1
|
Wang M, An Q, Li Z, Huang Z, Huang K, Li G, Ma Q, Zhao L. The alkylglycerone phosphate synthase sustains the resistance of gastric cancer cells to ferroptosis induced by Apatinib. Gastric Cancer 2025:10.1007/s10120-025-01610-0. [PMID: 40186794 DOI: 10.1007/s10120-025-01610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Apatinib is a targeted therapy used in the treatment of advanced gastric cancer. However, many gastric cancer patients develop resistance to Apatinib, and the mechanisms underlying this resistance remain unclear. Previous studies have shown that Apatinib can induce ferroptosis in gastric cancer cells. More recent research suggests that polyunsaturated ether phospholipids are closely associated with tumor cell sensitivity to ferroptosis, and may represent key molecules involved in the resistance of tumor cells to ferroptosis. METHODS We established Apatinib-resistant gastric cancer cell lines and assessed their tolerance to ferroptosis. We identified key enzymes responsible for the ferroptosis tolerance observed in drug-resistant cells using lipidomics and transcriptomics analysis. Molecular and biological experiments were conducted to elucidate the molecular mechanisms underlying Apatinib resistance mediated by ferroptosis tolerance in gastric cancer cells. RESULTS Apatinib resistance is closely linked to ferroptosis resistance, which is driven by a reduction in the levels of polyunsaturated ether phospholipids-phospholipids that are particularly susceptible to oxidation and induce ferroptosis. The downregulation of key enzymes involved in polyunsaturated ether phospholipid synthesis, such as AGPS, mediates tolerance to both ferroptosis and Apatinib in gastric cancer cells, both in vitro and in vivo. Mechanistically, the expression of AGPS in tumor cells is regulated by the transcription factor ELK1. Drug-resistant cells acquire Apatinib tolerance by downregulating both ELK1 and AGPS expression. CONCLUSIONS Apatinib-resistant gastric cancer cells exhibit reduced expression of the transcription factor ELK1, which regulates the expression of AGPS. This reduction contributes to the resistance and malignancy of gastric cancer cells.
Collapse
Affiliation(s)
- Minghao Wang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiyuan An
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Zhiwei Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhicheng Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kaihua Huang
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Guoxin Li
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qiang Ma
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, China
| | - Liying Zhao
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [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: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
3
|
Xu X, Wang Y, Wang Q, LV S, Mao G. Elevated expression of ELK1 promotes breast cancer cell growth and correlates with poor prognosis of breast cancer patients. Ann Med Surg (Lond) 2024; 86:5767-5775. [PMID: 39359848 PMCID: PMC11444635 DOI: 10.1097/ms9.0000000000002256] [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: 04/15/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024] Open
Abstract
Background Breast cancer is the most common tumor in women and poses a serious threat to women's physical and mental health. The ETS-like gene 1 (ELK1), upregulated in various malignancies, serves as a transcription regulatory factor. This study primarily investigates the biological functions and prognostic significance of ELK1 in breast cancer. Materials and methods The authors conducted an analysis of ELK1 expression in breast cancer and adjacent tissues using data from The Cancer Genome Atlas (TCGA), and validated these findings with clinical specimens. Additionally, the authors employed siRNA transfection, proliferation and apoptosis assays to elucidate the roles of ELK1 in breast cancer cells. Furthermore, we assessed the correlations between ELK1 expression and the tumor microenvironment, as well as tumor-infiltrating immune cells (TIICs), utilizing the ESTIMATE and CIBERSORT algorithms. Finally, we used Kaplan-Meier plots and COX regressions to identify prognostic factors, and developed a predictive alignment diagram to evaluate the prognostic significance of ELK1 in breast cancer. Results A marked increase in ELK1 expression is evident in breast cancer tissues (P<0.01). Experimental findings demonstrate that silencing ELK1 suppresses proliferation and promotes apoptosis in breast cancer cells. ELK1 plays a pivotal role in regulating the immune microenvironment of breast cancer. Furthermore, the alignment diagram indicates that ELK1 may serve as an independent prognostic factor for breast cancer patients. Conclusion The authors' study reveals that ELK1 exhibits a high expression level in breast cancer tissues and is associated with disease progression and poor prognosis.
Collapse
Affiliation(s)
- Xiuping Xu
- Medical Laboratory, Shaoxing People’s Hospital
| | - Yanan Wang
- Medical Laboratory, Shaoxing University Affiliated Hospital, Shaoxing, Zhejiang, China
| | - Qing Wang
- Medical Laboratory, Shaoxing People’s Hospital
| | - Shanmei LV
- Medical Laboratory, Shaoxing People’s Hospital
| | - Guofeng Mao
- Medical Laboratory, Shaoxing People’s Hospital
| |
Collapse
|
4
|
Zhang Y, Zhou Y, Li X, Pan X, Bai J, Chen Y, Lai Z, Chen Q, Ma F, Dong Y. Small-molecule α-lipoic acid targets ELK1 to balance human neutrophil and erythrocyte differentiation. Stem Cell Res Ther 2024; 15:100. [PMID: 38589882 PMCID: PMC11003016 DOI: 10.1186/s13287-024-03711-6] [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: 10/30/2022] [Accepted: 03/31/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Erythroid and myeloid differentiation disorders are commonly occurred in leukemia. Given that the relationship between erythroid and myeloid lineages is still unclear. To find the co-regulators in erythroid and myeloid differentiation might help to find new target for therapy of myeloid leukemia. In hematopoiesis, ALA (alpha lipoic acid) is reported to inhibit neutrophil lineage determination by targeting transcription factor ELK1 in granulocyte-monocyte progenitors via splicing factor SF3B1. However, further exploration is needed to determine whether ELK1 is a common regulatory factor for erythroid and myeloid differentiation. METHODS In vitro culture of isolated CD34+, CMPs (common myeloid progenitors) and CD34+ CD371- HSPCs (hematopoietic stem progenitor cells) were performed to assay the differentiation potential of monocytes, neutrophils, and erythrocytes. Overexpression lentivirus of long isoform (L-ELK1) or the short isoform (S-ELK1) of ELK1 transduced CD34+ HSPCs were transplanted into NSG mice to assay the human lymphocyte and myeloid differentiation differences 3 months after transplantation. Knocking down of SRSF11, which was high expressed in CD371+GMPs (granulocyte-monocyte progenitors), upregulated by ALA and binding to ELK1-RNA splicing site, was performed to analyze the function in erythroid differentiation derived from CD34+ CD123mid CD38+ CD371- HPCs (hematopoietic progenitor cells). RNA sequencing of L-ELK1 and S-ELK1 overexpressed CD34+ CD123mid CD38+ CD371- HPCs were performed to assay the signals changed by ELK1. RESULTS Here, we presented new evidence that ALA promoted erythroid differentiation by targeting the transcription factor ELK1 in CD34+ CD371- hematopoietic stem progenitor cells (HSPCs). Overexpression of either the long isoform (L-ELK1) or the short isoform (S-ELK1) of ELK1 inhibited erythroid-cell differentiation, but knockdown of ELK1 did not affect erythroid-cell differentiation. RNAseq analysis of CD34+ CD123mid CD38+ CD371- HPCs showed that L-ELK1 upregulated the expression of genes related to neutrophil activity, phosphorylation, and hypoxia signals, while S-ELK1 mainly regulated hypoxia-related signals. However, most of the genes that were upregulated by L-ELK1 were only moderately upregulated by S-ELK1, which might be due to a lack of serum response factor interaction and regulation domains in S-ELK1 compared to L-ELK1. In summary, the differentiation of neutrophils and erythrocytes might need to rely on the dose of L-ELK1 and S-ELK1 to achieve precise regulation via RNA splicing signals at early lineage commitment. CONCLUSIONS ALA and ELK1 are found to regulate both human granulopoiesis and erythropoiesis via RNA spliceosome, and ALA-ELK1 signal might be the target of human leukemia therapy.
Collapse
Affiliation(s)
- Yimeng Zhang
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Xindu Road 783, Chengdu, 610500, China
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Ya Zhou
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xiaohong Li
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xu Pan
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Ju Bai
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yijin Chen
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | | | - Qiang Chen
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Feng Ma
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China.
| | - Yong Dong
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Xindu Road 783, Chengdu, 610500, China.
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China.
| |
Collapse
|
5
|
Lund C, Ranea-Robles P, Falk S, Rausch DM, Skovbjerg G, Vibe-Petersen VK, Krauth N, Skytte JL, Vana V, Roostalu U, Pers TH, Lund J, Clemmensen C. Protection against overfeeding-induced weight gain is preserved in obesity but does not require FGF21 or MC4R. Nat Commun 2024; 15:1192. [PMID: 38331907 PMCID: PMC10853283 DOI: 10.1038/s41467-024-45223-0] [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: 05/02/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Overfeeding triggers homeostatic compensatory mechanisms that counteract weight gain. Here, we show that both lean and diet-induced obese (DIO) male mice exhibit a potent and prolonged inhibition of voluntary food intake following overfeeding-induced weight gain. We reveal that FGF21 is dispensable for this defense against weight gain. Targeted proteomics unveiled novel circulating factors linked to overfeeding, including the protease legumain (LGMN). Administration of recombinant LGMN lowers body weight and food intake in DIO mice. The protection against weight gain is also associated with reduced vascularization in the hypothalamus and sustained reductions in the expression of the orexigenic neuropeptide genes, Npy and Agrp, suggesting a role for hypothalamic signaling in this homeostatic recovery from overfeeding. Overfeeding of melanocortin 4 receptor (MC4R) KO mice shows that these mice can suppress voluntary food intake and counteract the enforced weight gain, although their rate of weight recovery is impaired. Collectively, these findings demonstrate that the defense against overfeeding-induced weight gain remains intact in obesity and involves mechanisms independent of both FGF21 and MC4R.
Collapse
Affiliation(s)
- Camilla Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Falk
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Dylan M Rausch
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Grethe Skovbjerg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Gubra ApS, Hørsholm, Denmark
| | | | - Nathalie Krauth
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Vasiliki Vana
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Chai B, Li Y, Guo Y, Zhang Z, Jia K, Chai X, Suo Y. ETV7 promotes colorectal cancer progression through upregulation of IFIT3. Funct Integr Genomics 2024; 24:8. [PMID: 38200280 PMCID: PMC10781848 DOI: 10.1007/s10142-023-01282-y] [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: 10/22/2023] [Revised: 12/04/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024]
Abstract
Members of the E26 transformation-specific (ETS) variant transcription factor family act as either tumor suppressors or oncogenic factors in numerous types of cancer. ETS variant transcription factor 7 (ETV7) participates in the development of malignant tumors, whereas its involvement in colorectal cancer (CRC) is less clear. In this study, The Cancer Genome Atlas (TCGA) and immunochemistry staining were applied to check the clinical relevance of ETV7 and interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in CRC patients. Overexpression and knockdown of ETV7 and IFIT3 were conducted by transfecting the cells with pCDNA3.1 plasmids and siRNAs, respectively. Western blotting was used to detect the protein expression of ETV7 in CRC cells. Cell Counting Kit-8, cell colony formation, and Transwell assays, as well as flow cytometry, were used to evaluate the proliferation, migration, cell cycle, and apoptosis of CRC cells. Furthermore, western blotting, RT-qPCR, and luciferase assay were used to explore the regulation of ETV7 on IFIT3. Rescue assay was used to investigate the significance of ETV7/IFIT3 axis on CRC progression. We found that ETV7 was upregulated in CRC tissues and cells. Overexpression of ETV7 stimulated the proliferation, migration, and cell cycle amplification, and reduced the apoptosis of CRC cells. Downregulation of ETV7 exerted the opposite effect on CRC cell progression. Moreover, we demonstrated that ETV7 stimulated the transcription activity, the mRNA and protein expression of IFIT3 in CRC cells. There was a positive correlation between ETV7 and IFIT3 in CRC patients. IFIT3 knockdown reversed the promotive effect exerted by overexpression of ETV7 on the amplification and migration of CRC cells. By contrast, overexpression of IFIT3 blocked the inhibitory effect of ETV7-targeting siRNA. In summary, ETV7 induces progression of CRC by activating the transcriptional expression of IFIT3. The EVT7/IFIT3 axis may be a novel target for CRC therapy.
Collapse
Affiliation(s)
- Bao Chai
- Department of Gastroenterology, Shanxi Academy of Medical Science, Shanxi Bethune Hospital, Taiyuan, China
| | - Yanjun Li
- Department of Surgery, Shanxi Academy of Medical Science, Shanxi Bethune Hospital, Taiyuan, China
| | - Yarong Guo
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, 85 South Jiefang Road, TaiyuanTaiyuan, 030001, Shanxi Province, China.
| | - Zhuowei Zhang
- Medical Imaging Department, Shanxi Medical University, Taiyuan, China
| | - Kai Jia
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinhao Chai
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, 85 South Jiefang Road, TaiyuanTaiyuan, 030001, Shanxi Province, China
| | - Yuhong Suo
- Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Taiyuan, China
| |
Collapse
|
7
|
Ahmed SA, Mendonca P, Messeha SS, Oriaku ET, Soliman KFA. The Anticancer Effects of Marine Carotenoid Fucoxanthin through Phosphatidylinositol 3-Kinase (PI3K)-AKT Signaling on Triple-Negative Breast Cancer Cells. Molecules 2023; 29:61. [PMID: 38202644 PMCID: PMC10779870 DOI: 10.3390/molecules29010061] [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: 11/19/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks specific targets such as estrogen, progesterone, and HER2 receptors. TNBC affects one in eight women in the United States, making up 15-20% of breast cancer cases. Patients with TNBC can develop resistance to chemotherapy over time, leading to treatment failure. Therefore, finding other options like natural products is necessary for treatment. The advantages of using natural products sourced from plants as anticancer agents are that they are less toxic, more affordable, and have fewer side effects. These products can modulate several cellular processes of the tumor microenvironment, such as proliferation, migration, angiogenesis, cell cycle arrest, and apoptosis. The phosphatidyl inositol 3-kinase (PI3K)-AKT signaling pathway is an important pathway that contributes to the survival and growth of the tumor microenvironment and is associated with these cellular processes. This current study examined the anticancer effects of fucoxanthin, a marine carotenoid isolated from brown seaweed, in the MDA-MB-231 and MDA-MB-468 TNBC cell lines. The methods used in this study include a cytotoxic assay, PI3K-AKT signaling pathway PCR arrays, and Wes analysis. Fucoxanthin (6.25 µM) + TNF-α (50 ng/mL) and TNF-α (50 ng/mL) showed no significant effect on cell viability compared to the control in both MDA-MB-231 and MDA-MB-468 cells after a 24 h treatment period. PI3K-AKT signaling pathway PCR array studies showed that in TNF-α-stimulated (50 ng/mL) MDA-MB-231 and MDA-MB-468 cells, fucoxanthin (6.25 µM) modulated the mRNA expression of 12 genes, including FOXO1, RASA1, HRAS, MAPK3, PDK2, IRS1, EIF4EBP1, EIF4B, PTK2, TIRAP, RHOA, and ELK1. Additionally, fucoxanthin significantly downregulated the protein expression of IRS1, EIF4B, and ELK1 in MDA-MB-231 cells, and no change in the protein expression of EIF4B and ELK1 was shown in MDA-MB-468 cells. Fucoxanthin upregulated the protein expression of RHOA in both cell lines. The modulation of the expression of genes and proteins of the PI3K-AKT signaling pathway may elucidate fucoxanthin's effects in cell cycle progression, apoptotic processes, migration, and proliferation, which shows that PI3K-AKT may be the possible molecular mechanism for fucoxanthin's effects. In conclusion, the results obtained in this study elucidate fucoxanthin's molecular mechanisms and indicate that fucoxanthin may be considered a promising candidate for breast cancer-targeted therapy.
Collapse
Affiliation(s)
- Shade’ A. Ahmed
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Samia S. Messeha
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Ebenezer T. Oriaku
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| |
Collapse
|
8
|
Gharbaran R. Insights into the molecular roles of FOXR2 in the pathology of primary pediatric brain tumors. Crit Rev Oncol Hematol 2023; 192:104188. [PMID: 37879492 DOI: 10.1016/j.critrevonc.2023.104188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/23/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Forkhead box gene R2 (FOXR2) belongs to the family of FOX genes which codes for highly conserved transcription factors (TFs) with critical roles in biological processes ranging from development to organogenesis to metabolic and immune regulation to cellular homeostasis. A number of FOX genes are associated with cancer development and progression and poor prognosis. A growing body of evidence suggests that FOXR2 is an oncogene. Studies suggested important roles for FOXR2 in cancer cell growth, metastasis, and drug resistance. Recent studies showed that FOXR2 is overexpressed by a subset of newly identified entities of embryonal tumors. This review discusses the role(s) FOXR2 plays in the pathology of pediatric brain cancers and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Rajendra Gharbaran
- Biological Sciences Department, Bronx Community College/City University of New York, 2155 University Avenue, Bronx, NY 10453, USA.
| |
Collapse
|
9
|
Saadh MJ, Baher H, Li Y, Chaitanya M, Arias-Gonzáles JL, Allela OQB, Mahdi MH, Carlos Cotrina-Aliaga J, Lakshmaiya N, Ahjel S, Amin AH, Gilmer Rosales Rojas G, Ameen F, Ahsan M, Akhavan-Sigari R. The bioengineered and multifunctional nanoparticles in pancreatic cancer therapy: Bioresponisive nanostructures, phototherapy and targeted drug delivery. ENVIRONMENTAL RESEARCH 2023; 233:116490. [PMID: 37354932 DOI: 10.1016/j.envres.2023.116490] [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: 05/24/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
The multidisciplinary approaches in treatment of cancer appear to be essential in term of bringing benefits of several disciplines and their coordination in tumor elimination. Because of the biological and malignant features of cancer cells, they have ability of developing resistance to conventional therapies such as chemo- and radio-therapy. Pancreatic cancer (PC) is a malignant disease of gastrointestinal tract in which chemotherapy and radiotherapy are main tools in its treatment, and recently, nanocarriers have been emerged as promising structures in its therapy. The bioresponsive nanocarriers are able to respond to pH and redox, among others, in targeted delivery of cargo for specific treatment of PC. The loading drugs on the nanoparticles that can be synthetic or natural compounds, can help in more reduction in progression of PC through enhancing their intracellular accumulation in cancer cells. The encapsulation of genes in the nanoparticles can protect against degradation and promotes intracellular accumulation in tumor suppression. A new kind of therapy for cancer is phototherapy in which nanoparticles can stimulate both photothermal therapy and photodynamic therapy through hyperthermia and ROS overgeneration to trigger cell death in PC. Therefore, synergistic therapy of phototherapy with chemotherapy is performed in accelerating tumor suppression. One of the important functions of nanotechnology is selective targeting of PC cells in reducing side effects on normal cells. The nanostructures are capable of being surface functionalized with aptamers, proteins and antibodies to specifically target PC cells in suppressing their progression. Therefore, a specific therapy for PC is provided and future implications for diagnosis of PC is suggested.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | - Hala Baher
- Department of Radiology and Ultrasonography Techniques, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | - Yuanji Li
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, 066004, China
| | - Mvnl Chaitanya
- Department of Pharmacognosy, School of Pharmacy, Lovely Professional University, Phagwara, Punjab, 144001, India
| | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, University of British Columbia, Vancouver, Canada
| | | | | | | | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Salam Ahjel
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | | | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Muhammad Ahsan
- Department of Measurememts and Control Systems, Silesian University of Technology, Gliwice, 44-100, Poland.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
10
|
Boucher Y, Posada JM, Subudhi S, Kumar AS, Rosario SR, Gu L, Kumra H, Mino-Kenudson M, Talele NP, Duda DG, Fukumura D, Wo JY, Clark JW, Ryan DP, Fernandez-Del Castillo C, Hong TS, Pittet MJ, Jain RK. Addition of Losartan to FOLFIRINOX and Chemoradiation Reduces Immunosuppression-Associated Genes, Tregs, and FOXP3+ Cancer Cells in Locally Advanced Pancreatic Cancer. Clin Cancer Res 2023; 29:1605-1619. [PMID: 36749873 PMCID: PMC10106451 DOI: 10.1158/1078-0432.ccr-22-1630] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 12/31/2022] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
PURPOSE Adding losartan (LOS) to FOLFIRINOX (FFX) chemotherapy followed by chemoradiation (CRT) resulted in 61% R0 surgical resection in our phase II trial in patients with locally advanced pancreatic cancer (LAPC). Here we identify potential mechanisms of benefit by assessing the effects of neoadjuvant LOS on the tumor microenvironment. EXPERIMENTAL DESIGN We performed a gene expression and immunofluorescence (IF) analysis using archived surgical samples from patients treated with LOS+FFX+CRT (NCT01821729), FFX+CRT (NCT01591733), or surgery upfront, without any neoadjuvant therapy. We also conducted a longitudinal analysis of multiple biomarkers in the plasma of treated patients. RESULTS In comparison with FFX+CRT, LOS+FFX+CRT downregulated immunosuppression and pro-invasion genes. Overall survival (OS) was associated with dendritic cell (DC) and antigen presentation genes for patients treated with FFX+CRT, and with immunosuppression and invasion genes or DC- and blood vessel-related genes for those treated with LOS+FFX+CRT. Furthermore, LOS induced specific changes in circulating levels of IL-8, sTie2, and TGF-β. IF revealed significantly less residual disease in lesions treated with LOS+FFX+CRT. Finally, patients with a complete/near complete pathologic response in the LOS+FFX+CRT-treated group had reduced CD4+FOXP3+ regulatory T cells (Tregs), fewer immunosuppressive FOXP3+ cancer cells (C-FOXP3), and increased CD8+ T cells in pancreatic ductal adenocarcinoma lesions. CONCLUSIONS Adding LOS to FFX+CRT reduced pro-invasion and immunosuppression-related genes, which were associated with improved OS in patients with LAPC. Lesions from responders in the LOS+FFX+CRT-treated group had reduced Tregs, decreased C-FOXP3 and increased CD8+ T cells. These findings suggest that LOS may potentiate the benefit of FFX+CRT by reducing immunosuppression.
Collapse
Affiliation(s)
- Yves Boucher
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Jessica M. Posada
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Pathology, Brigham and Women’s Hospital, Boston, University of Geneva, CH-1211 Geneva, Switzerland
| | - Sonu Subudhi
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Ashwin S. Kumar
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
- Harvard–MIT Division of Health Sciences and Technology, Cambridge, University of Geneva, CH-1211 Geneva, Switzerland
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, University of Geneva, CH-1211 Geneva, Switzerland
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, University of Geneva, CH-1211 Geneva, Switzerland
| | - Liqun Gu
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Heena Kumra
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Mari Mino-Kenudson
- Department of Pathology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Nilesh P. Talele
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Dan G. Duda
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Dai Fukumura
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Jennifer Y. Wo
- Department of Radiation Oncology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Jeffrey W. Clark
- Department of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - David P. Ryan
- Department of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | | | - Theodore S. Hong
- Department of Radiation Oncology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Mikael J. Pittet
- Department of Pathology and Immunology, University of Geneva, CH-1211 Geneva, Switzerland
- Ludwig Institute for Cancer Research, 1005 Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Rakesh K. Jain
- Steele Laboratories of Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
11
|
Wang K, Sun X, Sun Y, Jiao B, Yao J, Hu Y, Deng Q, Dong J, Wang W, Wang Y, Li C. Transcriptional regulation of macrophages in heart failure. Front Cardiovasc Med 2023; 10:1148041. [PMID: 37063966 PMCID: PMC10097991 DOI: 10.3389/fcvm.2023.1148041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
Adverse cardiac remodeling after acute myocardial infarction is the most important pathological mechanism of heart failure and remains a major problem in clinical practice. Cardiac macrophages, derived from tissue resident macrophages and circulating monocyte, undergo significant phenotypic and functional changes following cardiac injury and play crucial roles in inflammatory response and tissue repair response. Currently, numerous studies indicate that epigenetic regulatory factors and transcription factors can regulate the transcription of inflammatory and reparative genes and timely conversion of inflammatory macrophages into reparative macrophages and then alleviate cardiac remodeling. Accordingly, targeting transcriptional regulation of macrophages may be a promising option for heart failure treatment. In this review, we not only summarize the origin and function of cardiac macrophages, but more importantly, describe the transcriptional regulation of macrophages in heart failure, aiming to provide a potential therapeutic target for heart failure.
Collapse
Affiliation(s)
- Keyan Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoqian Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Boyang Jiao
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junkai Yao
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yueyao Hu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qiong Deng
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jianteng Dong
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
- Correspondence: Wei Wang Yong Wang Chun Li
| | - Yong Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
- Correspondence: Wei Wang Yong Wang Chun Li
| | - Chun Li
- Beijing Key Laboratory of Traditional Chinese Medicine (TCM) Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, China
- Modern Research Center for Traditional Chinese Medicine (TCM), Beijing University of Chinese Medicine, Beijing, China
- Correspondence: Wei Wang Yong Wang Chun Li
| |
Collapse
|
12
|
CircAMOTL1 RNA and AMOTL1 Protein: Complex Functions of AMOTL1 Gene Products. Int J Mol Sci 2023; 24:ijms24032103. [PMID: 36768425 PMCID: PMC9916871 DOI: 10.3390/ijms24032103] [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: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The complexity of the cellular proteome facilitates the control of a wide range of cellular processes. Non-coding RNAs, including microRNAs and long non-coding RNAs, greatly contribute to the repertoire of tools used by cells to orchestrate various functions. Circular RNAs (circRNAs) constitute a specific class of non-coding RNAs that have recently emerged as a widely generated class of molecules produced from many eukaryotic genes that play essential roles in regulating cellular processes in health and disease. This review summarizes current knowledge about circRNAs and focuses on the functions of AMOTL1 circRNAs and AMOTL1 protein. Both products from the AMOTL1 gene have well-known functions in physiology, cancer, and other disorders. Using AMOTL1 as an example, we illustrate how focusing on both circRNAs and proteins produced from the same gene contributes to a better understanding of gene functions.
Collapse
|
13
|
Xuan Z, Fang L, Zhang G, Zhang X, Jiang J, Wang K, Huang P. The Heterogeneity of Tumour-Associated Macrophages Contributes to the Recurrence and Outcomes of Glioblastoma Patients. J Mol Neurosci 2023; 73:1-14. [PMID: 36542317 DOI: 10.1007/s12031-022-02081-z] [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: 08/22/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022]
Abstract
Cellular heterogeneity and immune cell molecular phenotypes may be involved in the malignant progression of glioblastoma (GBM). In this study, we aimed to know whether the heterogeneity of tumour-associated macrophages contributes to the recurrence and outcomes of glioblastoma patients. Single-cell RNA sequencing (scRNA-Seq) data were used to assess the heterogeneity of CD45 + immune cells in recurrent GBM and analyse differentially expressed genes (DEGs) in master cells. Then, a prognostic signature based on the identified DEGs was established and validated, the correlation between risk score and tumour microenvironment (TME) was explored. The correlation between immune infiltration and LGMN, an important DEG in GBM tumour-associated macrophages (TAMs) was illuminated, using integrated bioinformatics analyses. Finally, immunohistochemistry and multiplex immunohistochemistry (mIHC) were used to analyse the expression of LGMN in GBM tissues from our hospital. scRNA-Seq analysis showed that the heterogeneity of recurrent GBM mainly comes from TAMs, which can be divided into 8 cell subclusters. Among these subclusters, TAM1 (markers: CXCL10, ADORA3), TAM3 (markers: MRC1, CFP), TAM4 (markers: GPNMB, PLTP), and TAM5 (markers: CCL4, IRAK2) were specifically present in recurrent GBM. After 342 DEGs in TAMs were identified, a prognostic signature was established based on 13 TAM-associated DEGs, and this signature could serve as an excellent prognostic predictor for patients with GBM. LGMN, one of 13 TAM-associated DEGs, was an important gene in lysosome pathway, we found that macrophage infiltration levels were higher after LGMN upregulation. GBM tissues from our hospital were collected for histopathologic validation, then LGMN was co-expressed with CD68, which is associated with the immune regulation of GBM. In conclusion, cell heterogeneity of TAMs is important for recurrent GBM, a prognostic signature based on 13 TAM-related DEGs can predict the survival outcome of GBM patients. An important DEG, LGMN may regulate the immune cell infiltration of GBM.
Collapse
Affiliation(s)
- Zixue Xuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Ling Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guobing Zhang
- Quality Management Office, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xin Zhang
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jinying Jiang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| | - Kai Wang
- Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China. .,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China.
| |
Collapse
|
14
|
Wang J, Luo J, Wu X, Li Z. ELK1 suppresses SYTL1 expression by recruiting HDAC2 in bladder cancer progression. Hum Cell 2022; 35:1961-1975. [DOI: 10.1007/s13577-022-00789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022]
|
15
|
Wei S, Yu Z, Shi R, An L, Zhang Q, Zhang Q, Zhang T, Zhang J, Wang H. GPX4 suppresses ferroptosis to promote malignant progression of endometrial carcinoma via transcriptional activation by ELK1. BMC Cancer 2022; 22:881. [PMID: 35962333 PMCID: PMC9373394 DOI: 10.1186/s12885-022-09986-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/08/2022] [Indexed: 12/21/2022] Open
Abstract
Background Glutathione Peroxidase 4 (GPX4) is a key protein that inhibits ferroptosis. However, its biological regulation and mechanism in endometrial cancer (EC) have not been reported in detail. Methods The expression of GPX4 in EC tissues was determined by TCGA databases, qRT-PCR, Western blot, and immunohistochemistry (IHC). The effects of GPX4 on EC cell proliferation, migration, apoptosis, and tumorigenesis were studied in vivo and in vitro. In addition, ETS Transcription Factor ELK1 (ELK1) was identified by bioinformatics methods, dual-luciferase reporter assay, and chromatin immunoprecipitation (ChIP). Pearson correlation analysis was used to evaluate the association between ELK1 and GPX4 expression. Results The expression of GPX4 was significantly up-regulated in EC tissues and cell lines. Silencing GPX4 significantly inhibited the proliferation, migration ability, induced apoptosis, and arrested the cell cycle of Ishikawa and KLE cells. Knockdown of GPX4 accumulated intracellular ferrous iron and ROS, disrupted MMP, and increased MDA levels. The xenograft tumor model also showed that GPX4 knockdown markedly reduced tumor growth in mice. Mechanically, ELK1 could bind to the promoter of GPX4 to promote its transcription. In addition, the expression of ELK1 in EC was positively correlated with GPX4. Rescue experiments confirmed that GPX4 knockdown could reverse the strengthens of cell proliferation and migration ability and the lower level of Fe2+ and MDA caused by upregulating ELK1. Conclusion The results of the present study suggest that ELK1 / GPX4 axis plays an important role in the progress of EC by promoting the malignant biological behavior and inducing ferroptosis of EC cells, which provides evidence for investigating the potential therapeutic strategies of endometrial cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09986-3.
Collapse
Affiliation(s)
- Sitian Wei
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Zhicheng Yu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Rui Shi
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Lanfen An
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qi Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qian Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tangansu Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jun Zhang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| | - Hongbo Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
16
|
Dong Y, Zhang Y, Zhang Y, Pan X, Bai J, Chen Y, Zhou Y, Lai Z, Chen Q, Hu S, Zhou Q, Zhang Y, Ma F. Dissecting the process of human neutrophil lineage determination by using alpha-lipoic acid inducing neutrophil deficiency model. Redox Biol 2022; 54:102392. [PMID: 35797799 PMCID: PMC9287745 DOI: 10.1016/j.redox.2022.102392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/25/2022] Open
Abstract
Granulocyte-monocyte progenitors (GMPs) differentiate into both neutrophils and monocytes. Recently, uni-potential neutrophil progenitors have been identified both in mice and humans using an array of surface markers. However, how human GMPs commit to neutrophil progenitors and the regulatory mechanisms of fate determination remain incompletely understood. In the present study, we established a human neutrophil deficiency model using the small molecule alpha-lipoic acid. Using this neutrophil deficiency model, we determined that the neutrophil progenitor commitment process from CD371+ CD115– GMPs defined by CD34 and CD15 and discovered that critical signals generated by RNA splicing and rRNA biogenesis regulate the process of early commitment for human early neutrophil progenitors derived from CD371+ CD115– GMPs. These processes were elucidated by single-cell RNA sequencing both in vitro and in vivo derived cells. Sequentially, we identified that the transcription factor ELK1 is essential for human neutrophil lineage commitment using the alpha-lipoic acid (ALA)-inducing neutrophil deficiency model. Finally, we also revealed differential roles for long-ELK1 and short-ELK1, balanced by SF3B1, in the commitment process of neutrophil progenitors. Taken together, we discovered a novel function of ALA in regulating neutrophil lineage specification and identified that the SF3B1-ELK axis regulates the commitment of human neutrophil progenitors from CD371+ CD115– GMPs. ALA completely blocks the differentiation of human neutrophils derived from CD34+ stem cells in ex-vivo culture. CD34 and CD15 could be used to define the early differentiation stages of human neutrophil lineage determination. SF3B1-ELK1 signal axis regulates human neutrophil lineage determination.
Collapse
|