1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [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: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Zhang H, Wang J, Liu C, Yan K, Wang X, Sheng X. Interactions between long non-coding RNAs and m6 A modification in cancer. Discov Oncol 2025; 16:579. [PMID: 40253659 PMCID: PMC12009795 DOI: 10.1007/s12672-025-02387-5] [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: 01/08/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of transcripts exceeding 200 nucleotides (nt) in length, which are broadly implicated in a broad spectrum of physiological and pathological processes, including allelic imprinting, genome packaging, chromatin remodeling, transcriptional activation and disruption, as well as the occurrence and progression of oncogenesis. N6-methyladenosine (m6 A) methylation stands as the most prevalent RNA modification, affecting multiple facets of RNA biology such as stability, splicing, transport, translation, degradation, and tertiary structure. Aberrant m6 A modifications are intimately implicated in cancer progression. In recent years, there has been a growing number of studies illuminating the dynamic interplay between lncRNAs and m6 A modifications, revealing that lncRNAs can modulate the activity of m6 A regulators, while m6 A not only affects the structural integrity but also the translational efficiency and stability of lncRNAs. Together, the interactions between lncRNAs and m6 A modifications significantly impact downstream oncogenes, cancer suppressor genes, cellular metabolism, epithelial-mesenchymal transition, angiogenesis, drug transport, DNA homology repair, and epigenetics, subsequently influencing tumorigenesis, metastasis, and drug resistance. This article endeavors to clarify the functions and mechanisms of lncRNAs and m6 A modifications interaction in cancer to provide promising insights for cancer diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Han Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Junjie Wang
- Department of Pathophysiology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China
| | - Chunyi Liu
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Kaiqin Yan
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Xiaomeng Wang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Xiumei Sheng
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
| |
Collapse
|
3
|
Kesimoglu ZN, Rifat JIM, Bozdag S. Computational inference of co-regulatory modules from transcription factors, MicroRNAs, and their targets using CanMod2. Sci Rep 2025; 15:12521. [PMID: 40216929 PMCID: PMC11992115 DOI: 10.1038/s41598-025-97476-4] [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: 08/26/2023] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Gene regulators such as Transcription Factors (TFs) and microRNAs (miRNAs) regulate genes at the transcriptional and post-transcriptional levels, respectively. There is a complex interplay of regulatory patterns of TFs and miRNAs. Some TFs and miRNAs regulate the activity of their target genes individually, some co-regulate the activity of the same set of genes, some TFs regulate miRNA activity, and some miRNAs regulate TFs. As dysregulation in gene regulation can lead to various diseases like cancer, it is a significant problem to find the interplay among TFs, miRNAs, and their target genes. Here, we propose a computational pipeline, CanMod2, which infers modules of TFs, miRNAs, and their co-regulatory targets that are involved in a common biological process. In this work, we have introduced several algorithmic enhancements to the earlier version of CanMod2. We applied CanMod2 to five cancer types and analyzed the inferred modules extensively. Our results show that the inferred modules were enriched in cancer-related biological processes and pathways. The hub regulators that occur in many modules were among cancer-related genes and miRNAs. The inferred regulator-target interactions were significantly enriched in ground truth interactions. CanMod2 source code and documentation are publicly available at https://github.com/bozdaglab/CanMod2 .
Collapse
Affiliation(s)
- Ziynet Nesibe Kesimoglu
- Department of Computer Science & Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Jubair Ibn Malik Rifat
- Department of Computer Science & Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
- Center for Computational Life Sciences, University of North Texas, Denton, TX, USA
| | - Serdar Bozdag
- Department of Computer Science & Engineering, University of North Texas, Denton, TX, USA.
- Department of Mathematics, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
- Center for Computational Life Sciences, University of North Texas, Denton, TX, USA.
| |
Collapse
|
4
|
Tao M, Xue M, Zhou D, Zhang L, Hou X, Zhu X, Feng S, Yan H, Qian X, Wei L, Zong C, Yang X, Zhang L. Lipopolysaccharide Induces Resistance to CAR-T Cell Therapy of Colorectal Cancer Cells through TGF-β-Mediated Stemness Enhancement. Mol Pharm 2025; 22:1790-1803. [PMID: 40116228 DOI: 10.1021/acs.molpharmaceut.4c00264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy is a cellular immunotherapy that has emerged in recent years, and increasing studies showed that therapeutic resistance to CAR-T cell therapy presents in colorectal cancer patients. Lipopolysaccharide (LPS), a component of the cell wall of Gram-negative bacteria, is known to preserve a high concentration in the colon. Whether LPS is a contributing factor to the development of resistance in colorectal cancer cells against CAR-T cell therapy remains unclear. For in vivo experiments, colorectal cancer cells COLO205 were pretreated with LPS for 24 h and then were injected into nude mice through the tail vein, followed by CAR-T cells transplantation one day later. Later, the number of tumors in the lung tissues of the mice was observed. The in vitro experiments were performed on COLO205 cells, which were treated with LPS for 24 h. The effect of LPS on the stemness of COLO205 and SW620 cells was observed by using the colony formation assay and spheroidization experiments. The effect of LPS on the expression of stemness-related genes, including CD44, SOX2, and NANOG, was observed by qRT-PCR assay, Western blotting assay, and immunofluorescence staining. Inhibitors of TGF-β and the MYD88 inhibitor were used to study the mechanisms by which LPS induces the stemness enhancement and resistance to CAR-T cell therapy of COLO205 cells. The correlation between MYD88 and TGFB1, as well as the correlation between TGFB1 and stemness-related genes was analyzed using the TCGA database. Both the in vivo assay of nude mice and the in vitro assay showed that LPS pretreatment could induce resistance to CAR-T cell therapy of colorectal cancer cells. LPS could enhance COLO205 and SW620 cells stemness presented by upregulation of CD44, SOX2, and NANOG. The reverse interfering assay using the TGF-β inhibitor indicated that the autosecretion of TGF-β induced by LPS played a critical role in the stemness enhancement of colorectal cancer cells. The TCGA database analysis revealed a strong positive correlation between MYD88 and TGFB1. Additionally, TGFB1 has been found to upregulate the expression of genes associated with stemness. Further mechanism studies showed that the TLR4/MYD88 pathway medicates LPS-induced TGF-β expression. Our results suggested that LPS-induced resistance to CAR-T cell therapy of colorectal cancer cells through stemness enhancement. TLR4/MYD88 signal pathway-dependent TGF-β expression was involved in stemness enhancement and CAR-T cell therapy resistance. In conclusion, our findings help us to understand the underlying mechanisms of CAR-T cell therapy resistance and indicate that inhibitors of TGF-β and MYD88 are promising targeting candidates to promote a therapeutic effect of CAR-T cell therapy in colorectal cancer in the clinic.
Collapse
Affiliation(s)
- Min Tao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengmeng Xue
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Daoyu Zhou
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Luyao Zhang
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaojuan Hou
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Xinyu Zhu
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shiyao Feng
- Anhui Medical University, Hefei 230032, China
| | - Haixin Yan
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Xiaofeng Qian
- Shanghai Putuo District Liqun Hospital, Shanghai 200061, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Chen Zong
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Xue Yang
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Li Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
5
|
Lee D, Jeong HS, Hwang SY, Lee YG, Kang YJ. ABCB1 confers resistance to carboplatin by accumulating stem-like cells in the G2/M phase of the cell cycle in p53 null ovarian cancer. Cell Death Discov 2025; 11:132. [PMID: 40175339 PMCID: PMC11965561 DOI: 10.1038/s41420-025-02435-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/19/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
High-grade ovarian serous carcinoma, mostly bearing the various mutations in the TP53 gene, typically relapses within six months after first-line therapy due to chemoresistance, with a median overall survival of less than a year. However, the molecular mechanisms of action behind acquired drug resistance, particularly in relation to different TP53 mutation types, have not been fully elucidated. In this study, we demonstrated that acquired resistance to carboplatin in SKOV3 harboring a p53null mutation, but not in OVCAR3 with a p53R248Q, induces a significant portion of cells accumulated in the G2/M phase of the cell cycle, where cells highly expressed stemness marker with elevated proliferative capacity, which we believe was reversed by ABCB1 inhibition to the levels observed in non-resistant parental cells. ABCB1 suppression re-sensitized carboplatin-resistant cells to additional genotoxic stress and reduced their proliferative ability by recovering DNA repair activity and lowering stemness-like features, especially in the G2/M-distributed fraction. This suggests that high levels of stemness and attenuated DNA repair function exhibited in the G2/M-accumulated portion may be a key contributor of chemoresistance in patients with ovarian cancer bearing a p53null mutation, but not other types of mutations expressing p53. Furthermore, the inhibition of ΔNp73 resulted in the suppression of ABCB1, which consequently restricted cell growth in carboplatin-resistant SKOV3, suggesting that the ΔNp73 may act as an upstream regulator of the ABCB1. Notably, combinatorial treatment of carboplatin with the p53 reactivator, APR-246, proved effective in overcoming chemoresistance in OVCAR3 with the p53R248Q. Our findings suggest that the ΔNp73-ABCB1 axis is a promising molecular target for carboplatin-resistant ovarian cancers harboring p53null mutations, which we uncovered could be utilized to increase the efficacy of conventional anti-cancer therapies, to develop more efficient combinatorial therapeutic interventions directed toward overcoming the chemoresistance and improving the survival rates in patients with ovarian cancer.
Collapse
Affiliation(s)
- Danbi Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Hyun-Seok Jeong
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Sun-Young Hwang
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Yu-Gyeong Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Youn-Jung Kang
- Department of Biochemistry, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam-si, South Korea.
| |
Collapse
|
6
|
Wang J, Liao J, Cheng Y, Chen M, Huang A. LAPTM4B enhances the stemness of CD133 + liver cancer stem-like cells via WNT/β-catenin signaling. JHEP Rep 2025; 7:101306. [PMID: 40171299 PMCID: PMC11960653 DOI: 10.1016/j.jhepr.2024.101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 04/03/2025] Open
Abstract
Background & Aims Lysosome-associated protein transmembrane 4β (LAPTM4B) is an oncogene implicated in the malignant progression of hepatocellular carcinoma (HCC). Previous research established a strong association between LAPTM4B and HCC stemness. However, specific mechanisms by which LAPTM4B regulates and maintains the stemness of liver cancer stem cells remain unclear. Therefore, we investigated the effects of LAPTM4B on the stemness regulation of cluster of differentiation 133 (CD133)+ liver cancer stem-like cells (CSLCs). Methods We used RNA interference and overexpression techniques in both in vitro and in vivo models. The involvement of LAPTM4B in wingless/integrated (WNT)/β-catenin signaling was examined through western blotting, immunofluorescence, and immunoprecipitation. The impact of LAPTM4B on β-catenin phosphorylation and ubiquitination was analyzed to elucidate its role in promoting stemness. Clinical relevance was evaluated in an in-house cohort of 105 specimens from patients with HCC through immunohistochemical and microarray analysis, enabling investigation of correlations with clinical outcomes. Results LAPTM4B promoted the self-renewal ability, chemoresistance, and tumorigenicity of CD133+ CSLCs. Mechanistically, aberrant LAPTM4B upregulation facilitated β-catenin nuclear translocation (nucleocytoplasmic separation assay, p <0.001) and inhibited its phosphorylation (p <0.01). In addition, LAPTM4B interacts with the deubiquitinating enzymes ubiquitin carboxyl-terminal hydrolase (USP)-1 and USP14, reducing β-catenin ubiquitination. Furthermore, patients with high LAPTM4B and β-catenin expression had markedly shorter 3-year overall survival rate (42.9% vs. 74.4%; hazard ratio, 5.174; 95% CI 2.280-11.741, p <0.001). Conclusions LAPTM4B promotes CD133+ CSLC stemness by activating WNT/β-catenin signaling by inhibiting β-catenin phosphorylation and ubiquitination degradation. The role of LAPTM4B in regulating WNT/β-catenin signaling suggests that LAPTM4B serves as a therapeutic target for impairing HCC stemness and progression. Impact and implications LAPTM4B contributes significantly to CD133+ CSLC stemness and inhibits β-catenin phosphorylation and ubiquitination degradation, activating WNT/β-catenin signaling. WNT inhibitors suppress LAPTM4B-induced CD133+ CSLC stemness. Thus, targeting the LAPTM4B-WNT/β-catenin axis could improve antitumor efficacy.
Collapse
Affiliation(s)
- Jiahong Wang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| | - Jianping Liao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou 510060, China
| | - Ye Cheng
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Diagnostical Pathology Center, Fujian Medical University, Fuzhou, Fujian 350004, China
| | - Meirong Chen
- Department of Pathology, Quanzhou Maternity and Children’s Hospital, Quanzhou, Fujian 362000, China
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian 350004, China
| |
Collapse
|
7
|
Yang C, Zhang Y, Yan L, Liu A, Li F, Li Y, Zhang Y. Comprehensive Analysis of GPSM2: From Pan-Cancer Analysis to Experimental Validation. J Cell Mol Med 2025; 29:e70527. [PMID: 40208185 PMCID: PMC11984320 DOI: 10.1111/jcmm.70527] [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: 04/26/2024] [Revised: 11/04/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
G-protein signalling modulator 2 (GPSM2) plays an important role in maintaining cell polarisation and regulating the cell cycle; however, a systematic and comprehensive analysis of GPSM2 in cancer is still lacking. Using extensive multi-omics data, we explored the pan-cancer expression levels of GPSM2 from multiple perspectives and its association with prognosis, diagnosis, tumour stemness, immune-related genes, immune cell infiltration, genomic instability, and response to immunotherapy. We also elucidated the potential pan-cancer biological functions of GPSM2 using gene set enrichment analysis (GSEA) and searched for targeted drugs that affect GPSM2 expression using connectivity map analysis. To elucidate the effect of GPSM2 on colon cancer, we evaluated its effect on the biological behaviour of two colon cancer cell lines. In this study, GPSM2 was systematically analysed and shown to have satisfactory performance in disease diagnosis and prognostic prediction of various cancers. G-protein signalling modulator 2 plays an important role in the genesis and development of various tumours and is a potential tumour diagnostic and prognostic biomarker as well as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Chunjiao Yang
- Department of OncologyThe Fifth Affiliated Hospital of Guangxi Medical University & The First People's Hospital of NanningNanningChina
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yuzhe Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Lirong Yan
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Aoran Liu
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Fang Li
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| | - Yanke Li
- Department of Anorectal SurgeryThe First Hospital of China Medical UniversityShenyangChina
| | - Ye Zhang
- The First Laboratory of Cancer InstituteThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
8
|
Hong WC, Kim M, Kim JH, Kang HW, Fang S, Jung HS, Kwon W, Jang JY, Kim HJ, Park JS. The FOXP1-ABCG2 axis promotes the proliferation of cancer stem cells and induces chemoresistance in pancreatic cancer. Cancer Gene Ther 2025:10.1038/s41417-025-00896-7. [PMID: 40169859 DOI: 10.1038/s41417-025-00896-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025]
Abstract
Pancreatic cancer is an aggressive disease with low survival and high recurrence rates. A major obstacle in treating pancreatic cancer is the frequent development of chemoresistance to the standard therapeutic drug, gemcitabine. One mechanism by which pancreatic cancer develops chemoresistance is through the proliferation of cancer stem cells (CSC). However, the mechanisms regulating stemness in chemoresistant tumors remain unclear. Here, we found that the expression of the transcription factor Forkhead Box P1 (FOXP1) was elevated in chemoresistant pancreatic cancer and crucial for establishing CSC characteristics. Silencing FOXP1 reduced the expressions of stemness-associated genes and diminished the formation of both spheroids and colonies, highlighting the crucial role of FOXP1 in regulating stemness in chemoresistant tumor cells. Mechanistically, we discovered that FOXP1 regulates the expression of ATP-binding cassette superfamily G member 2 (ABCG2), which induces the efflux of gemcitabine. Knockdown of FOXP1 reduced the expression of ABCG2, resulting in decreased proliferation and increased sensitivity to gemcitabine. Moreover, the inhibition of FOXP1 in orthotopic mouse models reduced tumor growth and proliferation, and enhanced sensitivity to gemcitabine. Together, our data reveal FOXP1 as a potent oncogene that promotes CSC growth in chemoresistant pancreatic cancer.
Collapse
Affiliation(s)
- Woosol Chris Hong
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minsoo Kim
- Korea Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Ju Hyun Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeon Woong Kang
- Korea Brain Korea 21 PLUS Project for Medical Science, Yonsei University, College of Medicine, Seoul, Republic of Korea
| | - Sungsoon Fang
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye-Sol Jung
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jung Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Joon Seong Park
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Zou J, Chen J, Deng L, Xu B, Yu T, Wang J, He C. Mechanistic insights into SENP1 and OCT4 interaction in promoting drug resistance and stem cell features in colon cancer. Am J Physiol Cell Physiol 2025; 328:C1260-C1278. [PMID: 40063360 DOI: 10.1152/ajpcell.00817.2024] [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: 11/04/2024] [Revised: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025]
Abstract
This study explores the molecular mechanism by which sentrin/SUMO-specific protease 1 (SENP1) promotes cisplatin (Cis) resistance and tumor stem cell characteristics in colon adenocarcinoma (COAD) through deSUMOylation-mediated modification of octamer-binding transcription factor 4 (OCT4). By analyzing single-cell and transcriptome sequencing datasets, we identified key genes and regulatory pathways in both resistant and sensitive COAD cells. Malignant cells were isolated and evaluated for stemness using the infercnv package, and differential genes between Cis-resistant and -sensitive groups were identified. Machine learning algorithms highlighted essential genes, and databases predicted interaction sites between OCT4 and SENP1. In vitro experiments using enriched HCT116 stem cells revealed that SENP1 and OCT4 expression significantly elevated CD44 and CD133 levels, enhancing stemness. Functional assays showed that SENP1's deSUMOylation of OCT4 intensified Cis resistance, migration, and invasion in cisplatin-resistant cell line 116 (Cis-116) cells. In vivo, SENP1 knockdown reduced tumor growth and stem cell markers, whereas OCT4 overexpression escalated tumor metastasis and structural damage. These findings demonstrate that SENP1's modulation of OCT4 is central to COAD's resistance and stem cell properties, offering a novel target for COAD therapy.NEW & NOTEWORTHY This study uncovers the critical role of SENP1 in regulating OCT4 through deSUMOylation, driving Cis resistance and tumor stemness in COAD. Targeting this pathway may provide novel therapeutic strategies for COAD management.
Collapse
Affiliation(s)
- Jun Zou
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jing Chen
- Department of nursing, Nanchang Medical College, Nanchang, People's Republic of China
| | - Lei Deng
- Department of Medical Oncology, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Bangran Xu
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jun Wang
- General Surgery Department of the Trauma Center, Nanchang First Hospital, Nanchang, People's Republic of China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| |
Collapse
|
10
|
Huang C, Tang B, Chen W, Chen J, Zhang H, Bai M. Multiomic traits reveal that critical irinotecan-related core regulator FSTL3 promotes CRC progression and affects ferroptosis. Cancer Cell Int 2025; 25:115. [PMID: 40140870 PMCID: PMC11938592 DOI: 10.1186/s12935-025-03753-7] [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: 12/09/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Irinotecan is a widely used chemotherapy drug in colorectal cancer (CRC). The evolution and prognosis of CRC involve complex mechanisms and depend on the drug administered, especially for irinotecan. However, the specific mechanism and prognostic role of irinotecan-related regulators remain to be elucidated. METHODS Data from public databases were used to explore the multiomic traits of irinotecan-related regulators through bioinformatics analysis. RT‒qPCR, western blotting, transmission electron microscopy and flow cytometry were used as experimental validations. RESULTS Iriscore (irinotecan-related score) was constructed based on irinotecan-related regulators, and a high iriscore predicted a poor prognosis, poor therapeutic response and the MSS/MSI-L status. Single-cell analysis revealed that FSTL3 and TMEM98 were mainly expressed in CRC stem cells. Potential transcription factors (E2F1, STAT1, and TTF2) and therapeutic drugs (telatinib) that target irinotecan-related regulators were identified. FSTL3 was the core risk irinotecan-related regulator. Some ferroptosis regulators (GPX4, HSPB1 and RGS4) and related metabolic pathways (lipid oxidation and ROS metabolism) were correlated significantly with FSTL3. In vitro, irinotecan inhibited the expression of FSTL3 and ferroptotic defence proteins (GPX4 and SLC7A11), and induced lipid peroxidation and intracellular Fe (2+) ions concentration increased. CONCLUSIONS We confirmed that irinotecan-related regulators, especially FSTL3, have effective prognostic value in CRC and speculated that FSTL3 may promote CRC progression and affect ferroptosis, which is beneficial for identifying candidate targeted irinotecan-related regulators and accurate individualized treatment strategies for CRC.
Collapse
Affiliation(s)
- Chengyi Huang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Bufu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenjuan Chen
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China
| | - Jinggang Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China
| | - Huojun Zhang
- Department of Radiation Oncology, Changhai Hospital Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Minghua Bai
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
11
|
Chen H, Ling R, Lai J, Liu Z, Wang Z, Yang H, Kong Y. CD44v6-mediated regulation of gastric cancer stem cells: a potential therapeutic target. Clin Exp Med 2025; 25:80. [PMID: 40069421 PMCID: PMC11897096 DOI: 10.1007/s10238-025-01611-4] [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: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Gastric cancer is the fourth most common cause of cancer-related deaths globally. Cancer stem cells (CSCs) play an essential role in tumor initiation, development, and chemoresistance. However, the molecular mechanisms that regulate CSC traits in gastric cancer, particularly the role of CD44v6 as a key CSC marker, remain poorly understood. Here, we demonstrate that CD44v6 is markedly upregulated in gastric cancer tissues and correlates with poor prognosis. Functional assays, including colony formation, wound healing, proliferation, and apoptosis assays, show that CD44v6 enhances CSC characteristics, such as self-renewal, proliferation, migration, and cisplatin chemoresistance. CD44v6 knockdown effectively suppresses these aggressive phenotypes. Mechanistically, CD44v6 regulates the expression of key CSC markers, including CD24, CD133, EpCAM, as well as stemness-related transcription factors Oct-4 and Nanog. Additionally, CD44v6 enhances cell proliferation and drug resistance in both in vitro and in vivo experiments. Collectively, our findings highlight the significant role of CD44v6 in regulating gastric CSC traits, suggesting it's a potential as a biomarker and therapeutic target for improving gastric cancer treatment outcomes, particularly in overcoming chemoresistance.
Collapse
Affiliation(s)
- Hao Chen
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Ruoyu Ling
- Department of General Surgery and Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Jiayu Lai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Zhiqi Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, Guangdong Province, China.
| | - Hua Yang
- Department of Basic Medicine, School of Medicine, Foshan University, Foshan, 528225, Guangdong Province, China.
| | - Yi Kong
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology (SUAT), Shenzhen, 518107, Guangdong Province, China.
| |
Collapse
|
12
|
Wei JR, Lu MY, Wei TH, Fleishman JS, Yu H, Chen XL, Kong XT, Sun SL, Li NG, Yang Y, Ni HW. Overcoming cancer therapy resistance: From drug innovation to therapeutics. Drug Resist Updat 2025; 81:101229. [PMID: 40081221 DOI: 10.1016/j.drup.2025.101229] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
One of the major limitations of cancer therapy is the emergence of drug resistance. This review amis to provide a focused analysis of the multifactorial mechanisms underlying therapy resistance,with an emphasis on actionable insights for developing novel therapeutic strategies. It concisely outlines key factors contributing to therapy resistance, including drug delivery barriers, cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer heterogeneity, tumor microenvironment (TME), genetic mutations, and alterlations in gene expression. Additionally, we explore how tumors evade targeted therapies through pathway-specific mechanisms that restore disrupted signaling pathways. The review critically evaluates innovative strategies designed to sensitize resistant tumor cells, such as targeted protein dedgradation, antibody-drug conjugates, structure-based drug design, allosteric drugs, multitarget drugs, nanomedicine and others We also highlight the importance of understanding the pharmacological actions of these agents and their integration into treatment regimens. By synthesizing current knowledge and identifying gaps in our understanding, this review aims to guide future research and improve patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Jin-Rui Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China; The First Clinical College of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meng-Yi Lu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hui Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiao-Li Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiang-Tu Kong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hai-Wen Ni
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
13
|
Tian J, Zhang L, La X, Fan X, Li Z. MiR-769-5p of macrophage exosomes induced by GRP78 promotes stemness and chemoresistance in colorectal cancer. Cell Death Dis 2025; 16:156. [PMID: 40044682 PMCID: PMC11882909 DOI: 10.1038/s41419-025-07466-7] [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: 10/05/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
The tumor microenvironment (TME) plays an important role in tumorigenesis and development. Tumor-associated macrophages (TAMs) are essential members of the TME, the exosomes and miRNAs they secrete are crucial in tumor regulation. Our previous study showed that GRP78-induced macrophages infinitely tend to be M2-type TAMs. In this study, the exosomes of M0 and GRP78-induced macrophage were collected and co-incubated with colorectal cancer (CRC) cells. The results implied that macrophage exosomes induced by GRP78 (GRP78-exos) significantly promoted stemness and chemoresistance in CRC in vitro and in vivo. Further, the top 5 miRNAs upregulated in GRP78-exos were obtained from miRNA sequencing data. The qRT-PCR validation revealed that miR-769-5p was the most observably upregulated and could be directly transferred into CRC cells via GRP78-exos. Mechanistically, the study indicated that miR-769-5p targeted MAPK1 to regulate the cell cycle-related proteins RB1, cyclin D1, and cyclin E1. This contributes to CRC cells entering a quiescent state, which leads to the development of chemoresistance. Moreover, miR-769-5p is also expressed higher in the tissues of 5-FU-resistant CRC patients. In summary, the findings indicate a novel function of miR-769-5p as a potential marker for the diagnosis and treatment of chemotherapy resistance in CRC.
Collapse
Affiliation(s)
- Jinmiao Tian
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China
| | - Lichao Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Xiaoqin La
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Xiaxia Fan
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China
| | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of the National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
14
|
Li Y, Liu H, Li J, Fu C, Jiang B, Chen B, Zou Y, Yu B, Song B. MLLT3 Regulates Melanoma Stemness and Progression by Inhibiting HMGB1 Nuclear Entry and MAGEA1 M 5C Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408529. [PMID: 39716999 PMCID: PMC11904942 DOI: 10.1002/advs.202408529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/03/2024] [Indexed: 12/25/2024]
Abstract
Melanoma stem cells are a kind of cells with self-renewal and multi-directional differentiation potential. They are one of the key factors in the occurrence, development and metastasis of melanoma. This study demonstrates that MLLT3 is a transcription factor that regulates the stemness and progression of melanoma. MLLT3 interacted with HMGB1 to inhibit its entry into the nucleus, MLLT3 interacted with YBX1 to inhibit its reading of m5C of MAGEA1, thereby inhibiting the mRNA stability of MAGEA1, and directly transcribed P53 to inhibit the stemness, proliferation and metastasis of melanoma cells. This study further explored the potential mechanism of the interaction between miR-542-3p/miR-3922-3p and MLLT3. Furthermore, the scRNA-seq of melanoma cells with MLLT3 knock-out resulted in important changes in cell subsets, activating the TP53 and MAPK pathways and transforming into stem cells. The results indicate that the transcription factor MLLT3 is a suppressor gene that regulates the stemness and progression of melanoma, and is expected to become a target for melanoma therapy.
Collapse
Affiliation(s)
- Yaling Li
- Institute of Biomedical and Health Engineering, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Hong Liu
- Department of Otorhinolaryngology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361000, China
| | - Jingyi Li
- Institute of Biomedical and Health Engineering, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Chang Fu
- Institute of Biomedical and Health Engineering, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Bin Jiang
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Bancheng Chen
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Yanfen Zou
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Bo Yu
- Department of Dermatology, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| |
Collapse
|
15
|
Choi SH, Lee HY, Yun SH, Jang SJ, Kim SU, Park JY, Ahn SH, Kim DY. Identification of new biomarkers of hepatic cancer stem cells through proteomic profiling. JOURNAL OF LIVER CANCER 2025; 25:123-133. [PMID: 40108769 PMCID: PMC12010825 DOI: 10.17998/jlc.2025.03.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUNDS/AIMS In hepatocellular carcinoma (HCC), which exhibits high mortality and recurrence rates globally, the traits of cancer stem cells (CSCs) that significantly influence recurrence and metastasis are not well understood. CSCs are self-renewing cell types identified in most liquid and solid cancers, contributing to tumor initiation, growth, resistance, recurrence, and metastasis following chemo-radiotherapy or trans-arterial chemoembolization therapy. METHODS CSCs are classified based on the expression of cell surface markers such as CD133, which varies depending on the tumor type. Proteomic analysis of liver cancer cell lines with cancer stem cell potential and HCC cancer cell lines lacking stem cell propensity was conducted to compare and analyze specific expression patterns. RESULTS Proteomic profiling and enrichment analysis revealed higher expression of the calcium-binding protein S100 family in CD133+ Huh7 cells than in CD133- or wild-type cells. Furthermore, elevated expression of S100 family members was confirmed in an actual CD133+ liver cancer cell line via protein-protein network analysis and quantitative polymerase chain reaction (qPCR). CONCLUSION The S100 family members are not only new markers of cancer stem cells but will also assist in identifying new treatment strategies for CSC metastasis and tumor advancement.
Collapse
Affiliation(s)
- Sung Hoon Choi
- Institute of Health & Environment, Seoul National University, Seoul, Korea
- KoBioLabs Inc., Seoul, Korea
| | - Ha Young Lee
- Digital Omics Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute, Cheongju, Korea
| | - Sung Ho Yun
- Digital Omics Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute, Cheongju, Korea
| | - Sung Jae Jang
- Institute of Health & Environment, Seoul National University, Seoul, Korea
- Department of Environmental Health Sciences, Seoul National University Graduate School of Public Health, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul, Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul, Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul, Korea
| |
Collapse
|
16
|
Xu DM, Chen LX, Han H, Mo M. Single-cell and spatial transcriptomics reveal pre-metastatic subsets and therapeutic targets in penile carcinoma. iScience 2025; 28:111765. [PMID: 39925432 PMCID: PMC11804784 DOI: 10.1016/j.isci.2025.111765] [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: 07/15/2024] [Revised: 09/28/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Tumor heterogeneity, driven by branching evolution and genomic mutations, complicates cancer treatment. Understanding malignant cell evolution across various tumors aids in identifying pre-metastatic subpopulations for optimized therapies. Using bulk RNA sequencing (6 primary penile carcinomas, 6 metastatic lymph nodes, GSE196978), single-cell RNA sequencing (4 advanced penile carcinomas), spatial transcriptomics (Squamous cell carcinoma [SCC]: GSE144239-GSM4565823 and SCC: GSE144239-GSM4565826), and cell assays with Silmitasertib, we mapped heterogeneity and pinpointed therapeutic targets. In penile carcinoma, we discovered an MMP3+SPP1+ pre-metastatic subset and casein kinase 2 alpha 1 (CK2α) overexpression. The nuclear factor κB (NF-κB) pathway may drive metastasis. Pan-cancer analysis showed that MMP3 and SPP1 link to epithelial mesenchymal transition (EMT) and drug resistance, while CK2α activates oncogenes. Silmitasertib, a CK2α inhibitor, exhibited anti-tumor effects in penile carcinoma cells. Validated across 98 single-cell and 6 spatial datasets, our study advances the understanding of tumorigenesis and metastasis, highlighting Silmitasertib as a potential therapeutic agent.
Collapse
Affiliation(s)
- Da-Ming Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Ling-Xiao Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Hui Han
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| |
Collapse
|
17
|
Lu X, Du W, Zhou J, Li W, Fu Z, Ye Z, Chen G, Huang X, Guo Y, Liao J. Integrated genomic analysis of the stemness index signature of mRNA expression predicts lung adenocarcinoma prognosis and immune landscape. PeerJ 2025; 13:e18945. [PMID: 39959839 PMCID: PMC11830367 DOI: 10.7717/peerj.18945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
mRNA expression-based stemness index (mRNAsi) has been used for prognostic assessment in various cancers, but its application in lung adenocarcinoma (LUAD) is limited, which is the focus of this study. Low mRNAsi in LUAD predicted a better prognosis. Eight genes (GNG7, EIF5A, ANLN, FKBP4, GAPDH, GNPNAT1, E2F7, CISH) associated with mRNAsi were screened to establish a risk model. The differentially expressed genes between the high and low risk groups were mainly enriched in the metabolism, cell cycle functions pathway. The low risk score group had higher immune cell scores. Patients with lower TIDE scores in the low risk group had better immunotherapy outcomes. In addition, risk score was effective in assessing drug sensitivity of LUAD. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) data showed that eight genes were differentially expressed in LUAD cell lines, and knockdown of EIF5A reduced the invasion and migration ability of LUAD cells. This study designed a risk model based on the eight mRNAsi-related genes for predicting LUAD prognosis. The model accurately predicted the prognosis and survival of LUAD patients, facilitating the assessment of the sensitivity of patients to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xingzhao Lu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Wei Du
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jianping Zhou
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Weiyang Li
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhimin Fu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhibin Ye
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Guobiao Chen
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Xian Huang
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Yuliang Guo
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jingsheng Liao
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| |
Collapse
|
18
|
Zhao K, Sun T, Sun Q, Chen Z, Wang T, Yang J, Li L, Zhu Y, Liu X, Yang D, Lin B, Lu N. Nerve Growth Factor Signaling Promotes Nuclear Translocation of TRAF4 to Enhance Tumor Stemness and Metastatic Dormancy Via C-Jun-mediated IL-8 Autocrine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414437. [PMID: 39716976 PMCID: PMC11831473 DOI: 10.1002/advs.202414437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/03/2024] [Indexed: 12/25/2024]
Abstract
Tumor necrosis factor receptor-associated factor 4 (TRAF4), an E3 ubiquitin ligase, is frequently overexpressed in tumors. Although its cytoplasmic role in tumor progression is well-documented, the precise mechanisms underlying its nuclear localization and functional contributions in tumor cells remain elusive. This study demonstrated a positive correlation between the expression of nuclear TRAF4 and both tumor grades and stemness signatures in human cancer tissues. Notably, reduced nuclear TRAF4 led to decreased stemness properties and metastatic dormancy of tumor cells. Conversely, restoring nuclear TRAF4 in TRAF4-knockout (TRAF4-KO) cells augmented these cellular capabilities. Within the nucleus, the TRAF domain of TRAF4 interacted with c-Jun, thereby stimulating its transcriptional activity. This interaction subsequently led to an enhancement of the promoter activity of interleukin-8 (IL-8), which is identified as a mediator of nuclear TRAF4-induced tumor dormancy. Additionally, activation of AKT signaling by nerve growth factor facilitated TRAF4 phosphorylation at Ser242, enhancing its interaction with 14-3-3θ and promoting its nuclear translocation. Importantly, pharmacological modulation of TRAF4 nuclear translocation is found to suppress tumor tumorigenicity and metastasis in tumor models. This study highlights the critical role of nuclear TRAF4 in regulating tumor stemness and dormancy, positioning it as a potential therapeutic target for metastatic and refractory cancers.
Collapse
Affiliation(s)
- Kai Zhao
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Tifan Sun
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Qiruo Sun
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Zhenzhong Chen
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Tiepeng Wang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
- School of PharmacyNanjing University of Chinese Medicine138 Xianlin Rd.Nanjing210023China
| | - Jinming Yang
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
- Department of PharmacyThe Second Hospital of NanjingAffiliated Hospital to Nanjing University of Chinese MedicineNanjing210003China
| | - Lei Li
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yanan Zhu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Xinye Liu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| | - Dawei Yang
- Department of PharmacyThe Second Hospital of NanjingAffiliated Hospital to Nanjing University of Chinese MedicineNanjing210003China
| | - Binyan Lin
- School of PharmacyNanjing University of Chinese Medicine138 Xianlin Rd.Nanjing210023China
| | - Na Lu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of Carcinogenesis and InterventionDepartment of PhysiologySchool of Basic Medicine and Clinical PharmacyChina Pharmaceutical University24 TongjiaxiangNanjing210009China
| |
Collapse
|
19
|
Ragab EM, Gamal DME, El-Najjar FF, Elkomy HA, Ragab MA, Elantary MA, Basyouni OM, Moustafa SM, El-Naggar SA, Elsherbiny AS. New insights into Notch signaling as a crucial pathway of pancreatic cancer stem cell behavior by chrysin-polylactic acid-based nanocomposite. Discov Oncol 2025; 16:107. [PMID: 39891818 PMCID: PMC11787125 DOI: 10.1007/s12672-025-01846-3] [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: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Pancreatic cancer is an extremely deadly illness for which there are few reliable treatments. Recent research indicates that malignant tumors are highly variable and consist of a tiny subset of unique cancer cells, known as cancer stem cells (CSCs), which are responsible for the beginning and spread of tumors. These cells are typically identified by the expression of specific cell surface markers. A population of pancreatic cancer stem cells with aberrantly active developmental signaling pathways has been identified in recent studies of human pancreatic tumors. Among these Notch signaling pathway has been identified as a key regulator of CSCs self-renewal, making it an attractive target for therapeutic intervention. Chrysin-loaded polylactic acid (PLA) as polymeric nanoparticles systems have been growing interest in using as platforms for improved drug delivery. This review aims to explore innovative strategies for targeted therapy and optimized drug delivery in pancreatic CSCs by manipulating the Notch pathway and leveraging PLA-based drug delivery systems. Furthermore, we will assess the capability of PLA nanoparticles to enhance the bioavailability and effectiveness of gemcitabine in pancreatic cancer cells. The insights gained from this review have the potential to contribute to the development of novel treatment approaches that combine targeted therapy with advanced drug delivery utilizing biodegradable polymeric nanoparticles.
Collapse
Affiliation(s)
- Eman M Ragab
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Fares F El-Najjar
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hager A Elkomy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A Ragab
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mariam A Elantary
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Omar M Basyouni
- Chemistry/Zoology Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sherif M Moustafa
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa A El-Naggar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Abeer S Elsherbiny
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
20
|
Meerovich G, Kogan E, Romanishkin I, Zharkov N, Avraamova S, Shchelokova E, Akhlyustina E, Strakhovskaya M, Meerovich I, Demura S, Tiganova I, Romanova Y, Chen ZL, Reshetov I. Potential of photodynamic therapy using polycationic photosensitizers in the treatment of lung cancer patients with SARS-CoV-2 infection and bacterial complications: Our recent experience. Photodiagnosis Photodyn Ther 2025; 51:104447. [PMID: 39681293 DOI: 10.1016/j.pdpdt.2024.104447] [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: 09/30/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
The problem of treating cancer patients with lung cancer has become more difficult due to the SARS-CoV-2 viral infection and concomitant bacterial lesions. The analysis shows that the photodynamic effect of long-wavelength polycationic photosensitizers suppresses the tumor process (including the destruction of cancer stem cells), SARS-CoV-2 coronavirus infection, Gram-positive and Gram-negative bacteria, including those that can cause pneumonia. Therefore, the photodynamic approach using such photosensitizers is promising for the development of an effective treatment method for patients with lung cancer, including those with SARS-CoV-2 infection and bacterial complications.
Collapse
Affiliation(s)
- Gennady Meerovich
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia; Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | - Evgeniya Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Igor Romanishkin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia.
| | - Nikolay Zharkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Sofiya Avraamova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Elena Shchelokova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Ekaterina Akhlyustina
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | | | - Irina Meerovich
- Research Center of Biotechnology of the Russian Academy of Sciences, A.N. Bach Institute of Biochemistry, Moscow 119071, Russia
| | - Sofya Demura
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | - Irina Tiganova
- Lomonosov Moscow State University, Moscow 119991, Russia
| | - Yulia Romanova
- Lomonosov Moscow State University, Moscow 119991, Russia
| | - Zhi-Long Chen
- Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Igor Reshetov
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| |
Collapse
|
21
|
Muniyan S, Vengoji R, Nimmakayala RK, Seshacharyulu P, Perumalsamy B, Alsafwani ZW, Kakar SS, Smith LM, Shonka N, Teply BA, Lele SM, Ponnusamy MP, Batra SK. PAF1-mediated transcriptional reprogramming confers docetaxel resistance in advanced prostate cancer. Cancer Lett 2025; 609:217355. [PMID: 39603380 PMCID: PMC11912633 DOI: 10.1016/j.canlet.2024.217355] [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: 03/21/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Advanced prostate cancer (PCa) remains a significant clinical challenge, and docetaxel plays a significant role in disease management. Despite the efficacy of docetaxel as a first-line chemotherapy, resistance often develops. We developed three clinically relevant in vitro PCa cell models and transcriptomic analysis identified that the Paf1/RNA polymerase II complex component (PAF1)-associated pluripotent-transcription factor (TF), SOX2, plays a crucial role in docetaxel resistance. The cancer stem cell (CSC) transcriptional master regulator PAF1 is significantly higher in PCa cell lines, tumor tissues, and docetaxel resistant (DR) PCa cells than in age-matched control cells. To determine the molecular underlying and functional characteristics of PAF1 in resistance mechanisms, we performed coimmunoprecipitation, embryonic stem cell network proteins, in vitro tumor-initiating ability, and 3D multicellular organoid growth using PAF1 knockdown cells. Tet-inducible PAF1 depletion reduced the drug-efflux phenotype, tumor-initiating frequencies, and three-dimensional organoid growth of the docetaxel-resistant PCa cell lines. Functional studies also showed restoration of docetaxel sensitivity in a 3D tumorsphere model upon PAF1 depletion. PAF1 depletion was also associated with decreased pluripotent TFs and other CSC markers. This study provides a novel regulatory mechanism of docetaxel resistance in PCa through PAF1.
Collapse
Affiliation(s)
- Sakthivel Muniyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Balaji Perumalsamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Zahraa Wajih Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sham S Kakar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lynette M Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nicole Shonka
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benjamin A Teply
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Subodh M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
22
|
Gangadaran P, Onkar A, Rajendran RL, Goenka A, Oh JM, Khan F, Nagarajan AK, Muthu S, Krishnan A, Hong CM, Ahn BC. Noninvasive in vivo imaging of macrophages: understanding tumor microenvironments and delivery of therapeutics. Biomark Res 2025; 13:20. [PMID: 39865337 PMCID: PMC11770947 DOI: 10.1186/s40364-025-00735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Macrophages are pivotal in the body's defense and response to inflammation. They are present in significant numbers and are widely implicated in various diseases, including cancer. While molecular and histological techniques have advanced our understanding of macrophage biology, their precise function within the cancerous microenvironments remains underexplored. Enhancing our knowledge of macrophages and the dynamics of their extracellular vesicles (EVs) in cancer development can potentially improve therapeutic management. Notably, macrophages have also been harnessed to deliver drugs. Noninvasive in vivo molecular imaging of macrophages is crucial for investigating intricate cellular processes, comprehending the underlying mechanisms of diseases, tracking cells and EVs' migration, and devising macrophage-dependent drug-delivery systems in living organisms. Thus, in vivo imaging of macrophages has become an indispensable tool in biomedical research. The integration of multimodal imaging approaches and the continued development of novel contrast agents hold promise for overcoming current limitations and expanding the applications of macrophage imaging. This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach. The review concludes by offering insights into the applicability of molecular imaging techniques for real time monitoring of macrophages in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ramya Lakshmi Rajendran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - ArulJothi Kandasamy Nagarajan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, 603203, Tamilnadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Tamil Nadu, 639004, Karur, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Tamil Nadu, 641021, Coimbatore, India
| | - Anand Krishnan
- Precision Medicine and Integrated Nano-Diagnostics (P-MIND) Research Group, Office of the Dean, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Korea.
| |
Collapse
|
23
|
Luan F, Cui Y, Huang R, Yang Z, Qiao S. Comprehensive pan-cancer analysis reveals NTN1 as an immune infiltrate risk factor and its potential prognostic value in SKCM. Sci Rep 2025; 15:3223. [PMID: 39863609 PMCID: PMC11762998 DOI: 10.1038/s41598-025-85444-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Netrin-1 (NTN1) is a laminin-related secreted protein involved in axon guidance and cell migration. Previous research has established a significant connection between NTN1 and nervous system development. In recent years, mounting evidence indicates that NTN1 also plays a crucial role in tumorigenesis and tumor progression. For instance, inhibiting Netrin-1 has been shown to suppress tumor growth and epithelial-mesenchymal transition (EMT) characteristics in endometrial cancer. To further elucidate the influence of genes on tumors, we utilized a variety of machine learning techniques and found that NTN1 is strongly linked to multiple cancer types, suggesting it as a potential therapeutic target. This study aimed to elucidate the role of NTN1 in pan-cancer using multi-omics data and explore its potential as a prognostic biomarker in SKCM. Analysis of the TCGA, GTEx, and UALCAN databases revealed significant differences in NTN1 expression at both the mRNA and protein levels. Prognostic value was evaluated through univariate Cox regression and Kaplan-Meier methods. Mutation and methylation analyses were conducted using the cBioPortal and SMART databases. We identified genes interacting with and correlated to NTN1 through STRING and GEPIA2, respectively. Subsequently, we performed GO and KEGG enrichment analyses. The results suggested that NTN1 might be involved in crucial biological processes and pathways related to cancer development and progression, including cell adhesion, axon guidance, immune response, and various signaling pathways. We then explored the correlation between NTN1 and immune infiltration as well as immunotherapy using the ESTIMATE package, TIMER2.0, TISIDB, TIDE, TIMSO, and TCIA. The relationship between NTN1 and tumor heterogeneity, stemness, DNA methyltransferases, and MMR genes was also examined. Lastly, we constructed a nomogram based on NTN1 in SKCM and investigated its association with drug sensitivity. NTN1 expression was significantly associated with tumor immune infiltration, molecular subtypes, and clinicopathological features in various cancers. Genetic analysis revealed that Deep deletions were the most common type of NTN1 alteration. Additionally, a positive correlation was observed between NTN1 CNAs and its expression levels. In most cancers, NTN1 showed positive correlations with immune and stromal scores, as well as with specific immune cell populations. Its predictive value for immunotherapy response was comparable to that of tumor mutational burden. Furthermore, NTN1 exhibited positive correlations with tumor heterogeneity, stemness, DNA methyltransferase genes, and MMR genes. In SKCM, NTN1 was identified as an independent risk factor and demonstrated potential associations with multiple drugs. NTN1 exhibits substantial clinical utility as a prognostic marker and indicator of immune response across various tumor types. This comprehensive analysis provides insights into its potential implications in pan-cancer research.
Collapse
Affiliation(s)
- Fuxiang Luan
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Yuying Cui
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Ruizhe Huang
- The First Clinical College of Changsha Medical University, Changsha, China
| | - Zhuojie Yang
- Academy of medical sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Shishi Qiao
- The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
24
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
25
|
Huang T, Fan L, Tang J, Chen S, Du G, Zhang N. Advances in research on the carcinogenic mechanisms and therapeutic potential of YAP1 in bladder cancer (Review). Oncol Rep 2025; 53:10. [PMID: 39540392 PMCID: PMC11599795 DOI: 10.3892/or.2024.8843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Bladder cancer is the most common malignant tumor of the urinary system with high morbidity and no clear pathogenesis. The Hippo signaling pathway is an evolutionarily conserved pathway that regulates organ size and maintains tissue homeostasis. Yes‑associated protein 1 (YAP1) is a key effector of this pathway and regulates downstream target genes by binding to transcriptional co‑activators with PDZ binding sequences (TAZ). Several studies have demonstrated that YAP1 is overexpressed in bladder cancer and is involved in adverse outcomes such as bladder cancer occurrence, progression, resistance to cisplatin and the recurrence of tumours. The present review summarized the involvement of YAP1 in bladder cancer disease onset and progression, and the mechanism of YAP1 involvement in bladder cancer treatment. In addition, this study further explored the potential of YAP1 in the diagnosis and treatment of bladder cancer. This study aimed to explore the potential mechanism of YAP1 in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Tianyu Huang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Longmei Fan
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiajia Tang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Shicheng Chen
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guotu Du
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Neng Zhang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
26
|
Yang J, Wang H, Li B, Liu J, Zhang X, Wang Y, Peng J, Gao L, Wang X, Hu S, Zhang W, Hong L. Inhibition of ACSS2 triggers glycolysis inhibition and nuclear translocation to activate SIRT1/ATG5/ATG2B deacetylation axis, promoting autophagy and reducing malignancy and chemoresistance in ovarian cancer. Metabolism 2025; 162:156041. [PMID: 39362518 DOI: 10.1016/j.metabol.2024.156041] [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: 07/05/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Metabolic reprogramming is a hallmark of cancer, characterized by a high dependence on glycolysis and an enhanced utilization of acetate as an alternative carbon source. ACSS2 is a critical regulator of acetate metabolism, playing a significant role in the development and progression of various malignancies. ACSS2 facilitates the conversion of acetate to acetyl-CoA, which participates in multiple metabolic pathways and functions as an epigenetic regulator of protein acetylation, thereby modulating key cellular processes such as autophagy. However, the roles and intrinsic connections of ACSS2, glycolysis, protein acetylation, and autophagy in ovarian cancer (OC) remain to be elucidated. BASIC PROCEDURES Utilizing clinical specimens and online databases, we analysed the expression of ACSS2 in OC and its relationship with clinical prognosis. By knocking down ACSS2, we evaluated its effects on the malignant phenotype, acetate metabolism, glycolysis, and autophagy. The metabolic alterations in OC cells were comprehensively analysed using Seahorse assays, transmission electron microscopy, membrane potential measurements, and stable-isotope labeling techniques. CUT&TAG and co-immunoprecipitation techniques were employed to explore the deacetylation of autophagy-related proteins mediated by ACSS2 via SIRT1. Additionally, through molecular docking, transcriptome sequencing, and metabolomics analyses, we validated the pharmacological effects of paeonol on ACSS2 and the glycolytic process in OC cells. Finally, both in vitro and in vivo experiments were performed to investigate the impact of paeonol on autophagy and its anti-OC effects mediated through the ACSS2/SIRT1 deacetylation axis. MAIN FINDINGS ACSS2 is significantly upregulated in OC and is associated with poor prognosis. Knockdown of ACSS2 inhibits OC cells proliferation, migration, invasion, angiogenesis, and platinum resistance, while reducing tumour burden in vivo. Mechanistically, inhibiting ACSS2 reduces acetate metabolism and suppresses glycolysis by targeting HXK2. This glycolytic reduction promotes the translocation of ACSS2 from the cytoplasm to the nucleus, leading to increased expression of the deacetylase SIRT1. SIRT1 mediates the deacetylation of autophagy-related proteins, such as ATG5 and ATG2B, thereby significantly activating autophagy in OC cells and exerting antitumor effects. Paeonol inhibits acetate metabolism and glycolysis in OC cells by targeting ACSS2. Paeonol activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation axis, demonstrating inhibition of OC in vitro and in vivo. PRINCIPAL CONCLUSIONS Pae can serve as an effective, low-toxicity, multi-targeted drug targeting ACSS2 and glycolysis. It activates autophagy through the ACSS2/SIRT1/ATG5/ATG2B deacetylation signalling cascade, thereby exerting anti-OC effects. Our study provides new insights into the malignant mechanisms of OC and offers a novel strategy for its treatment.
Collapse
Affiliation(s)
- Jiang Yang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China; Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, PR China
| | - Haoyu Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Bingshu Li
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jingchun Liu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xiaoyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Ying Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Jiaxin Peng
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Likun Gao
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Xinqi Wang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Siyuan Hu
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Wenyi Zhang
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Li Hong
- Department of Obstetrics and Gynaecology, Renmin Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
27
|
Yu Y, Lu XH, Mu JS, Meng JY, Sun JS, Chen HX, Yan Y, Meng K. N6-methyladenosine-modified long non-coding RNA KIF9-AS1 promotes stemness and sorafenib resistance in hepatocellular carcinoma by upregulating SHOX2 expression. World J Gastroenterol 2024; 30:5174-5190. [PMID: 39735272 PMCID: PMC11612700 DOI: 10.3748/wjg.v30.i48.5174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/26/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevalent and aggressive tumor. Sorafenib is the first-line treatment for patients with advanced HCC, but resistance to sorafenib has become a significant challenge in this therapy. Cancer stem cells play a crucial role in sorafenib resistance in HCC. Our previous study revealed that the long non-coding RNA (lncRNA) KIF9-AS1 is an oncogenic gene in HCC. However, the role of KIF9-AS1 in drug resistance and cancer stemness in HCC remains unclear. Herein, we aimed to investigate the function and mechanism of the lncRNA KIF9-AS1 in cancer stemness and drug resistance in HCC. AIM To describe the role of the lncRNA KIF9-AS1 in cancer stemness and drug resistance in HCC and elucidate the underlying mechanism. METHODS Tumor tissue and adjacent non-cancerous tissue samples were collected from HCC patients. Sphere formation was quantified via a tumor sphere assay. Cell viability, proliferation, and apoptosis were evaluated via Cell Counting Kit-8, flow cytometry, and colony formation assays, respectively. The interactions between the lncRNA KIF9-AS1 and its downstream targets were confirmed via RNA immunoprecipitation and coimmunoprecipitation. The tumorigenic role of KIF9-AS1 was validated in a mouse model. RESULTS Compared with that in normal controls, the expression of the lncRNA KIF9-AS1 was upregulated in HCC tissues. Knockdown of KIF9-AS1 inhibited stemness and attenuated sorafenib resistance in HCC cells. Mechanistically, N6-methyladenosine modification mediated by methyltransferase-like 3/insulin-like growth factor 2 mRNA-binding protein 1 stabilized and increased the expression of KIF9-AS1. Additionally, KIF9-AS1 increased the stability and expression of short stature homeobox 2 by promoting ubiquitin-specific peptidase 1-induced deubiquitination. Furthermore, depletion of KIF9-AS1 alleviated sorafenib resistance in a xenograft mouse model of HCC. CONCLUSION The N6-methyladenosine-modified lncRNA KIF9-AS1 promoted stemness and sorafenib resistance in HCC by upregulating short stature homeobox 2 expression.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Drug Resistance, Neoplasm/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Animals
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Mice
- Gene Expression Regulation, Neoplastic
- Up-Regulation
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Adenosine/pharmacology
- Cell Line, Tumor
- Male
- Cell Proliferation/drug effects
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Xenograft Model Antitumor Assays
- Apoptosis/drug effects
- Mice, Nude
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Female
Collapse
Affiliation(s)
- Yong Yu
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Xiang-Hong Lu
- Department of Intensive Care Medicine, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Jin-Song Mu
- Department of Intensive Care Medicine, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100039, China
| | - Jiang-Yun Meng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Jiang-Shan Sun
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Hai-Xu Chen
- Institute of Geriatrics and National Clinical Research Center of Geriatrics Disease, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Yang Yan
- Department of General Surgery, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| | - Ke Meng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
| |
Collapse
|
28
|
Hsu CY, Hisham Ateya N, Felix Oghenemaro E, Nathiya D, Kaur P, Hjazi A, Eldesoqui M, Yumashev A, Kadhim Abosaoda M, Adnan Abdulrahman M. Correlation between lncRNAs with human molecular chaperons in cancer immunopathogenesis and drug resistance. Int Immunopharmacol 2024; 143:113309. [PMID: 39405942 DOI: 10.1016/j.intimp.2024.113309] [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: 07/14/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/30/2024]
Abstract
The development of cancer immunology heavily relies on the interaction between long non-coding RNAs (lncRNAs) and molecular chaperones. By participating in gene regulation, lncRNAs interact with molecular chaperones, which play a critical role in protein folding and stress responses, to influence oncogenic pathways. This interaction has an impact on both the immune cells within the tumor microenvironment and the tumor cells themselves. Understanding these mechanisms provides valuable insights into innovative approaches for diagnosis and treatment. Targeting the lncRNA-chaperone axis has the potential to strengthen anti-tumor immunity and enhance cancer treatment outcomes. Further research is necessary to uncover specific associations, identify biomarkers, and develop personalized therapies aimed at disrupting this axis, which could potentially revolutionize cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ 85004, USA.
| | - Nabaa Hisham Ateya
- Biotechnology Department, College of Applied Science, Fallujah University, Iraq.
| | - Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Abraka, Delta State, Nigeria.
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India.
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia.
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Doctor of Medicine, Professor. Sechenov First Moscow State Medical University, Russia.
| | - Munther Kadhim Abosaoda
- College of Pharmacy, the Islamic University, Najaf, Iraq; College of Pharmacy, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of Pharmacy, the Islamic University of Babylon, Babylon, Iraq.
| | | |
Collapse
|
29
|
Li Z, Wang S, Guo T, Yan X, Chen C, Zhang W, Zhao J, Zhang J, Zhao S, Wang Y, Qi Y, Gu C. MACC1 ablation suppresses the dedifferentiation process of non-CSCs in lung cancer through stabilizing KLF4. Cell Death Discov 2024; 10:494. [PMID: 39695175 DOI: 10.1038/s41420-024-02256-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Metastasis-associated in colon cancer-1 (MACC1) was identified as a new player in lung cancer development, and some stemness-related genes can be novel transcriptional targets of MACC1. Cancer stem cells (CSCs) are responsible for sustaining tumorigenesis and plasticity. Both CSCs and non-CSCs are plastic and capable of undergoing phenotypic transition, especially the dedifferentiation of non-CSCs switch to CSC-like cells. However, the precise role of MACC1 during this process is largely unknown. Here, we showed that MACC1 promoted the transition from non-CSC to CSC in lung cancer. We found MACC1 was overexpressed in stemness enriched cells, enhancing the transition from no-CSCs to CSCs, while short-hairpin RNA-mediated Knockdown of MACC1 impaired this process. High-throughput sequencing and tumor specimen analysis revealed that MACC1 was negative correlated with Krüppel-like factor 4 (KLF4) expression level, which acts as a negative stemness regulator in lung cancer. Mechanistically, MACC1 delays the degradation of KLF4 mRNA by repressing the expression of microRNA-25, thereby promoting the KLF4 mRNA stabilization at the post-transcriptional level. Collectively, our findings may facilitate efforts to promote the development of precision targeted therapy for cancer stem cells in lung cancer.
Collapse
Affiliation(s)
- Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Shiqing Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Xinyi Yan
- Department of Nephrology, Dalian Municipal Central Hospital, Dalian University of Technology, Dalian, 116033, China
| | - Chaoqun Chen
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Wenjing Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Jinyao Zhao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Jinrui Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Yang Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Yangfan Qi
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China.
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116044, China.
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China.
| |
Collapse
|
30
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
31
|
Masciale V, Banchelli F, Grisendi G, Samarelli AV, Raineri G, Rossi T, Zanoni M, Cortesi M, Bandini S, Ulivi P, Martinelli G, Stella F, Dominici M, Aramini B. The molecular features of lung cancer stem cells in dedifferentiation process-driven epigenetic alterations. J Biol Chem 2024; 300:107994. [PMID: 39547513 PMCID: PMC11714729 DOI: 10.1016/j.jbc.2024.107994] [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: 03/23/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Cancer stem cells (CSCs) may be dedifferentiated somatic cells following oncogenic processes, representing a subpopulation of cells able to promote tumor growth with their capacities for proliferation and self-renewal, inducing lineage heterogeneity, which may be a main cause of resistance to therapies. It has been shown that the "less differentiated process" may have an impact on tumor plasticity, particularly when non-CSCs may dedifferentiate and become CSC-like. Bidirectional interconversion between CSCs and non-CSCs has been reported in other solid tumors, where the inflammatory stroma promotes cell reprogramming by enhancing Wnt signaling through nuclear factor kappa B activation in association with intracellular signaling, which may induce cells' pluripotency, the oncogenic transformation can be considered another important aspect in the acquisition of "new" development programs with oncogenic features. During cell reprogramming, mutations represent an initial step toward dedifferentiation, in which tumor cells switch from a partially or terminally differentiated stage to a less differentiated stage that is mainly manifested by re-entry into the cell cycle, acquisition of a stem cell-like phenotype, and expression of stem cell markers. This phenomenon typically shows up as a change in the form, function, and pattern of gene and protein expression, and more specifically, in CSCs. This review would highlight the main epigenetic alterations, major signaling pathways and driver mutations in which CSCs, in tumors and specifically, in lung cancer, could be involved, acting as key elements in the differentiation/dedifferentiation process. This would highlight the main molecular mechanisms which need to be considered for more tailored therapies.
Collapse
Affiliation(s)
- Valentina Masciale
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Federico Banchelli
- Department of Statistical Sciences "Paolo Fortunati", Alma Mater Studiorum- University of Bologna, Bologna, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Franco Stella
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy; Division of Oncology, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy.
| |
Collapse
|
32
|
Sher EK, Kalić A, Džidić-Krivić A, Zećo MB, Pinjić E, Sher F. Cellular therapeutic potential of genetically engineered stem cells in cancer treatment. Biotechnol Genet Eng Rev 2024; 40:4062-4097. [PMID: 37132363 DOI: 10.1080/02648725.2023.2204720] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/13/2023] [Indexed: 05/04/2023]
Abstract
Traditional therapeutic approaches in the treatment of cancer have many side effects and are often ineffective and non-specific, leading to the development of therapy-resistant tumour cells. Recently, numerous discoveries about stem cells have given a new outlook on their application in oncology. Stem cells are unique because of their biological attributes, including self-renewal, differentiation in different types of specialized cells and synthesis of molecules that interplay with tumour niche. They are already used as an effective therapeutic option for haematological malignancies, such as multiple myeloma and leukaemia. The main goal of this study is to investigate the possible applications of different types of stem cells in cancer treatment and to summarize novel advances, as well as the limitations of their application in cancer treatment. Research and clinical trials that are underway revealed and confirmed the enormous potential of regenerative medicine in the treatment of cancer, especially when combined with different nanomaterials. Nanoengineering of stem cells has been the focus of novel studies in the area of regenerative medicine, such as the production of nanoshells and nanocarriers that enhance the transport and uptake of stem cells in their targeted tumour niche and enable the effective monitoring of stem cell effects on tumour cells. Although nanotechnology has a lot of limitations, it provides new opportunities for the development of effective and innovative stem cell therapies.
Collapse
Affiliation(s)
- Emina Karahmet Sher
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Azra Kalić
- Faculty of pharmacy, University of modern sciences - CKM, Mostar, Bosnia and Herzegovina
| | - Amina Džidić-Krivić
- International Society of Engineering Science and Technology, Nottingham, UK
- Department of Neurology, Cantonal Hospital Zenica, Zenica, Bosnia and Herzegovina
| | - Merima Beća- Zećo
- Faculty of pharmacy, University of modern sciences - CKM, Mostar, Bosnia and Herzegovina
- International Society of Engineering Science and Technology, Nottingham, UK
| | - Emma Pinjić
- Department of Radiology, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
| | - Farooq Sher
- Department of Engineering, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
33
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
34
|
Li Q, He G, Yu Y, Li X, Peng X, Yang L. Exosome crosstalk between cancer stem cells and tumor microenvironment: cancer progression and therapeutic strategies. Stem Cell Res Ther 2024; 15:449. [PMID: 39578849 PMCID: PMC11583673 DOI: 10.1186/s13287-024-04061-z] [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: 09/27/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024] Open
Abstract
Cancer stem cells (CSCs) represent a small yet pivotal subset of tumor cells endowed with self-renewal capabilities. These cells are intricately linked to tumor progression and are central to drug resistance, metastasis, and recurrence. The tumor microenvironment (TME) encompasses the cancer cells and their surrounding milieu, including immune and inflammatory cells, cancer-associated fibroblasts, adjacent stromal tissues, tumor vasculature, and a variety of cytokines and chemokines. Within the TME, cells such as immune and inflammatory cells, endothelial cells, adipocytes, and fibroblasts release growth factors, cytokines, chemokines, and exosomes, which can either sustain or disrupt CSCs, thereby influencing tumor progression. Conversely, CSCs can also secrete cytokines, chemokines, and exosomes, affecting various components of the TME. Exosomes, a subset of extracellular vesicles (EVs), carry a complex cargo of nucleic acids, proteins, and lipids, playing a crucial role in the communication between CSCs and the TME. This review primarily focuses on the impact of exosomes secreted by CSCs (CSC-exo) on tumor progression, including their roles in maintaining stemness, promoting angiogenesis, facilitating metastasis, inducing immune suppression, and contributing to drug resistance. Additionally, we discuss how exosomes secreted by different cells within the TME affect CSCs. Finally, we explore the potential of utilizing exosomes to mitigate the detrimental effects of CSCs or to target and eliminate them. A thorough understanding of the exosome-mediated crosstalk between CSCs and the TME could provide valuable insights for developing targeted therapies against CSCs.
Collapse
Affiliation(s)
- Qi Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Yifan Yu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
35
|
Zhang J, Liu Z, Zhang Z, Yang H, Wang H, Yang Z, Xu Y, Li S, Yang D. Recent Advances in Silica-Based Nanomaterials for Enhanced Tumor Imaging and Therapy. ACS APPLIED BIO MATERIALS 2024; 7:7133-7169. [PMID: 39495482 DOI: 10.1021/acsabm.4c01318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Cancer remains a formidable challenge, inflicting profound physical, psychological, and financial burdens on patients. In this context, silica-based nanomaterials have garnered significant attention for their potential in tumor imaging and therapy owing to their exceptional properties, such as biocompatibility, customizable porosity, and versatile functionalization capabilities. This review meticulously examines the latest advancements in the application of silica-based nanomaterials for tumor imaging and therapy. It underscores their potential in enhancing various cancer imaging modalities, including fluorescence imaging, magnetic resonance imaging, computed tomography, positron emission tomography, ultrasound imaging, and multimodal imaging approaches. Moreover, the review delves into their therapeutic efficacy in chemotherapy, radiotherapy, phototherapy, immunotherapy, gas therapy, sonodynamic therapy, chemodynamic therapy, starvation therapy, and gene therapy. Critical evaluations of the biosafety profiles and degradation pathways of these nanomaterials within biological environments are also presented. By discussing the current challenges and prospects, this review aims to provide a nuanced perspective on the clinical translation of silica-based nanomaterials, thereby highlighting their promise in revolutionizing cancer diagnostics, enabling real-time monitoring of therapeutic responses, and advancing personalized medicine.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu 233030, China
| | - Zhenlu Yang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yunjian Xu
- School of Radiology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an 271000, China
- Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Shengke Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing 211816, China
| |
Collapse
|
36
|
Chen L, Rao W, Chen Y, Xie J. In vitro induction of anti‑lung cancer immune response by the A549 lung cancer stem cell lysate‑sensitized dendritic cell vaccine. Oncol Lett 2024; 28:550. [PMID: 39328277 PMCID: PMC11425031 DOI: 10.3892/ol.2024.14683] [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: 05/15/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Lung adenocarcinoma is one of the most fatal types of cancer worldwide, with non-small cell lung cancer being the most common subtype. Therefore, there is need for improved treatment approaches. Tumor growth results from the proliferation of a very small number of tumor stem cells, giving rise to the theory of cancer stem cells (CSCs). Lung CSCs are associated with lung cancer development, and although chemotherapy drugs can inhibit the proliferation of lung cancer cells, they have difficulty acting on lung CSCs. Even if the tumor appears to have disappeared after chemotherapy, the presence of a small number of residual tumor stem cells can lead to cancer recurrence and metastasis. Hence, targeting and eliminating lung CSCs is of significant therapeutic importance. In this study, we cultured A549 cells in sphere-forming conditions using B27, EGF, and bFGF, isolated peripheral blood mononuclear cells (PBMCs), and induced and characterized dendritic cells (DCs). We also isolated and expanded T lymphocytes. DC vaccines were prepared using A549 stem cell lysate or A549 cell lysate for sensitization and compared with non-sensitized DC vaccines. The content of IFN-γ in the supernatant of cultures with vaccines and T cells was measured by ELISA. The cytotoxic effects of the vaccines on A549 cells and stem cells were assessed using the Cytotox96 assay, and the impact of the vaccines on A549 cell migration and apoptosis was evaluated using Transwell assays and flow cytometry. DC vaccines sensitized with human lung CSC lysates induced significant in vitro cytotoxic effects on A549 lung cancer cells and CSCs by T lymphocytes, while not producing immune cytotoxic effects on human airway epithelial cells. Moreover, the immune-killing effect induced by DC vaccines sensitized with lung CSC lysates was superior to that of DC vaccines sensitized with lung cancer cells.
Collapse
Affiliation(s)
- Letian Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Wei Rao
- Department of Urology, Yingtan People's Hospital, Yingtan, Jiangxi 335000, P.R. China
| | - Yujuan Chen
- Department of Pulmonary and Critical Care Medicine, Gaoan People's Hospital, Yichun, Jiangxi 336000, P.R. China
| | - Junping Xie
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
37
|
Li F, Li Z, Wei C, Xu L, Liang Y, Yan J, Li Y, He B, Sun C. Application of hydrogels for targeting cancer stem cells in cancer treatment. Biomed Pharmacother 2024; 180:117486. [PMID: 39321506 DOI: 10.1016/j.biopha.2024.117486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024] Open
Abstract
Cancer stem cells (CSCs) are a major hindrance to clinical cancer treatment. Owing to their high tumorigenic and metastatic potential, CSCs are vital in malignant tumor initiation, growth, metastasis, and therapeutic resistance, leading to tumorigenesis and recurrence. Compared with normal tumor cells, CSCs express high levels of surface markers (CD44, CD90, CD133, etc.) and activate specific signaling pathways (Wnt/β-catenin, Notch, and Hedgehog). Although Current drug delivery systems (DDS) precisely target CSCs, the heterogeneity and multidrug resistance of CSCs impede CSC isolation and screening. Conversely, hydrogel DDSs exhibit good biocompatibility and high drug delivery efficiency. Hydrogels are three-dimensional (3D) spatial structures for drug encapsulation that facilitate the controlled release of bioactive molecules. Hence, hydrogels can be loaded with drugs to precisely target CSCs. Their 3D structure can also culture non-CSCs and facilitate their transformation into CSCs. for identification and isolation. Given that their elastic modulus and stiffness characteristics reflect those of the cellular microenvironment, hydrogels can simulate extracellular matrix pathways and markers to regulate CSCs, disrupting the equilibrium between CSC and non-CSC transformation. This article reviews the CSC microenvironment, metabolism, signaling pathway, and surface markers. Additionally, we summarize the existing CSC targeting strategies and explore the application of hydrogels for CSC screening and treatment. Finally, we discuss potential advances in CSC research that may lead to curative measures for tumors through targeted and precise attacks on CSCs.
Collapse
Affiliation(s)
- Fashun Li
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Chen Wei
- Department of Pharmacy, Qingdao Women and Children's Hospital, Qingdao 266034, China
| | - Long Xu
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Yifei Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Chong Sun
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
38
|
Chen C, Li P, Fan G, Yang E, Jing S, Shi Y, Gong Y, Zhang L, Wang Z. Role of TRIP13 in human cancer development. Mol Biol Rep 2024; 51:1088. [PMID: 39436503 DOI: 10.1007/s11033-024-10012-x] [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: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024]
Abstract
As an AAA + ATPase, thyroid hormone receptor interacting protein 13 (TRIP13) primarily functions in DNA double-strand break repair, chromosome recombination, and cell cycle checkpoint regulation; aberrant expression of TRIP13 can result in chromosomal instability (CIN). According to recent research, TRIP13 is aberrantly expressed in a variety of cancers, and a patient's poor prognosis and tumor stage are strongly correlated with high expression of TRIP13. Tumor cell and subcutaneous xenograft growth can be markedly inhibited by TRIP13 knockdown or TRIP13 inhibitor administration. In the initiation and advancement of human malignancies, TRIP13 seems to function as an oncogene. Based on available data, TRIP13 may function as a biological target and biomarker for cancer. The creation of inhibitors that specifically target TRIP13 may present novel approaches to treating cancer.
Collapse
Affiliation(s)
- Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China.
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China.
| |
Collapse
|
39
|
Zhou W, Su M, Jiang T, Xie Y, Shi J, Ma Y, Xu K, Xu G, Li Y, Xu J. Cancer Stemness Online: A Resource for Investigating Cancer Stemness and Associations with Immune Response. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae058. [PMID: 39141443 PMCID: PMC11522875 DOI: 10.1093/gpbjnl/qzae058] [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: 07/04/2023] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Cancer progression involves the gradual loss of a differentiated phenotype and the acquisition of progenitor and stem cell-like features, which are potential culprits of immunotherapy resistance. Although the state-of-the-art predictive computational methods have facilitated the prediction of cancer stemness, there remains a lack of efficient resources to accommodate various usage requirements. Here, we present the Cancer Stemness Online, an integrated resource for efficiently scoring cancer stemness potential at both bulk and single-cell levels. This resource integrates eight robust predictive algorithms as well as 27 signature gene sets associated with cancer stemness for predicting stemness scores. Downstream analyses were performed from five distinct aspects: identifying the signature genes of cancer stemness; exploring the associations with cancer hallmarks and cellular states; exploring the associations with immune response and the communications with immune cells; investigating the contributions to patient survival; and performing a robustness analysis of cancer stemness among different methods. Moreover, the pre-calculated cancer stemness atlas for more than 40 cancer types can be accessed by users. Both the tables and diverse visualizations of the analytical results are available for download. Together, Cancer Stemness Online is a powerful resource for scoring cancer stemness and expanding downstream functional interpretation, including immune response and cancer hallmarks. Cancer Stemness Online is freely accessible at http://bio-bigdata.hrbmu.edu.cn/CancerStemnessOnline.
Collapse
Affiliation(s)
- Weiwei Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Minghai Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Tiantongfei Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yunjin Xie
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Jingyi Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yingying Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Kang Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Gang Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
40
|
Li Y, Lin C, Chu Y, Wei Z, Ding Q, Gu S, Deng H, Liao Q, Shen Z. Characterization of Cancer Stem Cells in Laryngeal Squamous Cell Carcinoma by Single-cell RNA Sequencing. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae056. [PMID: 39107908 PMCID: PMC11522873 DOI: 10.1093/gpbjnl/qzae056] [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: 01/04/2023] [Revised: 04/03/2024] [Accepted: 07/23/2024] [Indexed: 11/01/2024]
Abstract
Cancer stem cells (CSCs) constitute a pivotal element within the tumor microenvironment (TME), driving the initiation and progression of cancer. However, the identification of CSCs and their underlying molecular mechanisms in laryngeal squamous cell carcinoma (LSCC) remains a formidable challenge. Here, we employed single-cell RNA sequencing of matched primary tumor tissues, paracancerous tissues, and local lymph nodes from three LSCC patients to comprehensively characterize the CSCs in LSCC. Two distinct clusters of stem cells originating from epithelial populations were delineated and verified as CSCs and normal stem cells (NSCs), respectively. CSCs were abundant in the paracancerous tissues compared to those in the tumor tissues. CSCs showed high expression of stem cell marker genes such as PROM1, ALDH1A1, and SOX4, and increased the activity of tumor-related hypoxia, Wnt/β-catenin, and Notch signaling pathways. We then explored the intricate crosstalk between CSCs and the TME cells and identified targets within the TME that related with CSCs. We also found eight marker genes of CSCs that were correlated significantly with the prognosis of LSCC patients. Furthermore, bioinformatics analyses showed that drugs such as erlotinib, OSI-027, and ibrutinib selectively targeted the CSC-specifically expressed genes. In conclusion, our results represent the first comprehensive characterization of CSC properties in LSCC at the single-cell level.
Collapse
Affiliation(s)
- Yanguo Li
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Chen Lin
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yidian Chu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zhengyu Wei
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Qi Ding
- The Ningbo Diagnostic Pathology Center, Ningbo 315021, China
| | - Shanshan Gu
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
| | - Hongxia Deng
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
| | - Qi Liao
- School of Public Health, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zhisen Shen
- The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
| |
Collapse
|
41
|
Niharika, Garg M. Understanding the autophagic functions in cancer stem cell maintenance and therapy resistance. Expert Rev Mol Med 2024; 26:e23. [PMID: 39375840 PMCID: PMC11488345 DOI: 10.1017/erm.2024.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/25/2023] [Accepted: 06/25/2024] [Indexed: 10/09/2024]
Abstract
Complex tumour ecosystem comprising tumour cells and its associated tumour microenvironment (TME) constantly influence the tumoural behaviour and ultimately impact therapy failure, disease progression, recurrence and poor overall survival of patients. Crosstalk between tumour cells and TME amplifies the complexity by creating metabolic changes such as hypoxic environment and nutrient fluctuations. These changes in TME initiate stem cell-like programmes in cancer cells, contribute to tumoural heterogeneity and increase tumour robustness. Recent studies demonstrate the multifaceted role of autophagy in promoting fibroblast production, stemness, cancer cell survival during longer periods of dormancy, eventual growth of metastatic disease and disease resistance. Recent ongoing studies examine autophagy/mitophagy as a powerful survival strategy in response to environmental stress including nutrient deprivation, hypoxia and environmental stress in TME. It prevents irreversible senescence, promotes dormant stem-like state, induces epithelial-mesenchymal transition and increases migratory and invasive potential of tumour cells. The present review discusses various theories and mechanisms behind the autophagy-dependent induction of cancer stem cell (CSC) phenotype. Given the role of autophagic functions in CSC aggressiveness and therapeutic resistance, various mechanisms and studies based on suppressing cellular plasticity by blocking autophagy as a powerful therapeutic strategy to kill tumour cells are discussed.
Collapse
Affiliation(s)
- Niharika
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
42
|
Klabukov I, Smirnova A, Yakimova A, Kabakov AE, Atiakshin D, Petrenko D, Shestakova VA, Sulina Y, Yatsenko E, Stepanenko VN, Ignatyuk M, Evstratova E, Krasheninnikov M, Sosin D, Baranovskii D, Ivanov S, Shegay P, Kaprin AD. Oncomatrix: Molecular Composition and Biomechanical Properties of the Extracellular Matrix in Human Tumors. JOURNAL OF MOLECULAR PATHOLOGY 2024; 5:437-453. [DOI: 10.3390/jmp5040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
The extracellular matrix is an organized three-dimensional network of protein-based molecules and other macromolecules that provide structural and biochemical support to tissues. Depending on its biochemical and structural properties, the extracellular matrix influences cell adhesion and signal transduction and, in general, can influence cell differentiation and proliferation through specific mechanisms of chemical and mechanical sensing. The development of body tissues during ontogenesis is accompanied by changes not only in cells but also in the composition and properties of the extracellular matrix. Similarly, tumor development in carcinogenesis is accompanied by a continuous change in the properties of the extracellular matrix of tumor cells, called ‘oncomatrix’, as the tumor matures, from the development of the primary focus to the stage of metastasis. In this paper, the characteristics of the composition and properties of the extracellular matrix of tumor tissues are considered, as well as changes to the composition and properties of the matrix during the evolution of the tumor and metastasis. The extracellular matrix patterns of tumor tissues can be used as biomarkers of oncological diseases as well as potential targets for promising anti-tumor therapies.
Collapse
Affiliation(s)
- Ilya Klabukov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anna Smirnova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Anna Yakimova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Alexander E. Kabakov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Dmitri Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Daria Petrenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Victoria A. Shestakova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Elena Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Vasiliy N. Stepanenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Michael Ignatyuk
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Evstratova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Michael Krasheninnikov
- Scientific and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Dmitry Sosin
- Center for Strategic Planning and Management of Medical and Biological Health Risks of the FMBA of Russia, 119121 Moscow, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Sergey Ivanov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Peter Shegay
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
43
|
Ciepła J, Smolarczyk R. Tumor hypoxia unveiled: insights into microenvironment, detection tools and emerging therapies. Clin Exp Med 2024; 24:235. [PMID: 39361163 PMCID: PMC11449960 DOI: 10.1007/s10238-024-01501-1] [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: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Hypoxia is one of the defining characteristics of the tumor microenvironment (TME) in solid cancers. It has a major impact on the growth and spread of malignant cells as well as their resistance to common treatments like radiation and chemotherapy. Here, we explore the complex functions of hypoxia in the TME and investigate its effects on angiogenesis, immunological evasion, and cancer cell metabolism. For prognostic and therapeutic reasons, hypoxia identification is critical, and recent developments in imaging and molecular methods have enhanced our capacity to precisely locate underoxygenated areas inside tumors. Furthermore, targeted therapies that take advantage of hypoxia provide a potential new direction in the treatment of cancer. Therapeutic approaches that specifically target hypoxic conditions in tumors without causing adverse effects are being led by hypoxia-targeted nanocarriers and hypoxia-activated prodrugs (HAPs). This review provides an extensive overview of this dynamic and clinically significant area of oncology research by synthesizing current knowledge about the mechanisms of hypoxia in cancer, highlighting state-of-the-art detection methodologies, and assessing the potential and efficacy of hypoxia-targeted therapies.
Collapse
Affiliation(s)
- Joanna Ciepła
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
44
|
Li L, Wang X, Jiang M, Li L, Wang D, Li Y. Advancements in a novel model of autophagy and immune network regulation in radioresistance of cancer stem cells. Biomed Pharmacother 2024; 179:117420. [PMID: 39255736 DOI: 10.1016/j.biopha.2024.117420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Radiotherapy, a precise modality for treating malignant tumors, has undergone rapid advancements in primary and clinical research. The mechanisms underlying tumor radioresistance have become significant research. With the introduction and in-depth study of cancer stem cells (CSCs) theory, CSCs have been identified as the primary factor contributing to the development of tumor radioresistance. The "stemness" of CSCs is a biological characteristic of a small subset of cells within tumor tissues, characterized by self-renewal solid ability. This characteristic leads to resistance to radiotherapy, chemotherapy, and targeted therapies, driving tumor recurrence and metastasis. Another study revealed that cellular autophagy plays a pivotal role in maintaining the "stemness" of CSCs. Autophagy is a cellular mechanism that degrades proteins and organelles to generate nutrients and energy in response to stress. This process maintains cellular homeostasis and contributes to CSCs radioresistance. Furthermore, ionizing radiation (IR) facilitates epithelial-to-mesenchymal transition (EMT), vascular regeneration, and other tumor processes by influencing the infiltration of M2-type tumor-associated macrophages (TAMs). IR promotes the activation of the classical immunosuppressive "switch," PD-1/PD-L1, which diminishes T-cell secretion, leading to immune evasion and promoting radioresistance. Interestingly, recent studies have found that the immune pathway PD-1/PD-L1 is closely related to cellular autophagy. However, the interrelationships between immunity, autophagy, and radioresistance of CSCs and the regulatory mechanisms involved remain unclear. Consequently, this paper reviews recent research to summarize these potential connections, aiming to establish a theoretical foundation for future studies and propose a new model for the network regulation of immunity, autophagy, and radioresistance of tumor cells.
Collapse
Affiliation(s)
- Leyao Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Xin Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Mei Jiang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Lei Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Di Wang
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yajun Li
- Department of Oncology, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China; Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China.
| |
Collapse
|
45
|
Behrooz AB, Shojaei S. Mechanistic insights into mesenchymal-amoeboid transition as an intelligent cellular adaptation in cancer metastasis and resistance. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167332. [PMID: 38960056 DOI: 10.1016/j.bbadis.2024.167332] [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: 01/22/2024] [Revised: 05/26/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Malignant cell plasticity is an important hallmark of tumor biology and crucial for metastasis and resistance. Cell plasticity lets cancer cells adapt to and escape the therapeutic strategies, which is the leading cause of cancer patient mortality. Epithelial cells acquire mobility via epithelial-mesenchymal transition (EMT), whereas mesenchymal cells enhance their migratory ability and clonogenic potential by acquiring amoeboid characteristics through mesenchymal-amoeboid transition (MAT). Tumor formation, progression, and metastasis depend on the tumor microenvironment (TME), a complex ecosystem within and around a tumor. Through increased migration and metastasis of cancer cells, the TME also contributes to malignancy. This review underscores the distinction between invasion pattern morphological manifestations and the diverse structures found within the TME. Furthermore, the mechanisms by which amoeboid-associated characteristics promote resistance and metastasis and how these mechanisms may represent therapeutic opportunities are discussed.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
46
|
Alcala S, Serralta San Martin G, Muñoz-Fernández de Legaria M, Moreno-Rubio J, Salinas S, López-Gil JC, Rojo López JA, Martínez Alegre J, Cortes Bandy DA, Zambrana F, Jiménez-Gordo AM, Casado E, López-Gómez M, Sainz B. Autofluorescent Cancer Stem Cells: Potential Biomarker to Predict Recurrence in Resected Colorectal Tumors. CANCER RESEARCH COMMUNICATIONS 2024; 4:2575-2588. [PMID: 39225547 PMCID: PMC11445700 DOI: 10.1158/2767-9764.crc-24-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/27/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cancer stem cells (CSC) in colorectal cancer drive intratumoral heterogeneity and distant metastases. Previous research from our group showed that CSCs can be easily detected by autofluorescence (AF). The aim of the present study was to evaluate the potential role of AF CSCs as a prognostic biomarker for colorectal cancer relapse. Seventy-five freshly resected tumors were analyzed by flow cytometry. AF was categorized as high (H-AF) or low, and the results were correlated with histologic features [grade of differentiation, presence of metastases in lymph nodes (LN), perivascular and lymphovascular invasion] and clinical variables (time to relapse and overall survival). Nineteen of the 75 (25.3%) patients experienced relapse (local or distant); of these 19 patients, 13 showed positive LNs and 6 had H-AF. Of note, four of them died before 5 years. Although patients with H-AF CSC percentages in the global population experienced 1.5 times increased relapse [HR, 1.47; 95% confidence interval (0.60-3.63)], patients with H-AF CSC percentages and LN metastases had the highest risk of relapse [HR, 7.92; P < 0.004; 95% confidence interval (1.97-31.82)]. These data support AF as an accurate and feasible marker to identify CSCs in resected colorectal cancer. A strong statistical association between H-AF CSCs and the risk of relapse was observed, particularly in patients with positive LNs, suggesting that H-AF patients might benefit from adjuvant chemotherapy regimens and intensive surveillance due to their high propensity to experience disease recurrence. Significance: AF has been proven to be an accurate biomarker for CSC identification; however, to date, their role as a prognostic factor after resection of colorectal cancer tumors has not been investigated. Our results show that determining the presence of AF CSCs after tumor resection has prognostic value and represents a potentially important tool for the management of patients with colorectal cancer.
Collapse
Affiliation(s)
- Sonia Alcala
- Department of Biochemistry, School of Medicine, Autónoma University of Madrid and Department of Cancer, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale (CSIC-UAM), Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Gonzalo Serralta San Martin
- Department of Internal Medicine, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
- Universidad Europea de Madrid, Madrid, Spain.
| | | | - Juan Moreno-Rubio
- Department of Medical Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
- Precision Nutrition and Cancer Program, Clinical Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain.
| | - Silvia Salinas
- Department of Pathology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
| | - Juan Carlos López-Gil
- Department of Biochemistry, School of Medicine, Autónoma University of Madrid and Department of Cancer, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale (CSIC-UAM), Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - José Alberto Rojo López
- Department of General Surgery, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
| | - Javier Martínez Alegre
- Universidad Europea de Madrid, Madrid, Spain.
- Department of General Surgery, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
| | | | - Francisco Zambrana
- Universidad Europea de Madrid, Madrid, Spain.
- Department of Medical Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
- Precision Nutrition and Cancer Program, Clinical Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain.
| | - Ana-María Jiménez-Gordo
- Universidad Europea de Madrid, Madrid, Spain.
- Department of Medical Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
- Precision Nutrition and Cancer Program, Clinical Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain.
| | - Enrique Casado
- Universidad Europea de Madrid, Madrid, Spain.
- Department of Medical Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
- Precision Nutrition and Cancer Program, Clinical Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain.
| | - Miriam López-Gómez
- Universidad Europea de Madrid, Madrid, Spain.
- Department of Medical Oncology, Infanta Sofía University Hospital, FIIB HUIS HHEN, Madrid, Spain.
| | - Bruno Sainz
- Department of Biochemistry, School of Medicine, Autónoma University of Madrid and Department of Cancer, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale (CSIC-UAM), Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
- Centro de Investigación Biomédica en Red, Área Cáncer, CIBERONC, ISCIII, Madrid, Spain.
| |
Collapse
|
47
|
Lei ZN, Teng QX, Koya J, Liu Y, Chen Z, Zeng L, Chen ZS, Fang S, Wang J, Liu Y, Pan Y. The correlation between cancer stem cells and epithelial-mesenchymal transition: molecular mechanisms and significance in cancer theragnosis. Front Immunol 2024; 15:1417201. [PMID: 39403386 PMCID: PMC11471544 DOI: 10.3389/fimmu.2024.1417201] [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: 04/14/2024] [Accepted: 09/06/2024] [Indexed: 01/03/2025] Open
Abstract
The connections between cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT) is critical in cancer initiation, progression, metastasis, and therapy resistance, making it a focal point in cancer theragnosis. This review provides a panorama of associations and regulation pathways between CSCs and EMT, highlighting their significance in cancer. The molecular mechanisms underlined EMT are thoroughly explored, including the involvement of key transcription factors and signaling pathways. In addition, the roles of CSCs and EMT in tumor biology and therapy resistance, is further examined in this review. The clinical implications of CSCs-EMT interplay are explored, including identifying mesenchymal-state CSC subpopulations using advanced research methods and developing targeted therapies such as inhibitors and combination treatments. Overall, understanding the reciprocal relationship between EMT and CSCs holds excellent potential for informing the development of personalized therapies and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Yangruiyu Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zizhou Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Leli Zeng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Shuo Fang
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jinxiang Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuchen Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yihang Pan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
- Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
48
|
Jin X, Lou X, Qi H, Zheng C, Li B, Siwu X, Liu R, Lv Q, Zhao A, Ruan J, Jiang M. NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma. Oncogenesis 2024; 13:35. [PMID: 39333079 PMCID: PMC11437035 DOI: 10.1038/s41389-024-00536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been observed in various cancers. Yet its exact contribution to the development of head and neck squamous cell carcinoma (HNSCC) remains undetermined. We previously found that NRF2 signaling is critical for the differentiation of squamous basal progenitor cells, while disruption of NRF2 causes basal cell hyperplasia. In this study, we revealed a correlation between elevated NRF2 activity and poor outcomes in HNSCC patients. We demonstrated that NRF2 facilitates tumor proliferation, migration, and invasion, as evidenced by both in vitro and in vivo studies. Significantly, NRF2 augments the expression of the antioxidant enzyme GPX2, thereby enhancing the proliferative, migratory, and invasive properties of HNSCC cells. Activation of GPX2 is critical for sustaining cancer stem cells (CSCs) by up-regulating NOTCH3, a key driver of cancer progression. These results elucidate that NRF2 regulates HNSCC progression through the NRF2-GPX2-NOTCH3 axis. Our findings proposed that pharmacological targeting of the NRF2-GPX2-NOTCH3 axis could be a potential therapeutic approach against HNSCC.
Collapse
Affiliation(s)
- Xiaoye Jin
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xiayuan Lou
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Haoxiang Qi
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Chao Zheng
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Bo Li
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xuerong Siwu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Ren Liu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Qiaoli Lv
- Institute of Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - An Zhao
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, China
| | - Ming Jiang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China.
| |
Collapse
|
49
|
Tufail M, Jiang CH, Li N. Altered metabolism in cancer: insights into energy pathways and therapeutic targets. Mol Cancer 2024; 23:203. [PMID: 39294640 PMCID: PMC11409553 DOI: 10.1186/s12943-024-02119-3] [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: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer cells undergo significant metabolic reprogramming to support their rapid growth and survival. This study examines important metabolic pathways like glycolysis, oxidative phosphorylation, glutaminolysis, and lipid metabolism, focusing on how they are regulated and their contributions to the development of tumors. The interplay between oncogenes, tumor suppressors, epigenetic modifications, and the tumor microenvironment in modulating these pathways is examined. Furthermore, we discuss the therapeutic potential of targeting cancer metabolism, presenting inhibitors of glycolysis, glutaminolysis, the TCA cycle, fatty acid oxidation, LDH, and glucose transport, alongside emerging strategies targeting oxidative phosphorylation and lipid synthesis. Despite the promise, challenges such as metabolic plasticity and the need for combination therapies and robust biomarkers persist, underscoring the necessity for continued research in this dynamic field.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
50
|
Qiu D, Wang T, Xiong Y, Li K, Qiu X, Feng Y, Lian Q, Qin Y, Liu K, Zhang Q, Jia C. TFCP2L1 drives stemness and enhances their resistance to Sorafenib treatment by modulating the NANOG/STAT3 pathway in hepatocellular carcinoma. Oncogenesis 2024; 13:33. [PMID: 39266516 PMCID: PMC11392926 DOI: 10.1038/s41389-024-00534-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy associated with high risks of recurrence and metastasis. Liver cancer stem cells (CSCs) are increasingly recognized as pivotal drivers of these processes. In our previous research, we demonstrated the involvement of TFCP2L1 in maintaining the pluripotency of embryonic stem cells. However, its relevance to liver CSCs remains unexplored. In this study, we report an inverse correlation between TFCP2L1 protein levels in HCC tissue and patient outcomes. The knockdown of TFCP2L1 significantly reduced HCC cell proliferation, invasion, metastasis, clonal formation, and sphere-forming capacity, while its overexpression enhanced these functions. In addition, experiments using a nude mouse model confirmed TFCP2L1's essential role in liver CSCs' function and tumorigenic potential. Mechanistically, we showed that TFCP2L1 promotes the stemness of CSCs by upregulating NANOG, which subsequently activates the JAK/STAT3 pathway, thereby contributing to HCC pathogenesis. Importantly, we identified a specific small molecule targeting TFCP2L1's active domain, which, in combination with Sorafenib, sensitizes hepatoma cells to treatment. Together, these findings underscore TFCP2L1's pathological significance in HCC progression, supporting its potential as a prognostic biomarker and therapeutic target in this disease.
Collapse
Affiliation(s)
- Dongbo Qiu
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiantian Wang
- Department of Medical Oncology; the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi Xiong
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kun Li
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiusheng Qiu
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuan Feng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qinghai Lian
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunfei Qin
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kunpeng Liu
- Medical college of Guangxi University, Nanning, Guangxi, China.
| | - Qi Zhang
- Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Changchang Jia
- Cell-gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|