1
|
Østergaard A, Boer JM, van Leeuwen FN, Pieters R, Den Boer ML. IKZF1 in acute lymphoblastic leukemia: the rise before the fall? Leuk Lymphoma 2024; 65:2077-2087. [PMID: 39210599 DOI: 10.1080/10428194.2024.2396046] [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: 04/09/2024] [Revised: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children and adolescents and in recent decades, the survival rates have risen to >90% in children largely due the introduction of risk adapted therapy. Therefore, knowledge of factors influencing risk of relapse is important. The transcription factor IKAROS is a regulator of lymphocyte development and alterations of its coding gene, IKZF1, are frequent in ALL and are associated with higher relapse risk. This concise review will discuss the normal function of IKAROS together with the effect of gene alterations in ALL such as relieved energy restriction and altered response to anti-leukemic drugs. Besides the biology, the clinical impact of gene alterations in the different subtypes of ALL will be discussed. Finally, possibilities for treating ALL with IKZF1 alterations will be considered including novel therapies like cell signaling inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Anna Østergaard
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Judith M Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
2
|
Yao J, Sun L, Gao F, Zhu W. Mesenchymal stem/stromal cells: dedicator to maintain tumor homeostasis. Hum Cell 2024; 38:21. [PMID: 39607530 DOI: 10.1007/s13577-024-01154-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) act as a factor in tumor recurrence after drug treatment with their involvement observed in various cancer types. As a constituent of the tumor microenvironment (TME), MSCs not only provide support to tumor growth but also establish connections with diverse cell populations within the TME, serving as mediators linking different tumor-associated components. MSCs play an important role in maintaining tumor progression due to their stem cell properties and remarkable differentiation capacity. Given the intensification of tumor research and the encouraging results achieved in recent years,the aim of this article is to investigate the supportive role of MSCs in tumor cells as well as in various cellular and non-cellular components of the tumor microenvironment. Furthermore, the article shows that MSCs do not have a specific anatomical ecological niche and describes the contribution of MSCs to the maintenance of tumor homeostasis on the basis of homing, plasticity and tumor-forming properties. By elucidating the critical roles of different components of TME, this study provides a comprehensive understanding of tumor therapy and may offer new insights into defeating cancer.
Collapse
Affiliation(s)
- Juncun Yao
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Li Sun
- Department of Clinical Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu Province, People's Republic of China
| | - Feng Gao
- Department of Surgery, Jingjiang People's Hospital, Jingjiang, 214500, People's Republic of China.
| | - Wei Zhu
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Ding X, Zhou Y, He L, Kang H, Wen Y, Xu J, Zhu C, Luo L, Zeng Q. Mitotic Arrest Deficient 2 Like 1 Contributes to Colorectal Cancer Cell Migration, Invasion, and Oxaliplatin Resistance Through the Wnt/β-Catenin Pathway. Chem Biol Drug Des 2024; 104:e70012. [PMID: 39487106 DOI: 10.1111/cbdd.70012] [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: 06/10/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
Colorectal cancer (CRC) is a highly prevalent malignancy, requiring chemotherapy for advanced stages of the disease. Previously, we found that mitotic arrest deficient 2 like 1 (MAD2L1) was upregulated and facilitated malignant proliferation in CRC. However, the association between MAD2L1 expression and tumor progression, as well as chemotherapy resistance in CRC, remains unclear. The progression capacities of CRC cells were assessed using transwell and wound healing assays, and the resistance to cisplatin in oxaliplatin-resistant CRC cells was assessed using CCK-8 assay and flow cytometry. Relevant protein levels of epithelial-to-mesenchymal transition (EMT) and Wnt/β-catenin pathway were analyzed using western blotting. Revealing the impact of MAD2L1 on metastasis and drug resistance in CRC through inhibition of the Wnt/β-catenin pathway. Knockdown of MAD2L1 attenuated the malignant progression of CRC cells, inhibited EMT, and blocked the Wnt/β-catenin pathway. MAD2L1 was significantly upregulated in oxaliplatin-resistant CRC cells, accompanied by the activation of the Wnt/β-catenin pathway. Knockdown of MAD2L1 effectively reversed oxaliplatin resistance, leading to apoptosis and downregulation of the protein expression levels of β-catenin, P-glycoprotein (P-gp), and ABCG2. After the knockdown of MAD2L1, the inhibition of the Wnt/β-catenin pathway exhibited a synergistic effect, effectively suppressing malignant progression and reversing oxaliplatin resistance in CRC cells. So, knockdown of MAD2L1 suppressed cell malignant progression, equally sensitized resistant CRC cells to oxaliplatin, potentially by blocking the activation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Xiang Ding
- Department of Gastroenterology, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Yonggui Zhou
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Linfang He
- Department of Gastroenterology, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Hubin Kang
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Youwu Wen
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Jia Xu
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Congbo Zhu
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Libing Luo
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Qingjun Zeng
- Department of Gastrointestinal Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| |
Collapse
|
4
|
Zhang X, Sun K, Gan R, Yan Y, Zhang C, Zheng D, Lu Y. WNT3 promotes chemoresistance to 5-Fluorouracil in oral squamous cell carcinoma via activating the canonical β-catenin pathway. BMC Cancer 2024; 24:564. [PMID: 38711026 PMCID: PMC11071218 DOI: 10.1186/s12885-024-12318-2] [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: 01/25/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND 5-Fluorouracil (5FU) is a primary chemotherapeutic agent used to treat oral squamous cell carcinoma (OSCC). However, the development of drug resistance has significantly limited its clinical application. Therefore, there is an urgent need to determine the mechanisms underlying drug resistance and identify effective targets. In recent years, the Wingless and Int-1 (WNT) signaling pathway has been increasingly studied in cancer drug resistance; however, the role of WNT3, a ligand of the canonical WNT signaling pathway, in OSCC 5FU-resistance is not clear. This study delved into this potential connection. METHODS 5FU-resistant cell lines were established by gradually elevating the drug concentration in the culture medium. Differential gene expressions between parental and resistant cells underwent RNA sequencing analysis, which was then substantiated via Real-time quantitative PCR (RT-qPCR) and western blot tests. The influence of the WNT signaling on OSCC chemoresistance was ascertained through WNT3 knockdown or overexpression. The WNT inhibitor methyl 3-benzoate (MSAB) was probed for its capacity to boost 5FU efficacy. RESULTS In this study, the WNT/β-catenin signaling pathway was notably activated in 5FU-resistant OSCC cell lines, which was confirmed through transcriptome sequencing analysis, RT-qPCR, and western blot verification. Additionally, the key ligand responsible for pathway activation, WNT3, was identified. By knocking down WNT3 in resistant cells or overexpressing WNT3 in parental cells, we found that WNT3 promoted 5FU-resistance in OSCC. In addition, the WNT inhibitor MSAB reversed 5FU-resistance in OSCC cells. CONCLUSIONS These data underscored the activation of the WNT/β-catenin signaling pathway in resistant cells and identified the promoting effect of WNT3 upregulation on 5FU-resistance in oral squamous carcinoma. This may provide a new therapeutic strategy for reversing 5FU-resistance in OSCC cells.
Collapse
Affiliation(s)
- Xuyang Zhang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China
| | - Kairui Sun
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China
| | - Ruihuan Gan
- Department of Preventive Dentistry, Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China
| | - Yuxiang Yan
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China
| | - Chaochao Zhang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China
| | - Dali Zheng
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China.
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China.
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China.
| | - Youguang Lu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China.
- Fujian Key Laboratory of Oral Diseases, Fuzhou, 350004, China.
- Fujian Provincial Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, 350004, China.
- Department of Preventive Dentistry, Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, China.
| |
Collapse
|
5
|
Panting RG, Kotecha RS, Cheung LC. The critical role of the bone marrow stromal microenvironment for the development of drug screening platforms in leukemia. Exp Hematol 2024; 133:104212. [PMID: 38552942 DOI: 10.1016/j.exphem.2024.104212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024]
Abstract
Extensive research over the past 50 years has resulted in significant improvements in survival for patients diagnosed with leukemia. Despite this, a subgroup of patients harboring high-risk genetic alterations still suffer from poor outcomes. There is a desperate need for new treatments to improve survival, yet consistent failure exists in the translation of in vitro drug development to clinical application. Preclinical screening conventionally utilizes tumor cell monocultures to assess drug activity; however, emerging research has acknowledged the vital role of the tumor microenvironment in treatment resistance and disease relapse. Current co-culture drug screening methods frequently employ fibroblasts as the designated stromal cell component. Alternative stromal cell types that are known to contribute to chemoresistance are often absent in preclinical evaluations of drug efficacy. This review highlights mechanisms of chemoresistance by a range of different stromal constituents present in the bone marrow microenvironment. Utilizing an array of stromal cell types at the early stages of drug screening may enhance the translation of in vitro drug development to clinical use. Ultimately, we highlight the need to consider the bone marrow microenvironment in drug screening platforms for leukemia to develop superior therapies for the treatment of high-risk patients with poor prognostic outcomes.
Collapse
Affiliation(s)
- Rhiannon G Panting
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Rishi S Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; School of Medicine, University of Western Australia, Perth, Western Australia, Australia; Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Laurence C Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.
| |
Collapse
|
6
|
Lu J, Chen XZ, Liu Y, Liu YJ, Liu B. Trends in confinement-induced cell migration and multi-omics analysis. Anal Bioanal Chem 2024; 416:2107-2115. [PMID: 38135761 DOI: 10.1007/s00216-023-05109-4] [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: 10/14/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023]
Abstract
Cell migration is an essential manner of different cell lines that are involved in embryological development, immune responses, tumorigenesis, and metastasis in vivo. Physical confinement derived from crowded tissue microenvironments has pivotal effects on migratory behaviors. Distinct migration modes under a heterogeneous extracellular matrix (ECM) have been extensively studied, uncovering potential molecular mechanisms involving a series of biological processes. Significantly, multi-omics strategies have been launched to provide multi-angle views of complex biological phenomena, facilitating comprehensive insights into molecular regulatory networks during cell migration. In this review, we describe biomimetic devices developed to explore the migratory behaviors of cells induced by different types of confined microenvironments in vitro. We also discuss the results of multi-omics analysis of intrinsic molecular alterations and critical pathway dysregulations of cell migration under heterogeneous microenvironments, highlighting the significance of physical confinement-triggered intracellular signal transduction in order to regulate cellular behaviors. Finally, we discuss both the challenges and promise of mechanistic analysis in confinement-induced cell migration, promoting the development of early diagnosis and precision therapeutics.
Collapse
Affiliation(s)
- Jiayin Lu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Xue-Zhu Chen
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Yixin Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China
| | - Yan-Jun Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China.
| | - Baohong Liu
- Department of ChemistryState Key Lab of Molecular Engineering of PolymersShanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Shanghai Stomatological HospitalShanghai Xuhui Central Hospital, Zhongshan-Xuhui HospitalFudan University, Shanghai, China.
| |
Collapse
|
7
|
Chai C, Tang H, Yi J, Li L, Yu C, Su Y, Miao L, Ye Z, Wang Z, Luo W, Hu J, Zhang H, Miao X, Xu H, Zhou W. Establishment and characterization of DPC-X4: a novel mixed-type ampullary cancer cell line. Hum Cell 2024; 37:531-545. [PMID: 38253956 DOI: 10.1007/s13577-023-01023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024]
Abstract
Mixed-type ampullary cancer is a distinct subtype of ampullary cancer that manifests a merging of the biological characteristics of both intestinal and pancreaticobiliary subtypes. The absence of established cell lines specific to this subtype has resulted in a concomitant scarcity of research on its tumorigenic mechanisms and the development of novel therapeutic modalities. The present study achieved the successful establishment of a novel mixed-type ampullary cancer cell line, designated DPC-X4 through primary culture techniques. Subsequent analyses pertaining to phenotypic characteristics, molecular profiling, biomarker identification, and histological features validated the DPC-X4 cell line as a potent model for delineating the pathogenesis of mixed-type ampullary cancer and facilitating the development of new pharmacological agents. This newly established cell line was subjected to continuous cultivation for 1 year, with stable passaging for over 50 generations. Notably, the DPC-X4 cell line manifested typical morphological features associated with epithelial tumors. Furthermore, the population doubling time for the DPC-X4 cell line was determined at 70 h. Short tandem repeat (STR) analysis confirmed that the DPC-X4 cell line exhibited a high genetic concordance with the primary tumor from the patient. Karyotypic profiling indicated an abnormal sub-triploid karyotype, with representative karyotypes of 57, XXY inv (9), 14p + , 15p + , der (17), + mar. The DPC-X4 cell line demonstrated a high capacity for efficient organoid formation under suspension culture conditions. In addition, the subcutaneous inoculation of DPC-X4 cells into NXG mice led to the formation of xenografted tumors. The results of drug sensitivity testing indicated that DPC-X4 cells were sensitive to paclitaxel and resistant to oxaliplatin, 5-fluorouracil, and gemcitabine. Immunohistochemistry revealed positive expression of CK7, CK19, and CK20 in DPC-X4 cells, while CDX2 demonstrated negative expression. In addition, positive expression of E-cadherin and vimentin was identified in DPC-X4 cells, with a proliferation index indicated by Ki-67 at 70%. The findings of our study establish DPC-X4 as a novel mixed-type ampullary cancer cell line, which can serve as a potential experimental model for exploring the pathogenesis of ampullary cancer and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Changpeng Chai
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Huan Tang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Jianfeng Yi
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Surgery, The First School of Clinical Medicine of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Lu Li
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Cheng Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of Anesthesiology, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yuanhui Su
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Long Miao
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Zhenzhen Ye
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- The First School of Clinical Medicine of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhengfeng Wang
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Wei Luo
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Jinjing Hu
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Hui Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
- Department of General Surgery, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Chengguan District, Lanzhou, 730000, China
| | - Xin Miao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| | - Hao Xu
- The Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
| | - Wence Zhou
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China.
- Department of General Surgery, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
8
|
Dong Y, Fan B, Li M, Zhang J, Xie S, Di S, Jia Q, Gong T. TESC acts as a prognostic factor and promotes epithelial-mesenchymal transition progression in esophageal squamous carcinoma. Pathol Res Pract 2024; 253:154964. [PMID: 38000203 DOI: 10.1016/j.prp.2023.154964] [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: 10/30/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND Tescalcin (TESC) is a critical regulator of cell differentiation and growth, promoting malignant progression in various tumors. However, the role of TESC in esophageal squamous carcinoma (ESCC) remains unclear. METHODS Immunohistochemistry (IHC), quantitative real-time PCR (qRT-PCR), and western blot were utilized to identify the difference in TESC expression between ESCC tissues and normal tissues adjacent to the carcinoma. The relationship between TESC and several clinicopathological features was shown by the chi-square test. Log-rank analysis and Cox regression were used to detect the relationship between TESC and the prognosis in ESCC. Clone formation and cell count kit-8 (CCK-8) were applied to detect the impact of TESC on ESCC proliferation. Wound healing assay and transwell assay were used to confirm the influence of TESC on the invasion and migration. Spearman correlation coefficient was used to describe the correlation between TESC and epithelial-mesenchymal transition (EMT)-related protein expression in ESCC. Western blot was used to detect the effect of TESC on the expression of E-cadherin, N-cadherin, and Vimentin as well as AKT signaling pathway. Xenograft tumors were developed to test the pro-tumorigenic impacts of TESC in vivo. RESULTS TESC was upregulated expression in ESCC tissues and was linked to poorer prognosis and worse tumor infiltration, TNM stage, and lymph node metastasis. Meanwhile, TESC was able to act as an independent prognostic factor in ESCC. TESC promoted tumor cell proliferation, invasion, migration, EMT progression, and activated the phosphorylation of the AKT pathway. Furthermore, TESC knockdown inhibited the growth of carcinoma in vivo. CONCLUSION TESC is a predictive factor for poor prognosis in ESCC and may provide a new strategy for ESCC treatment.
Collapse
Affiliation(s)
- Yanxin Dong
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Boshi Fan
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jiale Zhang
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shun Xie
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China
| | - Shouyin Di
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| | - Taiqian Gong
- Department of Thoracic Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China; School of Medicine, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
9
|
Li Z, Yin P. Tumor microenvironment diversity and plasticity in cancer multidrug resistance. Biochim Biophys Acta Rev Cancer 2023; 1878:188997. [PMID: 37832894 DOI: 10.1016/j.bbcan.2023.188997] [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: 08/23/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023]
Abstract
Multidrug resistance (MDR) poses a significant obstacle to effective cancer treatment, and the tumor microenvironment (TME) is crucial for MDR development and reversal. The TME plays an active role in promoting MDR through several pathways. However, a promising therapeutic approach for battling MDR involves targeting specific elements within the TME. Therefore, this comprehensive review elaborates on the research developments regarding the dual role of the TME in promoting and reversing MDR in cancer. Understanding the complex role of the TME in promoting and reversing MDR is essential to developing effective cancer therapies. Utilizing the adaptability of the TME by targeting novel TME-specific factors, utilizing combination therapies, and employing innovative treatment strategies can potentially combat MDR and achieve personalized treatment outcomes for patients with cancer.
Collapse
Affiliation(s)
- Zhi Li
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Department of General surgery, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| |
Collapse
|
10
|
Atiya HI, Gorecki G, Garcia GL, Frisbie LG, Baruwal R, Coffman L. Stromal-Modulated Epithelial-to-Mesenchymal Transition in Cancer Cells. Biomolecules 2023; 13:1604. [PMID: 38002286 PMCID: PMC10669774 DOI: 10.3390/biom13111604] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The ability of cancer cells to detach from the primary site and metastasize is the main cause of cancer- related death among all cancer types. Epithelial-to-mesenchymal transition (EMT) is the first event of the metastatic cascade, resulting in the loss of cell-cell adhesion and the acquisition of motile and stem-like phenotypes. A critical modulator of EMT in cancer cells is the stromal tumor microenvironment (TME), which can promote the acquisition of a mesenchymal phenotype through direct interaction with cancer cells or changes to the broader microenvironment. In this review, we will explore the role of stromal cells in modulating cancer cell EMT, with particular emphasis on the function of mesenchymal stromal/stem cells (MSCs) through the activation of EMT-inducing pathways, extra cellular matrix (ECM) remodeling, immune cell alteration, and metabolic rewiring.
Collapse
Affiliation(s)
- Huda I. Atiya
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Grace Gorecki
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Geyon L. Garcia
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leonard G. Frisbie
- Department of Integrative Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roja Baruwal
- Molecular Pharmacology Graduate Program, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Lan Coffman
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee Women’s Research Institute, Pittsburgh, PA15213, USA
| |
Collapse
|
11
|
Sarrand J, Soyfoo MS. Involvement of Epithelial-Mesenchymal Transition (EMT) in Autoimmune Diseases. Int J Mol Sci 2023; 24:14481. [PMID: 37833928 PMCID: PMC10572663 DOI: 10.3390/ijms241914481] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex reversible biological process characterized by the loss of epithelial features and the acquisition of mesenchymal features. EMT was initially described in developmental processes and was further associated with pathological conditions including metastatic cascade arising in neoplastic progression and organ fibrosis. Fibrosis is delineated by an excessive number of myofibroblasts, resulting in exuberant production of extracellular matrix (ECM) proteins, thereby compromising organ function and ultimately leading to its failure. It is now well acknowledged that a significant number of myofibroblasts result from the conversion of epithelial cells via EMT. Over the past two decades, evidence has accrued linking fibrosis to many chronic autoimmune and inflammatory diseases, including systemic sclerosis (SSc), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), Sjögren's syndrome (SS), and inflammatory bowel diseases (IBD). In addition, chronic inflammatory states observed in most autoimmune and inflammatory diseases can act as a potent trigger of EMT, leading to the development of a pathological fibrotic state. In the present review, we aim to describe the current state of knowledge regarding the contribution of EMT to the pathophysiological processes of various rheumatic conditions.
Collapse
Affiliation(s)
- Julie Sarrand
- Department of Rheumatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Muhammad S. Soyfoo
- Department of Rheumatology, Hôpital Erasme, Université Libre de Bruxelles, 1070 Brussels, Belgium
| |
Collapse
|