoxic environment. Capsaicin is known to inhibit hypoxia-induced HIF activity in lung cancer. Hence, in this study we tried to elucidate its inhibitory mechanism of action. In lung cancer cells, including H1299, H23, A549, and H2009 cells, capsaicin inhibited cell growth and HIF activation. Under hypoxic conditions, capsaicin reduced the accumulation of HIF-1α protein and the expression of its target genes, including pyruvate dehydrogenase kinase 1 (PDK1) and glucose transporter 1 (GLUT1), with no effect on overall HIF-1α mRNA levels in the H1299 cells. In addition, capsaicin increased intracellular oxygen levels by suppressing mitochondrial respiration, resulting in a reduction of HIF-1α accumulation. Furthermore, mitochondrial ATP production was reduced by capsaicin through the inhibition of mitochondrial respiration in the H1299, H23, A549, and H2009 cells. These results indicate that capsaicin potentially exhibits anticancer therapeutic effects in lung cancer under hypoxic conditions.
Collapse
Affiliation(s)
- Tae-Hee Han
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Min Kyung Park
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea
| | - Hiroyuki Nakamura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan.
| | - Hyun Seung Ban
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, South Korea.
| |
Collapse
|
16
|
de Souza PS, Bibá GCC, Melo EDDN, Muzitano MF. Chalcones against the hallmarks of cancer: a mini-review. Nat Prod Res 2021; 36:4809-4826. [PMID: 34865580 DOI: 10.1080/14786419.2021.2000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Chalcones (1,3-diphenylpropen-1-ones) are a class of flavonoids that have been shown a broad spectrum of biological activities with therapeutic potential. Naturally occurring chalcones or synthetic chalcone derivatives have been extensively investigated as anticancer compounds. Cancer is still among the leading causes of death globally, although cancer treatments have improved over the past decades. Most of chemotherapeutic drugs target proliferating tumor cells; however, the cancer cells capabilities are also associated to tumor surround microenvironment. Thereby, the search of new compounds with a broad antitumor activity is still a great challenge. The cytotoxicity mechanisms of chalcones are beyond apoptosis induction in tumor cells, which make them promising compound for cancer therapy. In this mini-review we summarized recent studies that describe the anticancer potential of chalcones related to some of hallmarks of cancer. We shed a light on sustaining proliferative signaling, tumor-promoting inflammation, activating invasion and metastasis, inducing angiogenesis and resisting cell death.
Collapse
Affiliation(s)
- Paloma Silva de Souza
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Geysa Cristina Caldas Bibá
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Evelynn Dalila do Nascimento Melo
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Frazão Muzitano
- Laboratório de Produtos Bioativos, Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Seebacher NA, Krchniakova M, Stacy AE, Skoda J, Jansson PJ. Tumour Microenvironment Stress Promotes the Development of Drug Resistance. Antioxidants (Basel) 2021; 10:1801. [PMID: 34829672 PMCID: PMC8615091 DOI: 10.3390/antiox10111801] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Multi-drug resistance (MDR) is a leading cause of cancer-related death, and it continues to be a major barrier to cancer treatment. The tumour microenvironment (TME) has proven to play an essential role in not only cancer progression and metastasis, but also the development of resistance to chemotherapy. Despite the significant advances in the efficacy of anti-cancer therapies, the development of drug resistance remains a major impediment to therapeutic success. This review highlights the interplay between various factors within the TME that collectively initiate or propagate MDR. The key TME-mediated mechanisms of MDR regulation that will be discussed herein include (1) altered metabolic processing and the reactive oxygen species (ROS)-hypoxia inducible factor (HIF) axis; (2) changes in stromal cells; (3) increased cancer cell survival via autophagy and failure of apoptosis; (4) altered drug delivery, uptake, or efflux and (5) the induction of a cancer stem cell (CSC) phenotype. The review also discusses thought-provoking ideas that may assist in overcoming the TME-induced MDR. We conclude that stressors from the TME and exposure to chemotherapeutic agents are strongly linked to the development of MDR in cancer cells. Therefore, there remains a vast area for potential research to further elicit the interplay between factors existing both within and outside the TME. Elucidating the mechanisms within this network is essential for developing new therapeutic strategies that are less prone to failure due to the development of resistance in cancer cells.
Collapse
Affiliation(s)
| | - Maria Krchniakova
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Alexandra E. Stacy
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Patric J. Jansson
- Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia;
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St. Leonards, NSW 2065, Australia
| |
Collapse
|
18
|
Koklesova L, Liskova A, Samec M, Zhai K, AL-Ishaq RK, Bugos O, Šudomová M, Biringer K, Pec M, Adamkov M, Hassan STS, Saso L, Giordano FA, Büsselberg D, Kubatka P, Golubnitschaja O. Protective Effects of Flavonoids Against Mitochondriopathies and Associated Pathologies: Focus on the Predictive Approach and Personalized Prevention. Int J Mol Sci 2021; 22:ijms22168649. [PMID: 34445360 PMCID: PMC8395457 DOI: 10.3390/ijms22168649] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/07/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023] Open
Abstract
Multi-factorial mitochondrial damage exhibits a “vicious circle” that leads to a progression of mitochondrial dysfunction and multi-organ adverse effects. Mitochondrial impairments (mitochondriopathies) are associated with severe pathologies including but not restricted to cancers, cardiovascular diseases, and neurodegeneration. However, the type and level of cascading pathologies are highly individual. Consequently, patient stratification, risk assessment, and mitigating measures are instrumental for cost-effective individualized protection. Therefore, the paradigm shift from reactive to predictive, preventive, and personalized medicine (3PM) is unavoidable in advanced healthcare. Flavonoids demonstrate evident antioxidant and scavenging activity are of great therapeutic utility against mitochondrial damage and cascading pathologies. In the context of 3PM, this review focuses on preclinical and clinical research data evaluating the efficacy of flavonoids as a potent protector against mitochondriopathies and associated pathologies.
Collapse
Affiliation(s)
- Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
| | - Raghad Khalid AL-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
| | | | - Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 664 61 Rajhrad, Czech Republic;
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (L.K.); (A.L.); (M.S.); (K.B.)
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Prague, Czech Republic;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, 00185 Rome, Italy;
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (K.Z.); (R.K.A.-I.)
- Correspondence: (D.B.); (P.K.); (O.G.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1150 Brussels, Belgium
- Correspondence: (D.B.); (P.K.); (O.G.)
| | - Olga Golubnitschaja
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1150 Brussels, Belgium
- Predictive, Preventive, Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
- Correspondence: (D.B.); (P.K.); (O.G.)
| |
Collapse
|
19
|
Wang B, Wu Z, Li W, Liu G, Tang Y. Insights into the molecular mechanisms of Huangqi decoction on liver fibrosis via computational systems pharmacology approaches. Chin Med 2021; 16:59. [PMID: 34301291 PMCID: PMC8306236 DOI: 10.1186/s13020-021-00473-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/17/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The traditional Chinese medicine Huangqi decoction (HQD) consists of Radix Astragali and Radix Glycyrrhizae in a ratio of 6: 1, which has been used for the treatment of liver fibrosis. In this study, we tried to elucidate its action of mechanism (MoA) via a combination of metabolomics data, network pharmacology and molecular docking methods. METHODS Firstly, we collected prototype components and metabolic products after administration of HQD from a publication. With known and predicted targets, compound-target interactions were obtained. Then, the global compound-liver fibrosis target bipartite network and the HQD-liver fibrosis protein-protein interaction network were constructed, separately. KEGG pathway analysis was applied to further understand the mechanisms related to the target proteins of HQD. Additionally, molecular docking simulation was performed to determine the binding efficiency of compounds with targets. Finally, considering the concentrations of prototype compounds and metabolites of HQD, the critical compound-liver fibrosis target bipartite network was constructed. RESULTS 68 compounds including 17 prototype components and 51 metabolic products were collected. 540 compound-target interactions were obtained between the 68 compounds and 95 targets. Combining network analysis, molecular docking and concentration of compounds, our final results demonstrated that eight compounds (three prototype compounds and five metabolites) and eight targets (CDK1, MMP9, PPARD, PPARG, PTGS2, SERPINE1, TP53, and HIF1A) might contribute to the effects of HQD on liver fibrosis. These interactions would maintain the balance of ECM, reduce liver damage, inhibit hepatocyte apoptosis, and alleviate liver inflammation through five signaling pathways including p53, PPAR, HIF-1, IL-17, and TNF signaling pathway. CONCLUSIONS This study provides a new way to understand the MoA of HQD on liver fibrosis by considering the concentrations of components and metabolites, which might be a model for investigation of MoA of other Chinese herbs.
Collapse
Affiliation(s)
- Biting Wang
- Laboratory of Molecular Modeling and Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zengrui Wu
- Laboratory of Molecular Modeling and Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Weihua Li
- Laboratory of Molecular Modeling and Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Guixia Liu
- Laboratory of Molecular Modeling and Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yun Tang
- Laboratory of Molecular Modeling and Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
20
|
Synthetic methods and biological applications of retrochalcones isolated from the root of Glycyrrhiza species: A review. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|