1
|
Luo X, Li J, Cen Z, Feng G, Hong M, Huang L, Long Q. Exploring the therapeutic potential of lupeol: A review of its mechanisms, clinical applications, and advances in bioavailability enhancement. Food Chem Toxicol 2024; 196:115193. [PMID: 39662867 DOI: 10.1016/j.fct.2024.115193] [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: 07/18/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
Lupeol, a naturally occurring triterpenoid, has garnered significant attention for its diverse range of biological activities and potential therapeutic applications. This comprehensive review delves into the various aspects of lupeol, including its sources, extraction methods, chemical characteristics, pharmacokinetics, safety evaluation, mechanisms of action, and applications in disease treatment. We highlight the compound's unique carbon skeleton and its role in inflammation regulation, antioxidant activity, and broad-spectrum antimicrobial effects. The review also underscores lupeol's potential in cancer therapy, cardiovascular protection, metabolic disease management, and wound healing. Furthermore, we discuss the challenges and future perspectives of lupeol's clinical application, emphasizing the need for further research to improve its bioavailability and explore its full therapeutic potential. The review concludes by recognizing the significance of lupeol in drug development and healthcare, with expectations for future breakthroughs in medical applications.
Collapse
Affiliation(s)
- Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ji Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhifeng Cen
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Gang Feng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Meiqi Hong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Gajos-Michniewicz A, Czyz M. Therapeutic Potential of Natural Compounds to Modulate WNT/β-Catenin Signaling in Cancer: Current State of Art and Challenges. Int J Mol Sci 2024; 25:12804. [PMID: 39684513 DOI: 10.3390/ijms252312804] [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: 10/22/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Targeted therapies and immunotherapies have improved the clinical outcome of cancer patients; however, the efficacy of treatment remains frequently limited due to low predictability of response and development of drug resistance. Therefore, novel therapeutic strategies for various cancer types are needed. Current research emphasizes the potential therapeutic value of targeting WNT/β-catenin dependent signaling that is deregulated in various cancer types. Targeting the WNT/β-catenin signaling pathway with diverse synthetic and natural agents is the subject of a number of preclinical studies and clinical trials for cancer patients. The usage of nature-derived agents is attributed to their health benefits, reduced toxicity and side effects compared to synthetic agents. The review summarizes preclinical studies and ongoing clinical trials that aim to target components of the WNT/β-catenin pathway across a diverse spectrum of cancer types, highlighting their potential to improve cancer treatment.
Collapse
Affiliation(s)
- Anna Gajos-Michniewicz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| |
Collapse
|
3
|
Ren X, Zhang F, Gao G, Gao D, Su P. Biophysical and Morphological Cell Features Retrieved by Digital Holographic Microscopy Correlate with Drug-Induced Changes in Cell Migration Behavior. Anal Chem 2024. [PMID: 39513947 DOI: 10.1021/acs.analchem.4c04872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Quantitative analysis of cancer cell migration is critical for developing effective therapies to curb cancer metastasis. However, traditional methods are time-consuming and labor-intensive and lack quantitative capabilities. Cell volume change, a key physiological indicator of cell migration, is directly linked to phase change. In this work, we have developed a model that connects phase features from digital holographic microscopy (DHM) with cell healrate values from the wound healing assay. This approach aims to provide a rapid and quantitative evaluation of the breast cancer cell migration capability. Using DHM, six phase features of 231 cells treated with varying drug concentrations were extracted. It was observed that the rate of change of these phase features, termed characteristic parameters, showed a high linear correlation with cell healrate values from wound healing assays. Based on these linear correlations, a composite coefficient was derived by linearly combining the characteristic parameters of the six phase features. This composite coefficient was then linearly correlated with the cell healrate values to create a correlation model. This model establishes a strong connection between DHM-extracted morphological/biophysical features and cell migration metrics from a complementary assay. It provides a new, rapid, and quantitative method for assessing cancer cell migration in vitro and delivering valuable insights for cancer research.
Collapse
Affiliation(s)
- Xiaoxi Ren
- The State Key Laboratory of Chemical Oncogenomics and Open FIESTA, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055 Shenzhen, P. R. China
| | - Feiyang Zhang
- Chongqing Normal University, University Town Campus, 401331 Chongqing, P. R. China
| | - Ge Gao
- The State Key Laboratory of Chemical Oncogenomics and Open FIESTA, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055 Shenzhen, P. R. China
| | - Dan Gao
- The State Key Laboratory of Chemical Oncogenomics and Open FIESTA, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055 Shenzhen, P. R. China
| | - Ping Su
- The State Key Laboratory of Chemical Oncogenomics and Open FIESTA, Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055 Shenzhen, P. R. China
| |
Collapse
|
4
|
Chen Z, Luo R, Xu T, Wang L, Deng S, Wu J, Wang H, Lin Y, Bu M. Design, synthesis and antitumor effects of lupeol quaternary phosphonium salt derivatives. Bioorg Med Chem 2024; 113:117934. [PMID: 39369566 DOI: 10.1016/j.bmc.2024.117934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
Lupeol is a natural pentacyclic triterpenoid with a wide range of biological activities. To improve the water solubility and targeting of lupeol, in the following study, we synthesized 27 lupeol derivatives in the first series by introducing lipophilic cations with lupeol as the lead compound. Through the screening of different cancer cells, we found that some of the derivatives showed better activity than cisplatin against human non-small cell lung cancer A549 cells, among which compound 6c was found to have an IC50 value of 1.83 μM and a selectivity index of 21.02 (IC50MRC-5/IC50A549) against A549 cells. To further improve the antiproliferative activity of the compounds, we replaced the ester linkage of the linker with a carbamate linkage and synthesized a second series of five lupeol derivatives which were screened for activity, among which compound 14f was found to have an IC50 value of 1.36 μM and a selectivity index of 15.60 (IC50MRC-5/IC50A549) against A549 cells. We further evaluated the bioactivity of compounds 6c and 14f and found that both compounds induced apoptosis in A549 cells, promoted an increase in intracellular reactive oxygen species and decrease in mitochondrial membrane potential, and inhibited the cell cycle in the S phase. Of the compounds, compound 14f showed stronger bioactivity than compound 6c. We then selected compound 14f for molecular-level Western blot evaluation and in vivo evaluation in the zebrafish xenograft A549 tumor cell model. Compound 14f was found to significantly downregulate Bcl-2 protein expression and upregulate Bax, Cyt C, cleaved caspase-9, and cleaved caspase-3 protein expression, and 14f was found to be able to inhibit the proliferation of A549 cells in the zebrafish xenograft model. The above results suggest that compound 14f has great potential in the development of antitumor drugs targeting mitochondria.
Collapse
Affiliation(s)
- Zongxing Chen
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ran Luo
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Tianci Xu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jiale Wu
- College of Pharmacy, Hainan University, Haikou 570228, Hainan, PR China
| | - Haijun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| |
Collapse
|
5
|
Tanuja, Parani M. Identification of full-length genes involved in the biosynthesis of β-caryophyllene and lupeol from the leaf transcriptome of Ayapana triplinervis. Genome 2024; 67:440-444. [PMID: 38996388 DOI: 10.1139/gen-2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
β-Caryophyllene possesses potential anticancer properties against various cancers, including breast, colon, and lung cancer. Therefore, the essential oil of Ayapana triplinervis, which is rich in β-caryophyllene, can be a potential herbal remedy for treating cancer. However, molecular and genomic studies on A. triplinervis are still sparse. In this study, we obtained 14.7 Gb of RNA-Seq data from A. triplinervis leaf RNA and assembled 137 554 transcripts with an N50 value of 1437 bp. We annotated 72 436 (52.7%) transcripts and mapped 10 640 transcripts to 156 biochemical pathways. Among them, 218 were related to terpenoid backbone biosynthesis, while 27 were linked to sesquiterpenoid and triterpenoid pathways. Ninety-four transcripts were annotated in the β-caryophyllene and lupeol pathways. From these transcripts, for the first time, we identified 25 full-length genes encoding all the 17 enzymes involved in β-caryophyllene biosynthesis and an additional five genes involved in lupeol biosynthesis. These genes will be useful for the metabolic engineering of β-caryophyllene and lupeol biosynthesis, not just in A. triplinervis but also in other species.
Collapse
Affiliation(s)
- Tanuja
- Department of Genetic Engineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur 603203, Kanchipuram, Chennai, TN, India
| | - Madasamy Parani
- Department of Genetic Engineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur 603203, Kanchipuram, Chennai, TN, India
| |
Collapse
|
6
|
Sen K, Kumar Das S, Ghosh N, Sinha K, Sil PC. Lupeol: A dietary and medicinal triterpene with therapeutic potential. Biochem Pharmacol 2024; 229:116545. [PMID: 39293501 DOI: 10.1016/j.bcp.2024.116545] [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: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Lupeol, a triterpene derived from various plants, has emerged as a potent dietary supplement with extensive therapeutic potential. This review offers a comprehensive examination of lupeol's applications across diverse health conditions. By meticulously analyzing current scientific literature, we have synthesized findings that underscore lupeol's impact on cancer, diabetes, gastrointestinal disorders, neurological diseases, dermatological conditions, nephrological issues, and cardiovascular health. The review delves into molecular studies that reveal lupeol's ability to modulate disease pathways and alleviate symptoms, positioning it as a promising therapeutic agent. Moreover, we discuss the potential role of lupeol in clinical practice and public health strategies, emphasizing its substantial benefits as a natural compound. This thorough analysis serves as a critical resource for researchers, providing insights into the multifaceted therapeutic properties of lupeol and its potential to significantly enhance health outcomes.
Collapse
Affiliation(s)
- Koushik Sen
- Jhargram Raj College, Jhargram 721507, India
| | | | | | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India.
| |
Collapse
|
7
|
Deng S, Zhao Y, Guo X, Hong X, Li G, Wang Y, Li Q, Bu M, Wang M. Thiazolidinedione-Conjugated Lupeol Derivatives as Potent Anticancer Agents Through a Mitochondria-Mediated Apoptotic Pathway. Molecules 2024; 29:4957. [PMID: 39459325 PMCID: PMC11510666 DOI: 10.3390/molecules29204957] [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: 08/30/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
To improve the potential of lupeol against cancer cells, a privileged structure, thiazolidinedione, was introduced into its C-3 hydroxy group with ester, piperazine-carbamate, or ethylenediamine as a linker, and three series of thiazolidinedione-conjugated compounds (6a-i, 9a-i, and 12a-i) were prepared. The target compounds were evaluated for their cytotoxic activities against human lung cancer A549, human breast cancer MCF-7, human hepatocarcinoma HepG2, and human hepatic LO2 cell lines, and the results revealed that most of the compounds displayed improved potency over lupeol. Compound 12i exhibited significant activity against the HepG2 cell line, with an IC50 value of 4.40 μM, which is 9.9-fold more potent than lupeol (IC50 = 43.62 μM). Mechanistic studies suggested that 12i could induce HepG2 cell apoptosis, as evidenced by AO/EB staining and annexin V-FITC/propidium iodide dual staining assays. Western blot analysis suggested that compound 12i can upregulate Bax expression, downregulate Bcl-2 expression, and activate the mitochondria-mediated apoptotic pathway. Collectively, compound 12i is worthy of further investigation to support the discovery of effective agents against cancer.
Collapse
Affiliation(s)
- Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Yinxu Zhao
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Xiaoshan Guo
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Xian Hong
- Research Institute of Medicine & Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (X.H.); (G.L.)
| | - Gang Li
- Research Institute of Medicine & Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (X.H.); (G.L.)
| | - Yuchun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Qingyi Li
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| | - Ming Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (S.D.); (Y.Z.); (X.G.); (Y.W.); (Q.L.)
| |
Collapse
|
8
|
Fatma H, Jameel M, Siddiqui AJ, Kuddus M, Buali NS, Bahrini I, Siddique HR. Chemotherapeutic potential of lupeol against cancer in pre-clinical model: A systematic review and meta-analysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155777. [PMID: 38943695 DOI: 10.1016/j.phymed.2024.155777] [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: 03/25/2024] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Extensive research on Lupeol's potential in cancer prevention highlights its ability to target various cancer-related factors. It regulates proliferative markers, modulates signaling pathways, including PI3K/AKT/mTOR, and influences inflammatory and apoptotic mechanisms. Additionally, Lupeol demonstrates selectivity in killing cancer cells while sparing normal cells, thus minimizing the risk of toxic effects on healthy tissues. HYPOTHESIS Therefore, we aimed to explore Lupeol's potential roles as a chemotherapeutic agent and as a sensitizer to chemotherapy by reviewing various animal-based studies published on its effects. STUDY DESIGN We conducted a comprehensive search across databases, including PubMed, PMC, Cochrane, EuroPMC, and ctri.gov.in to identify pertinent articles. Our focus was solely on published animal studies examining Lupeol's anti-cancer effects, with reviewers independently assessing bias risk and resolving discrepancies through consensus. RESULT 20 studies were shortlisted. The results demonstrated that Lupeol brings changes in the tumor volume by [Hedges's g: -6.62; 95 % CI: -8.68, -4.56; τ2: 24.36, I2: 96.50 %; p < 0.05] and tumor weight by [Hedges's g: -3.97; 95 % CI: -5.20, -2.49; τ2: 2.70, I2: 79.27 %; p <0.05]. The high I2, negative Egger's value, and asymmetrical funnel plot show the publication bias among the studies. Further, Lupeol in combination with other chemotherapeutic agents showed better outcomes as compared to them alone [Hedges's g: -6.38; 95 % CI: -11.82, -0.94; τ2: 46.91; I2: 98.68 %; p <0.05]. Lupeol also targets various signaling molecules and pathways to exert an anti-cancer effect. CONCLUSION In conclusion, Lupeol significantly reduces tumor volume and weight. Combining Lupeol with other chemotherapy agents shows promise for enhancing anti-cancer effects. However, high variability among studies and evidence of publication bias suggest caution in interpreting results.
Collapse
Affiliation(s)
- Homa Fatma
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India
| | - Mohd Jameel
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia.
| | - Mohammed Kuddus
- Department of Biochemistry, College of Medicine, University of Ha'il, Hail, Kingdom of Saudi Arabia
| | - Nouha Saleh Buali
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia
| | - Insaf Bahrini
- Department of Biology, College of Science, University of Ha'il, Hail, PO Box 2440, Kingdom of Saudi Arabia
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Laboratory, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India.
| |
Collapse
|
9
|
Szoka L, Stocki M, Isidorov V. Dammarane-Type 3,4- seco-Triterpenoid from Silver Birch ( Betula pendula Roth) Buds Induces Melanoma Cell Death by Promotion of Apoptosis and Autophagy. Molecules 2024; 29:4091. [PMID: 39274939 PMCID: PMC11397366 DOI: 10.3390/molecules29174091] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
Despite unquestionable advances in therapy, melanoma is still characterized by a high mortality rate. For years, high expectations have been raised by compounds of natural origin as a component of pharmacotherapy, particularly by triterpenes found in the bark of birch trees. In this study, 3,4-seco-dammara-4(29),20(21),24(25)-trien-3-oic acid (SDT) was isolated from buds of silver birch and its mechanisms of cell death induction, including apoptosis and autophagy, were determined. Cytotoxicity of SDT was evaluated by the cell viability test and clonogenic assay, whereas induction of apoptosis and autophagy was determined by annexin V staining and Western blot. The results revealed dose- and time-dependent reductions in viability of melanoma cells. Treatment of cells for 48 h led to an increase in the percentage of annexin V-positive cells, activation of caspase-8, caspase-9, and caspase-3, and cleavage of PARP, confirming apoptosis. Simultaneously, it was found that SDT increased the level of autophagy marker LC3-II and initiator of autophagy beclin-1. Pretreatment of cells with caspase-3 inhibitor or autophagy inhibitor significantly reduced the cytotoxicity of SDT and revealed that both apoptosis and autophagy contribute to a decrease in cell viability. These findings suggest that 3,4-seco-dammaranes may become a promising group of natural compounds for searching for anti-melanoma agents.
Collapse
Affiliation(s)
- Lukasz Szoka
- Department of Medicinal Chemistry, Medical University of Bialystok, 15-222 Białystok, Poland
| | - Marcin Stocki
- Institute of Forest Sciences, Białystok University of Technology, 15-351 Białystok, Poland
| | - Valery Isidorov
- Institute of Forest Sciences, Białystok University of Technology, 15-351 Białystok, Poland
| |
Collapse
|
10
|
Mitra D, Saha D, Das G, Mukherjee R, Banerjee S, Alam N, Mustafi SM, Nath P, Majumder A, Majumder B, Murmu N. Lupeol synergizes with 5-fluorouracil to combat c-MET/EphA2 mediated chemoresistance in triple negative breast cancer. iScience 2023; 26:108395. [PMID: 38047085 PMCID: PMC10692664 DOI: 10.1016/j.isci.2023.108395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/02/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most elusive subtype of breast cancer that encounters treatment dilemmas owing to the paucity of druggable targets. We found hyperactivation of c-MET and ephrin type-A receptor 2 (EphA2) in patients treated with 5FU driven chemotherapy which correlated with lower disease-free survival. However, silencing of both these genes resulted in a marked decrease in the invasive, migratory, and tumorigenic potential of TNBC cells, indicating that a dual target strategy is actionable. Lupeol is a phytochemical, with potent anticancer efficacy and minimal side effects in preclinical studies. A synergistic strategy with 5FU and Lupeol elicited promising anticancer responses in vitro, in vivo, and in patient-derived ex vivo tumor culture models. This synergistic regimen is effective, even in the presence of HGF, which mechanistically orchestrates the activation of c-MET and EphA2. These data lay the foundation for the clinical validation of this combination therapy for TNBC patients.
Collapse
Affiliation(s)
- Debarpan Mitra
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Depanwita Saha
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Gaurav Das
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rimi Mukherjee
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Samir Banerjee
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Saunak Mitra Mustafi
- Department of Pathology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Partha Nath
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Anuj Majumder
- Department of Medicine, Harvard Medical School, 65 Lansdowne Street, Suite #317, Cambridge, MA 02139, USA
- Brookline High School, 115 Greenough Street, Brookline, MA 02445, USA
| | - Biswanath Majumder
- Departments of Molecular Profiling, Cancer Biology and Molecular Pathology, Mitra Biotech, Bangalore, India
| | - Nabendu Murmu
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| |
Collapse
|
11
|
Mani S, Ralph SJ, Swargiary G, Rani M, Wasnik S, Singh SP, Devi A. Therapeutic Targeting of Mitochondrial Plasticity and Redox Control to Overcome Cancer Chemoresistance. Antioxid Redox Signal 2023; 39:591-619. [PMID: 37470214 DOI: 10.1089/ars.2023.0379] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Significance: Mitochondria are subcellular organelles performing essential metabolic functions contributing to cellular bioenergetics and regulation of cell growth or death. The basic mitochondrial function in fulfilling the need for cell growth and vitality is evidenced whereby cancer cells with depleted mitochondrial DNA (rho zero, p0 cells) no longer form tumors until newly recruited mitochondria are internalized into the rho zero cells. Herein lies the absolute dependency on mitochondria for tumor growth. Hence, mitochondria are key regulators of cell death (by apoptosis, necroptosis, or other forms of cell death) and are, therefore, important targets for anticancer therapy. Recent Advances: Mitochondrial plasticity regulating their state of fusion or fission is key to the chemoresistance properties of cancer cells by promoting pro-survival pathways, enabling the mitochondria to mitigate against the cellular stresses and extreme conditions within the tumor microenvironment caused by chemotherapy, hypoxia, or oxidative stress. Critical Issues: This review discusses many characteristics of mitochondria, the processes and pathways controlling the dynamic changes occurring in the morphology of mitochondria, the roles of reactive oxygen species, and their relationship with mitochondrial fission or fusion. It also examines the relationship of redox to mitophagy when mitochondria become compromised and its effect on cancer cell survival, stemness, and the changes accompanying malignant progression from primary tumors to metastatic disease. Future Directions: A challenging question that arises is whether the changes in mitochondrial dynamics and their regulation can provide opportunities for improving drug targeting during cancer treatment and enhancing survival outcomes. Antioxid. Redox Signal. 39, 591-619.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Stephen J Ralph
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Australia
| | - Geeta Swargiary
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Madhu Rani
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Samiksha Wasnik
- Department of Regenerative Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - Shashi Prakash Singh
- Special Centre of Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Annu Devi
- Special Centre of Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
12
|
Hu J, Qi Q, Zhu Y, Wen C, Olatunji OJ, Jayeoye TJ, Eze FN. Unveiling the anticancer, antimicrobial, antioxidative properties, and UPLC-ESI-QTOF-MS/ GC–MS metabolite profile of the lipophilic extract of siam weed (Chromolaena odorata). ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
|
13
|
Inhibition of p62 and/or NFE2L2 induced autophagy impaires esophageal squamous cell cancer metastasis by reversing EMT. Gene 2023; 858:147194. [PMID: 36641074 DOI: 10.1016/j.gene.2023.147194] [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: 09/28/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) pathogenesis is influenced by both NFE2L2 (nuclear factor erythroid 2-related factor 2) and SQSTM1 (sequestosome 1), also known as p62. However, while there is evidence that these two proteins can interact with one another in a range of pathological contexts, whether these interactions govern the development or progression of ESCC remains unknown. In the present study, analyses of the GEPIA database revealed the simultaneous upregulation of both NFE2L2 and p62 in ESCC, as was further confirmed through biochemical analyses conducted with a human tumor microarray. Knocking down the expression of one or both of these factors demonstrated that both p62 and NFE2L2 mediate the progression of ESCC, as such downregulation altered the morphological characteristics of these cells and suppressed the epithelial-mesenchymal transition (EMT). Strikingly, these experiments revealed synergistic interactions between NFE2L2 and p62 in the promotion of ESCC invasivity and EMT induction. The treatment of cells with the autophagy inhibitors 3-MA, however, was sufficient to partially reverse the anti-metastatic effects of knocking down p62 and/or NFE2L2. Together, these data illustrate the ability of p62 and NFE2L2 to function in a synergistic manner, promoting ESCC cell metastatic progression and EMT induction through mechanisms linked to autophagic activity. As such, efforts to simultaneously target both of these proteins may represent a viable means of providing new treatment options to ESCC patients.
Collapse
|
14
|
Hashemi M, Arani HZ, Orouei S, Fallah S, Ghorbani A, Khaledabadi M, Kakavand A, Tavakolpournegari A, Saebfar H, Heidari H, Salimimoghadam S, Entezari M, Taheriazam A, Hushmandi K. EMT mechanism in breast cancer metastasis and drug resistance: Revisiting molecular interactions and biological functions. Biomed Pharmacother 2022; 155:113774. [DOI: 10.1016/j.biopha.2022.113774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
|
15
|
Ruellia tuberosa Ethyl Acetate Leaf Extract Induces Apoptosis and Cell Cycle Arrest in Human Breast Cancer Cell Line, MCF-7. Sci Pharm 2022. [DOI: 10.3390/scipharm90030044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Ruellia tuberosa L. has been previously shown to possess antioxidant and antiproliferative activities on cancer cells but its underlying mechanisms are largely unknown. This study aimed to elucidate the mode of action underlying this inhibitory effect on MCF-7 using ethyl acetate extract obtained after liquid-liquid partition of methanol crude extract. Antiproliferative effect of R. tuberosa ethyl acetate leaf extract (RTEAL) was evaluated using MTT assay. Its ability to induce apoptosis was assessed by DNA ladder formation, JC-1, Annexin V, and methylene blue staining assays. Perturbation of cell cycle progression was determined using flow cytometry. RTEAL was found to selectively inhibit the proliferation of MCF-7 cells with the IC50 value of 28 µg/mL. Morphological changes such as nuclear fragmentation and chromatin condensation were observed although DNA laddering was undetected in agarose gel. RTEAL-induced apoptotic pathways by inhibiting the expression of anti-apoptotic BCL-2 while upregulating pro-apoptotic BAX, caspase 7 and caspase 8. RTEAL also caused cell cycle arrests at the S and G2/M phase and dysregulation of cell cycle regulators. These findings collectively demonstrate that RTEAL extract inhibited cell growth by inducing apoptosis and cell cycle arrest, suggesting its therapeutic potential against breast cancer.
Collapse
|