1
|
Tang Y, Gao Z, Yang J, Li C, Wang W, Wu C, Wu M, Li M, Wu H, Sun Y, Zhang H, Chai Y, Xie F, Qian J, Shen H, Wang D. Breaking the synergism of iron overload and miR-122 to rescue lipid accumulation and peroxidation in MASLD. Pharmacol Res 2025; 215:107728. [PMID: 40188979 DOI: 10.1016/j.phrs.2025.107728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/01/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
MASLD is a multifactorial disease with specific subtypes being featured by hepatic iron overload and loss of miR-122, a liver-specific microRNA regulating hepatic lipid homeostasis. Previously we reported the mechanism of iron overload decreasing miR-122. Interestingly, we found that mice lacking miR-122 were highly sensitive to iron overload-induced steatosis and fibrosis. The present study aimed to disclose the downstream mechanisms and the preventive measures targeting miR-122. We first validated the decreases in iron-related genes and miR-122 in MASLD. By using LC-MS/MS and gas-chromatography, we found that the combination of miR-122 knockout and iron overload significantly increased the production and peroxidation of polyunsaturated fatty acids (PUFAs). However, miR-122 knockout itself only incurred lipid accumulation, suggesting a synergistic effect of miR-122 knockout and iron overload in lipid peroxidation. We then located the key enzymes involved in PUFA production and peroxidation by the transcriptome and proteome analysis. Mechanistically, miR-122 and iron regulated fatty acid synthesis through Aacs, fatty acid desaturation through Fads2, and PUFAs oxidation through CYPs. Re-supplementation of miR-122 by recombinant adeno-associated virus or agomir effectively broke the synergism of miR-122 knockout and iron overload in vivo. We further designed a miR-122 expression reporter cell model for high-throughput screening on 2543 natural compounds, and eventually found and validated that the dihydroberberine could upregulate miR-122 expression and correct iron overload-induced lipid disorders. These results identified the synergistic role of miR-122 and iron in PUFAs production and peroxidation, and also proposed the potential application of dihydroberberine as a preventive and therapeutic candidate for MASLD.
Collapse
Affiliation(s)
- Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China.
| | - Zelong Gao
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Chenqi Li
- Department of Nutrition, Second Military Medical University, Shanghai, China; Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Weili Wang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Chenghua Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengpu Wu
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Min Li
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Huiwen Wu
- Department of Clinical Nutrition, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Sun
- Department of Clinical Nutrition, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongwei Zhang
- Department of Clinical Nutrition, Brain Disease Hospital, Zhengzhou University, Henan, China
| | - Yifeng Chai
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Feng Xie
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China; Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Jianxin Qian
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China.
| | - Dongyao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China.
| |
Collapse
|
2
|
Teleb EK, Mehanna RA, Assem NM, Houssen ME. Antitumor effects of dauricine on sorafenib-treated human lung cancer cell lines via modulation of HIF-1α signaling pathways. Med Oncol 2025; 42:157. [PMID: 40205002 DOI: 10.1007/s12032-025-02679-4] [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: 12/23/2024] [Accepted: 03/11/2025] [Indexed: 04/11/2025]
Abstract
The majority of lung cancer cases are non-small cell lung cancer (NSCLC) which continues to be a serious global health concern. Hypoxia-inducible factor (HIF-α) pathway is a promising therapeutic target because it has a vital function in advanced non-small cell lung carcinoma. Antiangiogenic multi-kinase inhibitor, sorafenib may have a part in regulating HIF signaling in cancer. As a result, there is now more interest in employing it to target hypoxia-driven pathways in non-small cell lung cancer, especially when paired with natural bioactive products such as dauricine which is a naturally occurring alkaloid molecule targets multiple cellular pathways to provide strong anticancer effects. To examine molecular impacts of combining dauricine with sorafenib on HIF-mediated signaling pathways in human lung cancer cell lines. Cell viability was assessed using MTT assay in A549 and H1975 lung tumor cell lines. Levels of key proteins (AKT, mTORC1, HIF-1 α, ERK, VEGF, Cyclin-D1, BCL2, and E-Cadherin) were measured by ELISA.A colorimetric test was utilized to assess the activity of caspase-3, as a marker of apoptosis. qRT-PCR was employed to identify PI3K and VEGFR2 genes expression. Combination of sorafenib and dauricine significantly enhanced cytotoxicity compared to either agent alone. This combination also led to a marked reduction in VEGFR2, PI3K expression and VEGF, AKT, mTOR, HIF-1α, BCL2, ERK and E-Cadherin, and Cyclin-D1 levels. In addition, there was a significant increase in caspase-3 activity. Dauricine potentiates antitumor effects of sorafenib in human NSCLC by modulating HIF-1α-mediated pathways that are involved in several cancer hallmarks. This combination shows promise as a potential lung cancer treatment approach.
Collapse
Affiliation(s)
- Eman K Teleb
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, 22511, Egypt
| | - Radwa A Mehanna
- Medical Physiology, Faculty of Medicine, Alexanderia University, Alexanderia, Egypt
- Center of Excellence for Research in Regenerative Medicine and Its Applications, Faculty of Medicine, Alexandria University, Alexanderia, 22511, Egypt
| | - Nagwa M Assem
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexanderia, 22511, Egypt
| | - Maha E Houssen
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, 22511, Egypt.
| |
Collapse
|
3
|
Yang J, Wang D, Wang W, Wu C, Li C, Shi W, Qian J, Xie F, Shen H, Tang Y. Prolonged tamoxifen treatment induces iron deposition and ferroptosis in the liver. FASEB J 2025; 39:e70313. [PMID: 39792062 DOI: 10.1096/fj.202402553rr] [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: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025]
Abstract
Tamoxifen is an inhibitor of estrogen receptors and was originally developed for breast cancer therapy. Besides, tamoxifen is widely used for Cre-estrogen receptor-mediated conditional knockout in transgenic mice. However, we found that the 3-month feeding of 0.5 g/kg tamoxifen diet dramatically lowered the body weight of mice. The liver fat content and the serum lipid indicators were all decreased. But the liver injuries were identified, as illustrated by liver/body ratio and serum ALT and AST levels. The Sirius red staining and α-SMA staining even showed fibrosis in the liver. The increased lipid peroxidation indicators MDA and LPO, and ferroptosis markers COX-2, GPX4, SLC7A11, and ACSL4 implied the tamoxifen-induced ferroptosis in the liver. We further found that tamoxifen induced hepatic iron deposition. The investigation of iron transporters found that tamoxifen upregulated ferric iron reductase STEAP3, ferrous iron transporter DMT1, and iron storage molecule ferritin, which were probably the reasons for tamoxifen-induced iron deposition. The downregulation of the transferrin receptor and upregulation of hepcidin were more likely the responses to iron deposition. In conclusion, we found that tamoxifen disturbed the iron metabolism and induced liver injuries and ferroptosis, warranting attention to the applications of tamoxifen in cancer therapy and conditional gene knockout.
Collapse
Affiliation(s)
- Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Dongyao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- National Key Laboratory of Immunity and Inflammation, Second Military Medical University, Shanghai, China
| | - Weili Wang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chenghua Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenqi Li
- Department of Nutrition, Second Military Medical University, Shanghai, China
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Wenjing Shi
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jianxin Qian
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Xie
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China
- Key Laboratory of Biosafety Defense (Naval Medical University), Ministry of Education, Shanghai, China
| | - Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| |
Collapse
|
4
|
Zarychta J, Kowalczyk A, Słowik K, Przywara D, Petniak A, Kondracka A, Wójtowicz-Marzec M, Słyk-Gulewska P, Kwaśniewska A, Kocki J, Gil-Kulik P. Pilot Study on the Effect of Patient Condition and Clinical Parameters on Hypoxia-Induced Factor Expression: HIF1A, EPAS1 and HIF3A in Human Colostrum Cells. Int J Mol Sci 2024; 25:11042. [PMID: 39456823 PMCID: PMC11507067 DOI: 10.3390/ijms252011042] [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: 08/24/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1) may play a role in mammary gland development, milk production and secretion in mammals. Due to the limited number of scientific reports on the expression of HIF genes in colostrum cells, it was decided to examine the expression of HIF1A, HIF3A and EPAS1 in the these cells, collected from 35 patients who voluntarily agreed to provide their biological material for research, were informed about the purpose of the study and signed a consent to participate in it. The expression of HIF genes was assessed using qPCR. Additionally, the influence of clinical parameters (method of delivery, occurrence of stillbirths in previous pregnancies, BMI level before pregnancy and at the moment of delivery, presence of hypertension during pregnancy, presence of Escherichia coli in vaginal culture, iron supplement and heparin intake during pregnancy) on the gene expression was assessed, revealing statistically significant correlations. The expression of HIF1A was 3.5-fold higher in the case of patients with the presence of E. coli in vaginal culture (p = 0.041) and 2.5 times higher (p = 0.031) in samples from women who used heparin during pregnancy. Approximately 1.7-fold higher expression of the EPAS1 was observed in women who did not supplement iron during pregnancy (p = 0.046). To our knowledge, these are the first studies showing the relationship between HIF expression in cells from breast milk and the method of delivery and health condition of women giving birth. The assessment of HIF expression requires deeper examination in a larger study group, and the results of further studies will allow to determine whether HIF can become biomarkers in pregnancy pathology states.
Collapse
Affiliation(s)
- Julia Zarychta
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (J.Z.); (A.K.); (K.S.)
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Adrian Kowalczyk
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (J.Z.); (A.K.); (K.S.)
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Karolina Słowik
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (J.Z.); (A.K.); (K.S.)
| | - Dominika Przywara
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland;
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (A.P.); (J.K.)
| | - Alicja Petniak
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (A.P.); (J.K.)
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland; (A.K.); (P.S.-G.); (A.K.)
| | - Monika Wójtowicz-Marzec
- Chair and Department of Pediatric Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Patrycja Słyk-Gulewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland; (A.K.); (P.S.-G.); (A.K.)
| | - Anna Kwaśniewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland; (A.K.); (P.S.-G.); (A.K.)
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (A.P.); (J.K.)
| | - Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland; (A.P.); (J.K.)
| |
Collapse
|
5
|
Liang Y. Mechanisms of sorafenib resistance in hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2024; 48:102434. [PMID: 39084553 DOI: 10.1016/j.clinre.2024.102434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
Liver cancer is one of the most common and devastating causes of cancer-related deaths worldwide. Hepatocellular carcinoma (HCC) accounts for approximately 90% of primary liver cancers and represents a significant global health issue. There is currently no effective systemic treatment for patients with advanced liver cancer. One study suggests that sorafenib may be effective against hepatocellular carcinoma. Sorafenib can significantly extend the median survival time of patients, but only by 3-5 months. Furthermore, it is linked to severe adverse side effects and frequently leads to drug resistance. In this review, we offer a critical analysis of the factors contributing to sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Yuanjing Liang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China.
| |
Collapse
|
6
|
de Melo Silva AJ, de Melo Gama JE, de Oliveira SA. The Role of Bcl-2 Family Proteins and Sorafenib Resistance in Hepatocellular Carcinoma. Int J Cell Biol 2024; 2024:4972523. [PMID: 39188653 PMCID: PMC11347034 DOI: 10.1155/2024/4972523] [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: 04/13/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
Liver cancer has been reported to be one of the most malignant diseases in the world. It is late diagnosis consequently leads to a difficult treatment, as the cancer reached an advanced stage. Hepatocellular carcinoma (HCC) is the primary type of cancer diagnosed in the liver, with deadly characteristics and a poor prognosis. The first-in-line treatment for advanced HCC is sorafenib. Sorafenib acts by inhibiting cell proliferation and by inducing apoptosis as well as blocks receptors associated with these mechanisms. Due to its constant use, sorafenib resistance has been described, especially to proteins of the Bcl-2 family, and their overexpression of Bcl-XL and Mcl-1. This review focuses on the role of the Bcl-2 proteins in relation to sorafenib resistance as a consequence of first-in-line treatment in HCC.
Collapse
|
7
|
Hu X, Zhang P, Li S, Zhang J, Wang D, Wang Z, Zhu L, Wang L. Mitochondrial GCN5L1 acts as a novel regulator for iron homeostasis to promote sorafenib sensitivity in hepatocellular carcinoma. J Transl Med 2024; 22:593. [PMID: 38918793 PMCID: PMC11201091 DOI: 10.1186/s12967-024-05404-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Sorafenib resistance is becoming increasingly common and disadvantageous for hepatocellular carcinoma (HCC) treatment. Ferroptosis is an iron dependent programmed cell death underlying the mechanism of sorafenib. Iron is crucial for synthesis of cofactors essential to mitochondrial enzymes and necessary for HCC proliferation, while mitochondrial iron overload and oxidative stress are associated with sorafenib induced ferroptosis. However, the crosstalk among iron homeostasis and sorafenib resistance is unclear. METHODS We conducted bioinformatics analysis of sorafenib treated HCC datasets to analyze GCN5L1 and iron related gene expression with sorafenib resistance. GCN5L1 deleted HCC cell lines were generated by CRISPR technology. Sorafenib resistant HCC cell line was established to validate dataset analysis and evaluate the effect of potential target. RESULTS We identified GCN5L1, a regulator of mitochondrial acetylation, as a modulator in sorafenib-induced ferroptosis via affecting mitochondrial iron homeostasis. GCN5L1 deficiency significantly increased sorafenib sensitivity in HCC cells by down-regulating mitochondrial iron transporters CISD1 expression to induce iron accumulation. Mitochondrial iron accumulation leads to an acceleration in cellular and lipid ROS. Sorafenib resistance is related to CISD1 overexpression to release mitochondrial iron and maintaining mitochondrial homeostasis. We combined CISD1 inhibitor NL-1 with sorafenib, which significantly enhanced sorafenib-induced ferroptosis by promoting mitochondrial iron accumulation and lipid peroxidation. The combination of NL-1 with sorafenib enhanced sorafenib efficacy in vitro and in vivo. CONCLUSIONS Our findings demonstrate that GCN5L1/CISD1 axis is crucial for sorafenib resistance and would be a potential therapeutic strategy for sorafenib resistant HCC.
Collapse
Affiliation(s)
- Xiuya Hu
- State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Main Bldg., R1306 22 Qixiangtai Rd, Tianjin, 300070, China
| | - Peiyu Zhang
- State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Main Bldg., R1306 22 Qixiangtai Rd, Tianjin, 300070, China
| | - Sai Li
- The Province and Ministry co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaqi Zhang
- State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Main Bldg., R1306 22 Qixiangtai Rd, Tianjin, 300070, China
| | - Danni Wang
- The Province and Ministry co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zihan Wang
- The Province and Ministry co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lu Zhu
- State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Main Bldg., R1306 22 Qixiangtai Rd, Tianjin, 300070, China.
- The Province and Ministry co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.
| | - Lingdi Wang
- State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Main Bldg., R1306 22 Qixiangtai Rd, Tianjin, 300070, China.
| |
Collapse
|
8
|
Luan M, Feng Z, Zhu W, Xing Y, Ma X, Zhu J, Wang Y, Jia Y. Mechanism of metal ion-induced cell death in gastrointestinal cancer. Biomed Pharmacother 2024; 174:116574. [PMID: 38593706 DOI: 10.1016/j.biopha.2024.116574] [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: 01/17/2024] [Revised: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024] Open
Abstract
Gastrointestinal (GI) cancer is one of the most severe types of cancer, with a significant impact on human health worldwide. Due to the urgent demand for more effective therapeutic strategies against GI cancers, novel research on metal ions for treating GI cancers has attracted increasing attention. Currently, with accumulating research on the relationship between metal ions and cancer therapy, several metal ions have been discovered to induce cell death. In particular, the three novel modes of cell death, including ferroptosis, cuproptosis, and calcicoptosis, have become focal points of research in the field of cancer. Meanwhile, other metal ions have also been found to trigger cell death through various mechanisms. Accordingly, this review focuses on the mechanisms of metal ion-induced cell death in GI cancers, hoping to provide theoretical support for further GI cancer therapies.
Collapse
Affiliation(s)
- Muhua Luan
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Zhaotian Feng
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Wenshuai Zhu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Jingyu Zhu
- Department of Gastroenterology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yunshan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China
| | - Yanfei Jia
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan 250013, People's Republic of China; Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, People's Republic of China; Department of Medical Laboratory, Weifang Medical University, Weifang 261053, People's Republic of China.
| |
Collapse
|
9
|
Abdel-Wahab ND, Kabil MF, El-Sherbiny IM, Salama MF, El-Sayed G, El-Sherbini ES. Potential anticancer effect of free and nanoformulated Deferasirox for breast cancer treatment: in-vitro and in-vivo evaluation. Drug Dev Ind Pharm 2024; 50:223-235. [PMID: 38305197 DOI: 10.1080/03639045.2024.2314189] [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: 01/12/2024] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Breast cancer (BC) stands as the second-leading cause of mortality among women worldwide. Many chemotherapeutic treatments for BC come with significant adverse effects. Additionally, BC is recognized as one of the most resistant forms of malignancy to treatment. Consequently, there exists a critical need for innovative therapeutic agents that are both highly effective and exhibit reduced toxicity and side effects for patients. Deferasirox (DFX), an iron-chelating drug approved by the FDA for oral use, emerges as a promising contender in the fight against BC proliferation. DFX, primarily administered orally, is utilized to address chronic iron excess resulting from blood transfusions, and it is the inaugural treatment for chronic iron overload syndrome. However, DFX encounters limitations due to its poor water solubility. AIM This study aimed at incorporating DFX into lipid nanocapsules (DFX-LNCs) followed by investigating the anticancer effect of the DFX nanoform as compared to free DFX in-vitro and on an orthotopic BC mouse model in-vivo. METHODS The DFX-LNCs was prepared and imaged using TEM and also characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI) using DLS. Moreover, drug release, cytotoxicity, and anticancer effect were assessed in-vitro, and in-vivo. RESULTS The results revealed that DFX-LNCs are more cytotoxic than free DFX with IC50 of 4.417 µg/ml and 16.114 µg/ml, respectively, while the plain LNCs didn't show any cytotoxic effect on the 4T1 cell line (IC50 = 122.797 µg/ml). Besides, the apoptotic effect of DFX-LNCs was more pronounced than that of free DFX, as evidenced by Annexin V/PI staining, increased BAX expression, and decreased expression of BcL-2. Moreover, DFX-LNCs showed a superior antitumor effect in-vivo with potent antioxidant and anti-proliferative effects. CONCLUSION The newly developed DFX nanoform demonstrated a high potential as a promising therapeutic agent for BC treatment.
Collapse
Affiliation(s)
- Nadeen Diaa Abdel-Wahab
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed F Salama
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - Gehad El-Sayed
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| | - El-Said El-Sherbini
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Egypt
| |
Collapse
|
10
|
Wang Y, Yang W, Liu L, Liu L, Chen J, Duan L, Li Y, Li S. Vitamin K2 (MK-7) attenuates LPS-induced acute lung injury via inhibiting inflammation, apoptosis, and ferroptosis. PLoS One 2023; 18:e0294763. [PMID: 38011192 PMCID: PMC10681318 DOI: 10.1371/journal.pone.0294763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Acute lung injury (ALI) is a life-threatening disease that has received considerable critical attention in the field of intensive care. This study aimed to explore the role and mechanism of vitamin K2 (VK2) in ALI. Intraperitoneal injection of 7 mg/kg LPS was used to induce ALI in mice, and VK2 injection was intragastrically administered with the dose of 0.2 and 15 mg/kg. We found that VK2 improved the pulmonary pathology, reduced myeloperoxidase (MPO) activity and levels of TNF-α and IL-6, and boosted the level of IL-10 of mice with ALI. Moreover, VK2 played a significant part in apoptosis by downregulating and upregulating Caspase-3 and Bcl-2 expressions, respectively. As for further mechanism exploration, we found that VK2 inhibited P38 MAPK signaling. Our results also showed that VK2 inhibited ferroptosis, which manifested by reducing malondialdehyde (MDA) and iron levels, increasing glutathione (GSH) level, and upregulated and downregulated glutathione peroxidase 4 (GPX4) and heme oxygenase-1 (HO-1) expressions, respectively. In addition, VK2 also inhibited elastin degradation by reducing levels of uncarboxylated matrix Gla protein (uc-MGP) and desmosine (DES). Overall, VK2 robustly alleviated ALI by inhibiting LPS-induced inflammation, apoptosis, ferroptosis, and elastin degradation, making it a potential novel therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Yulian Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Weidong Yang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Lulu Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Lihong Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | | | - Lili Duan
- Sungen Bioscience Co., Ltd., Guangdong, China
| | - Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Shuzhuang Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|