1
|
Dong Y, Chang W, Lu B, Li Y, Liu Y. METTL5-mediated m 6A modification of SLC7A11 promotes cervical cancer by inhibiting ferroptosis. Int J Biochem Cell Biol 2025; 186:106822. [PMID: 40527406 DOI: 10.1016/j.biocel.2025.106822] [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: 11/30/2024] [Revised: 04/12/2025] [Accepted: 06/14/2025] [Indexed: 06/19/2025]
Abstract
Ferroptosis could suppress the viability of cervical cancer cells and trigger their death, thereby offering a unique perspective for exploring novel therapeutic approach for cervical cancer. Here, this study tried to explore the role of N6-methyladenosine (m6A) methyltransferase methyltransferase-like 5 (METTL5) on cervical cancer ferroptosis. Elevated METTL5 functioned as an oncogene in cervical cancer tumorigenesis by inhibiting the ferroptosis. Mechanistically, METTL5 was verified to target SLC7A11 and installed the m6A methylation on SLC7A11 mRNA. Moreover, YTHDF3 bound with the m6A site of SLC7A11 mRNA to enhance SLC7A11 mRNA stability. Rescue assays confirmed that METTL5/YTHDF3/SLC7A11 axis inhibited the ferroptosis of cervical cancer cells. In vivo, METTL5 silencing repressed the tumor growth of cervical cancer cells, as well as reducing the SLC7A11. In conclusion, these data inspired that METTL5-mediated m6A modification of SLC7A11 promoted cervical cancer by inhibiting ferroptosis, providing a novel insight for cervical cancer.
Collapse
Affiliation(s)
- Yujin Dong
- Department of Oncology, Zibo Central Hospital, Zibo 255036, China
| | - Wei Chang
- Department of Oncology, Zibo Central Hospital, Zibo 255036, China
| | - Bei Lu
- Department of Oncology, Zibo Central Hospital, Zibo 255036, China
| | - Yuanyuan Li
- Department of Oncology, Zibo Central Hospital, Zibo 255036, China.
| | - Yuanhua Liu
- Department of Oncology, Zibo Central Hospital, Zibo 255036, China.
| |
Collapse
|
2
|
Mahri S, Villa R, Shiau Y, Tang M, Racacho KJ, Zong Q, Chowdhury SI, Hua T, Godinez F, Birkeland A, Lin T, Li Y. Nanomedicine Approaches for Autophagy Modulation in Cancer Therapy. SMALL SCIENCE 2025; 5:2400607. [PMID: 40529859 PMCID: PMC12168626 DOI: 10.1002/smsc.202400607] [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: 12/02/2024] [Revised: 03/19/2025] [Indexed: 06/20/2025] Open
Abstract
Cancer is a daunting global health problem with a steadily rising incidence. Despite the wide arsenal of current anticancer therapies, challenges such as drug resistance, tumor heterogeneity, poor targeting, and severe side effects often lead to suboptimal efficacy and poor patient outcomes, highlighting the need for innovative therapies. Autophagy modulation has emerged as an attractive approach to complement existing therapies. The dual role of autophagy in cancer promotion and suppression has inspired the development of new drugs and therapeutic strategies focusing on both inhibition and induction. Despite the promising results of current autophagy modulators in preclinical studies, challenges such as the lack of selectivity and potency, toxicity, poor pharmacokinetics, and inadequate tumor targeting continue to limit their successful clinical translation. Many of these challenges could be overcome using nanomedicine. This review explores recent advancements in nanomedicine strategies for autophagy modulation. Successful combination strategies leveraging nanoparticles and autophagy modulators in synergy with chemotherapy, immunotherapy, phototherapy, gene therapy, and other modalities are presented. Additionally, nanomaterials with intrinsic autophagy-modulating capabilities, such as self-assembling autophagy inhibitors, are discussed. Finally, limitations of autophagy modulators currently in clinical trials are discussed, and future perspectives on designing nanomedicine for successful clinical implementation are explored.
Collapse
Affiliation(s)
- Sohaib Mahri
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Rodolfo Villa
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Ya‐Ping Shiau
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Menghuan Tang
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Kelsey Jane Racacho
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Qiufang Zong
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Saiful Islam Chowdhury
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
- Department of RadiologyUniversity of California DavisSacramentoCA95817USA
| | - Tan Hua
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
- Department of Otolaryngology, Head and Neck SurgeryUniversity of California DavisSacramentoCA95817USA
| | - Felipe Godinez
- Department of RadiologyUniversity of California DavisSacramentoCA95817USA
| | - Andrew Birkeland
- Department of Otolaryngology, Head and Neck SurgeryUniversity of California DavisSacramentoCA95817USA
| | - Tzu‐Yin Lin
- Division of Hematology/OncologyDepartment of Internal MedicineUniversity of California DavisSacramentoCA95817USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular MedicineUC Davis Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| |
Collapse
|
3
|
Chen C, Wang J, Zhu X, Zhang S, Yuan X, Hu J, Liu C, Liu L, Zhang Z, Li J. Lactylation as a metabolic epigenetic modification: Mechanistic insights and regulatory pathways from cells to organs and diseases. Metabolism 2025; 169:156289. [PMID: 40324589 DOI: 10.1016/j.metabol.2025.156289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/20/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
In recent years, lactylation, a novel post-translational modification, has demonstrated a unique role in bridging cellular metabolism and epigenetic regulation. This modification exerts a dual-edged effect in both cancer and non-cancer diseases by dynamically integrating the supply of metabolic substrates and the activity of modifying enzymes: on one hand, it promotes tissue homeostasis and repair through the activation of repair genes; on the other, it exacerbates pathological progression by driving malignant phenotypes. In the field of oncology, lactylation regulates key processes such as metabolic reprogramming, immune evasion, and therapeutic resistance, thereby shaping the heterogeneity of the tumor microenvironment. In non-cancerous diseases, including neurodegeneration and cardiovascular disorders, its aberrant activation can lead to mitochondrial dysfunction, fibrosis, and chronic inflammation. Existing studies have revealed a dynamic regulatory network formed by the cooperation of modifying and demodifying enzymes, and have identified mechanisms such as subcellular localization and RNA metabolism intervention that influence disease progression. Nevertheless, several challenges remain in the field. This article comprehensively summarizes the disease-specific regulatory mechanisms of lactylation, with the aim of providing a theoretical foundation for its targeted therapeutic application.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiandun Yuan
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100096, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhenpeng Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| |
Collapse
|
4
|
Liang Y, Lin W, Chen Y, Yang W, Zhou X, Ai S, Qiu L, Cao R, Wang J. Synthesis and in vitro and in vivo evaluation of novel bivalent quinolines as antitumor agents via targeting autophagy in cervical cancer. Eur J Med Chem 2025; 288:117421. [PMID: 39987834 DOI: 10.1016/j.ejmech.2025.117421] [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: 01/28/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
A series of novel bivalent quinolines with a spacer of four to six methylene units between the phenoxy group in the position-7 and various substituents in the position-4 of quinoline skeleton, respectively, were synthesized and evaluated as anticancer agents. The data showed that the majority of the compounds had significant antiproliferative activity with IC50 values less than 50 μM against human cancer cell lines. Among them, compound 4b exhibited the strongest antiproliferative activity against HCT116, A549, BGC823, HeLa and MCF-7 cell lines with an IC50 values of 0.26, 2.75, 4.06, 3.71 and 3.08 μM, respectively. Further studies on the anticancer effects in mice of compound 4b showed its capacity to inhibit tumor growth and markedly reduce tumor size of cervical cancer. Moreover investigation on the underlying mechanism of action indicated that compound 4b didn't trigger apoptotic processes in cervical cancer cell lines, but inhibit cervical cancer growth through inducing autophagy via the ATG5/ATG7 pathway.
Collapse
Affiliation(s)
- Yuexiu Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, 510275, PR China; Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China
| | - Wenxian Lin
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Yuzhen Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, 533000, PR China
| | - Weijie Yang
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Xiaoyu Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China; Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Baise, 533000, PR China
| | - Shishen Ai
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Liqin Qiu
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Rihui Cao
- School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China.
| | - Junli Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, 510275, PR China; Department of Obstetrics and Gynecology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, PR China.
| |
Collapse
|
5
|
Chen W, Zhou W, Liu S. The key role of natural products in the fight against endometrial Cancer. Int Immunopharmacol 2025; 151:114344. [PMID: 40015208 DOI: 10.1016/j.intimp.2025.114344] [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: 09/16/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Endometrial cancer (EC) is a common malignant disease in women, originating from the endometrial tissue. Over the past few decades, the global incidence rate of EC has gradually increased, and the affected population has become progressively younger. Traditional treatment methods, such as surgery and adjuvant therapy, have considerable toxic side effects. Furthermore, their therapeutic effectiveness is significantly very uncertain. Therefore, the search for a new type of treatment for EC is a top priority. Natural products are a class of compounds found in nature that have a wide range of biological functions; their derivatives have chemical structures that show great potential for developing new drugs. The latest studies have found that certain natural products, such as flavonoids, plant polyphenols, terpenoids and alkaloids, have inhibitory effects on EC cells in non-clinical models and animal studies. Despite challenges, including low extraction and bioavail ability, the potential of natural products for treating EC is still highly regarded by the scientific community. In the future, as research on natural products deepens and is combined with modern drug design and delivery technologies, it is hoped that more efficient and less toxic anti-cancer drugs will be developed, thereby offering EC patients more treatment options and hope. This article summarises the possible molecular mechanisms of various natural products and their bioactive components with regard to EC cells, as well as the latest research, to provide new ideas for further research and drug development.
Collapse
Affiliation(s)
- Wen Chen
- Tongde Hospital of Zhejiang Province Affiliated to Zhejiang Chinese Medical University (College of Integrated Traditional Chinese and Western Medicine Clinical Medicine), Hangzhou 310053, China
| | - Wencheng Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China
| | - Songjun Liu
- Tongde Hospital of Zhejiang Province Affiliated to Zhejiang Chinese Medical University (College of Integrated Traditional Chinese and Western Medicine Clinical Medicine), Hangzhou 310053, China; Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China.
| |
Collapse
|
6
|
Abdel Bar FM, Alonazi R, Elekhnawy E, Samra RM, Alqarni MH, Badreldin H, Magdy G. HPLC-PDA and in vivo anti-inflammatory potential of isorhamnetin-3-O-β-D-glucoside from Zygophyllum simplex L. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119089. [PMID: 39528120 DOI: 10.1016/j.jep.2024.119089] [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: 07/24/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammation is a biological process in response to injury, resulting in altered blood flow, increased vascular permeability, tissue destruction, and the production of reactive oxygen species (ROS) and inflammatory mediators. Zygophyllum simplex L., a medicinal plant traditionally used in the Arabian Peninsula for inflammatory disorders, has demonstrated promising in vitro anti-inflammatory activity due to its phenolic content. Additionally, the ethyl acetate fraction has exhibited notable in vivo anti-inflammatory effects. STUDY OBJECTIVE This research aimed to evaluate the in vivo anti-inflammatory effects of a Z. simplex plant extract and its principal ethyl acetate isolate, isorhamnetin-3-O-β-D-glucoside (Isor-3-Glu). The study seeks to develop a straightforward and robust HPLC method for quantifying Isor-3-Glu within the total methanolic extract of Z. simplex. MATERIALS AND METHODS The total methanol extract of Z. simplex was successively partitioned with a variety of organic solvents and the ethyl acetate fraction was used to isolate Isor-3-Glu on a Sephadex LH-20 column. The in vivo anti-inflammatory activity was investigated using carrageenan-triggered inflammation in rats. Histological features and immunohistochemical expression of cyclooxygenase-2 (COX-2) and tumor necrosis factor-alpha (TNF-α) were analyzed, and the levels of interleukins (IL-1β and IL-6) as well as prostaglandin E2 (PGE2) of the paw tissues were examined by qRT-PCR and ELISA, respectively. Quantification of Isor-3-Glu was achieved using an HPLC-PDA method. RESULTS Isor-3-Glu considerably (p < 0.05) lowered the weight of the paw edema. The histological abnormalities were improved, and the percentage of the COX-2 and TNF-α immunoreactive cells substantially decreased in the Isor-3-Glu-treated group in comparison with the positive control and Z. simplex extract group. Isor-3-Glu significantly ameliorated PGE2, IL-1β, and IL-6 levels. A straightforward and dependable HPLC technique was established for quantifying Isor-3-Glu in the total extract. The proposed methodology effectively determined Isor-3-Glu in less than 5 min. The calibration curve exhibited a linear relationship over the concentration range of 1.0-40.0 μg/mL, with a correlation coefficient (r) ≥ 0.9995. The developed method demonstrated a high level of sensitivity, with a detection limit as low as 0.139 μg/mL. The concentration of Isor-3-Glu in the total extract of Z. simplex was determined to be 0.05% w/w of dry extract. CONCLUSION Isor-3-Glu could be considered a promising anti-inflammatory compound that necessitates future clinical research. Isor-3-Glu was accurately quantified using a meticulously developed and optimized HPLC-PDA technique.
Collapse
Affiliation(s)
- Fatma M Abdel Bar
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Rana Alonazi
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia.
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Reham M Samra
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mohammed H Alqarni
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia.
| | - Hussein Badreldin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Galal Magdy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33511, Egypt; Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura National University, Gamasa, 7731168, Egypt.
| |
Collapse
|
7
|
Chen X, Cao Y, Ye K, Liu Y, Chi F, Niu Y, Lu D, Zhao B, Chen L, Zheng X, Feng W. Alkaloids with Their Protective Effects Against A β25-35-Induced PC-12 Cell Injury from the Tubers of Pinellia pedatisecta Schott. Molecules 2024; 29:5059. [PMID: 39519701 PMCID: PMC11547655 DOI: 10.3390/molecules29215059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Seven new alkaloids [1, (±)-2, (±)-3, 4, and 5] and one new natural product (6), along with eight known analogues, were isolated from the tubers of Pinellia pedatisecta Schott. Their structures were determined by a comprehensive analysis of spectroscopic data, including HRESIMS, and electronic circular dichroism (ECD). In addition, the results of the bioactivity evaluation showed that compounds (±)-3, 6, and 9 exhibited significantly protective effects against Aβ25-35-induced PC-12 cell injury and ameliorated cell viabilities by decreasing the levels of the reactive oxidative species (ROS) and mitochondrial membrane potential (MMP).
Collapse
Affiliation(s)
- Xu Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Yangang Cao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Kaili Ye
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Yanling Liu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Fangge Chi
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Ying Niu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Di Lu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Bingxian Zhao
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Lan Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
| | - Xiaoke Zheng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan and Education Ministry of P.R. China, Zhengzhou 450046, China
| | - Weisheng Feng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China; (X.C.); (Y.C.); (K.Y.); (Y.L.); (F.C.); (Y.N.); (D.L.); (B.Z.); (L.C.); (X.Z.)
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province China, Zhengzhou 450046, China
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Disease by Henan and Education Ministry of P.R. China, Zhengzhou 450046, China
| |
Collapse
|
8
|
Zhang L, Deng R, Guo R, Jiang Y, Guan Y, Chen C, Zhao W, Huang G, Liu L, Du H, Tang D. Recent progress of methods for cuproptosis detection. Front Mol Biosci 2024; 11:1460987. [PMID: 39297074 PMCID: PMC11408227 DOI: 10.3389/fmolb.2024.1460987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Varying from other identified cell death pathways, cuproptosis is a new type of regulated cell death characterized by excess Cu ions, abnormal aggregation of lipoylated proteins in TCA cycle, loss of Fe-S cluster proteins, upregulation of HSP70, leading to proteotoxic and oxidative stress. Cuproptosis is highly concerned by scientific community and as the field of cuproptosis further develops, remarkable progress has been made in the verification and mechanism of cuproptosis, and methods used to detect cuproptosis have been continuously improved. According to the characteristic changes of cuproptosis, techniques based on cell death verification, Cu content, morphology, molecular biology of protein levels of cuproptosis-related molecules and biochemical pathways of cuproptosis-related enzyme activity and metabolites of oxidative stress, lipoic acid, TCA cycle, Fe-S cluster proteins, oxidative phosphorylation, cell respiration intensity have been subject to cuproptosis verification and research. In order to further deepen the understanding of detecting cuproptosis, the principle and application of common cuproptosis detection methods are reviewed and categorized in cellular phenomena and molecular mechanism in terms of cell death, Cu content, morphology, molecular biology, biochemical pathways with a flow chart. All the indicating results have been displayed in response to the markers of cuproptosis, their advantages and limitations are summaried, and comparison of cuproptosis and ferroptosis detection is performed in this study. Our collection of methods for cuproptosis detection will provide a great basis for cuproptosis verification and research in the future.
Collapse
Affiliation(s)
- Ligang Zhang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Ruiting Deng
- Beijing Mercer United International Education Consulting Co., Ltd., Guangzhou, China
| | - Raoqing Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yawen Jiang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Yichen Guan
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Caiyue Chen
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Wudi Zhao
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Guobin Huang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Lian Liu
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dongsheng Tang
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, China
| |
Collapse
|
9
|
Zhong W, Zhang Q. Atractylodin: An Alkyne Compound with Anticancer Potential. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1729-1757. [PMID: 39192675 DOI: 10.1142/s0192415x24500551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Atractylodin is one of the main active ingredients of Atractylodis Rhizoma. It has various pharmacological properties, such as antigastric ulcer, immune regulation, antibacterial, anti-inflammatory, antitumor, anti-oxidant, and neuroprotective properties. In the past few decades, atractylodin has attracted the attention of researchers due to its excellent therapeutic effects. This paper aims to review the pharmacology of atractylodin, focusing mainly on its pharmacological effects in tumor treatment. Atractylodin exerts its antitumor effect by regulating different signaling pathways to induce important biological events such as apoptosis, cell cycle arrest, and autophagy, inhibiting cancer cell invasion and metastasis. In the process of cell apoptosis, atractylodin mainly induces cancer cell apoptosis by downregulating the Notch signaling pathway, affecting multiple upstream and downstream targets. In addition, atractylodin induces autophagy in cancer cells by regulating various signaling pathways such as PI3K/AKT/mTOR, p38MAPK, and hypothalamic Sirt1 and p-AMPK. Atractylodin effectively induces G1/M and G2/M phase arrest under the action of multiple signaling pathways. Among them, the pathways related to G1/M are more widely stagnated. In inhibiting the migration and invasion of cancer cells, atractylodin mainly regulates the Wnt signaling pathway, downregulates the expression of N-cadherin in cancer cells, and then blocks the PI3K/AKT/mTOR signaling pathway, inhibiting the phosphorylation of PI3K, AKT, and mTOR proteins, thereby having a significant impact on the invasion and migration of cancer cells. This paper systematically reviews the research progress on the antitumor effects and mechanisms of atractylodin, hoping to provide a reference and theoretical basis for its clinical application and new drug development.
Collapse
Affiliation(s)
- Wenxia Zhong
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Qi Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| |
Collapse
|
10
|
Liu C, Liu X, Ma Q, Su F, Cai E. Design, Synthesis, and Antitumor Activity of Isoliquiritigenin Amino Acid Ester Derivatives. Molecules 2024; 29:2641. [PMID: 38893517 PMCID: PMC11174122 DOI: 10.3390/molecules29112641] [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: 04/29/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Isoliquiritigenin (ISL) is a chalcone that has shown great potential in the treatment of cancer. However, its relatively weak activity and low water solubility limit its clinical application. In this study, we designed and synthesized 21 amino acid ester derivatives of ISL and characterized the compounds using 1H NMR and 13C NMR. Among them, compound 9 (IC50 = 14.36 μM) had a better inhibitory effect on human cervical cancer (Hela) than ISL (IC50 = 126.5 μM), and it was superior to the positive drug 5-FU (IC50 = 33.59 μM). The mechanism of the action experiment showed that compound 9 could induce Hela cell apoptosis and autophagy through the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
| | | | | | - Fengyan Su
- College of Chinese Medicinal Material, Jilin Agricultural University, 2888 Xincheng Street, Changchun 130118, China; (C.L.); (X.L.); (Q.M.)
| | - Enbo Cai
- College of Chinese Medicinal Material, Jilin Agricultural University, 2888 Xincheng Street, Changchun 130118, China; (C.L.); (X.L.); (Q.M.)
| |
Collapse
|
11
|
Meng Q, Sun H, Zhang Y, Yang X, Hao S, Liu B, Zhou H, Xu ZX, Wang Y. Lactylation stabilizes DCBLD1 activating the pentose phosphate pathway to promote cervical cancer progression. J Exp Clin Cancer Res 2024; 43:36. [PMID: 38291438 PMCID: PMC10829273 DOI: 10.1186/s13046-024-02943-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Discoidin, CUB, and LCCL domain-containing type I (DCBLD1) is identified as an oncogene involved in multiple regulation of tumor progression, but specific mechanisms remain unclear in cervical cancer. Lactate-mediated lactylation modulates protein function. Whether DCBLD1 can be modified by lactylation and the function of DCBLD1 lactylation are unknown. Therefore, this study aims to investigate the lactylation of DCBLD1 and identify its specific lactylation sites. Herein, we elucidated the mechanism by which lactylation modification stabilizes the DCBLD1 protein. Furthermore, we investigated DCBLD1 overexpression activating pentose phosphate pathway (PPP) to promote the progression of cervical cancer. METHODS DCBLD1 expression was examined in human cervical cancer cells and adjacent non-tumorous tissues using quantitative reverse transcription-polymerase chain reaction, western blotting, and immunohistochemistry. In vitro and in vivo studies were conducted to investigate the impact of DCBLD1 on the progression of cervical cancer. Untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics studies were used to characterize DCBLD1-induced metabolite alterations. Western blot, immunofuorescence and transmission electron microscopy were performed to detect DCBLD1 degradation of G6PD by activating autophagy. Chromatin immunoprecipitation, dual luciferase reporter assay for detecting the mechanism by which lactate increases DCBLD1 transcription. LC-MS/MS was employed to verify specific modification sites within the DCBLD1 protein. RESULTS We found that lactate increased DCBLD1 expression, activating the PPP to facilitate the proliferation and metastasis of cervical cancer cells. DCBLD1 primarily stimulated PPP by upregulating glucose-6-phosphate dehydrogenase (G6PD) expression and enzyme activity. The mechanism involved the increased enrichment of HIF-1α in the DCBLD1 promoter region, enhancing the DCBLD1 mRNA expression. Additionally, lactate-induced DCBLD1 lactylation stabilized DCBLD1 expression. We identified DCBLD1 as a lactylation substrate, with a predominant lactylation site at K172. DCBLD1 overexpression inhibited G6PD autophagic degradation, activating PPP to promote cervical cancer progression. In vivo, 6-An mediated inhibition of G6PD enzyme activity, inhibiting tumor proliferation. CONCLUSIONS Our findings revealed a novel post-translational modification type of DCBDL1, emphasizing the significance of lactylation-driven DCBDL1-mediated PPP in promoting the progression of cervical cancer.
Collapse
Affiliation(s)
- Qingfei Meng
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Huihui Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Xiangzhe Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Shiming Hao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China.
- School of Life Sciences, Henan University, Kaifeng, 475000, China.
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
12
|
Zhang S, Chen M, Geng Z, Liu T, Li S, Yu Q, Cao L, Liu D. Potential Application of Self-Assembled Peptides and Proteins in Breast Cancer and Cervical Cancer. Int J Mol Sci 2023; 24:17056. [PMID: 38069380 PMCID: PMC10706889 DOI: 10.3390/ijms242317056] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Ongoing research is gradually broadening the idea of cancer treatment, with attention being focused on nanoparticles to improve the stability, therapeutic efficacy, targeting, and other important metrics of conventional drugs and traditional drug delivery methods. Studies have demonstrated that drug delivery carriers based on biomaterials (e.g., protein nanoparticles and lipids) and inorganic materials (e.g., metal nanoparticles) have potential anticancer effects. Among these carriers, self-assembled proteins and peptides, which are highly biocompatible and easy to standardize and produce, are strong candidates for the preparation of anticancer drugs. Breast cancer (BC) and cervical cancer (CC) are two of the most common and deadly cancers in women. These cancers not only threaten lives globally but also put a heavy burden on the healthcare system. Despite advances in medical care, the incidence of these two cancers, particularly CC, which is almost entirely preventable, continues to rise, and the mortality rate remains steady. Therefore, there is still a need for in-depth research on these two cancers to develop more targeted, efficacious, and safe therapies. This paper reviews the types of self-assembling proteins and peptides (e.g., ferritin, albumin, and virus-like particles) and natural products (e.g., soy and paclitaxel) commonly used in the treatment of BC and CC and describes the types of drugs that can be delivered using self-assembling proteins and peptides as carriers (e.g., siRNAs, DNA, plasmids, and mRNAs). The mechanisms (including self-assembly) by which the natural products act on CC and BC are discussed. The mechanism of action of natural products on CC and BC and the mechanism of action of self-assembled proteins and peptides have many similarities (e.g., NF-KB and Wnt). Thus, natural products using self-assembled proteins and peptides as carriers show potential for the treatment of BC and CC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lingling Cao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (S.Z.); (M.C.); (Z.G.); (T.L.); (S.L.); (Q.Y.)
| |
Collapse
|