1
|
Kariya Y, Nishita M. Integrins in Cancer Drug Resistance: Molecular Mechanisms and Clinical Implications. Int J Mol Sci 2025; 26:3143. [PMID: 40243917 PMCID: PMC11989024 DOI: 10.3390/ijms26073143] [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: 03/03/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
It is estimated that between 80 and 90% of mortality in cancer patients is directly or indirectly related to drug resistance. Consequently, overcoming drug resistance represents a significant challenge in the treatment of cancer. Integrins are transmembrane adhesion molecules that facilitate the linkage between the extracellular matrix (ECM) and the cytoskeleton, thereby enabling the activation of various cellular signaling pathways. Integrins are highly expressed in various cancers and contribute to cancer progression through invasion and metastasis. In addition, recent studies have revealed that integrins play a pivotal role in the development of drug resistance in cancer. This review will first provide an overview of integrin function and classification. It then discusses recent advances in understanding how integrins contribute to drug resistance in cancer, with a focus on ECM, drug transporters, the epithelial-to-mesenchymal transition (EMT), cancer stemness, PD-L1, and glycosylation. Finally, the potential applications of integrins as targets for therapeutic agents against drug-resistant cancers are also summarized.
Collapse
Affiliation(s)
- Yoshinobu Kariya
- Department of Biochemistry, Fukushima Medical University, 1 Hikarigaoka, Fukushima City 960-1295, Fukushima, Japan
| | | |
Collapse
|
2
|
Hushmandi K, Alimohammadi M, Heiat M, Hashemi M, Nabavi N, Tabari T, Raei M, Aref AR, Farahani N, Daneshi S, Taheriazam A. Targeting Wnt signaling in cancer drug resistance: Insights from pre-clinical and clinical research. Pathol Res Pract 2025; 267:155837. [PMID: 39954370 DOI: 10.1016/j.prp.2025.155837] [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: 08/08/2024] [Revised: 12/22/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Cancer drug resistance, encompassing both acquired and intrinsic chemoresistance, remains a significant challenge in the clinical management of tumors. While advancements in drug discovery and the development of various small molecules and anti-cancer compounds have improved patient responses to chemotherapy, the frequent and prolonged use of these drugs continues to pose a high risk of developing chemoresistance. Therefore, understanding the primary mechanisms underlying drug resistance is crucial. Wnt proteins, as secreted signaling molecules, play a pivotal role in transmitting signals from the cell surface to the nucleus. Aberrant expression of Wnt proteins has been observed in a variety of solid and hematological tumors, where they contribute to key processes such as proliferation, metastasis, stemness, and immune evasion, often acting in an oncogenic manner. Notably, the role of the Wnt signaling pathway in modulating chemotherapy response in human cancers has garnered significant attention. This review focuses on the involvement of Wnt signaling and its related molecular pathways in drug resistance, highlighting their associations with cancer hallmarks, stemness, and tumorigenesis linked to chemoresistance. Additionally, the overexpression of Wnt proteins has been shown to accelerate cancer drug resistance, with regulation mediated by non-coding RNAs. Elevated Wnt activity reduces cell death in cancers, particularly by affecting mechanisms like apoptosis, autophagy, and ferroptosis. Furthermore, pharmacological compounds and small molecules have demonstrated the potential to modulate Wnt signaling in cancer therapy. Given its impact, Wnt expression can also serve as a prognostic marker and a factor influencing survival outcomes in human cancers.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Teimour Tabari
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
3
|
Song X, Liu S, Zeng Y, Cai Y, Luo H. BANCR-containing extracellular vesicles enhance breast cancer resistance. J Biol Chem 2025; 301:108304. [PMID: 39947472 PMCID: PMC11999273 DOI: 10.1016/j.jbc.2025.108304] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 04/04/2025] Open
Abstract
Extracellular vesicles (EVs) are nano-sized particles secreted by many cell types-including tumor cells-and play key roles in cellular communication by transporting functional RNAs. This study aims to elucidate the role of long noncoding RNA BRAF-activated nonprotein coding RNA (BANCR) in EVs derived from breast cancer (BC) cells in trastuzumab resistance. Differentially expressed long noncoding RNA and downstream targets in BC-resistant samples were identified. SKBR-3 cells were treated with trastuzumab to generate resistant cells (SKBR-3TR), and EVs from these cells (SKBR-3TR-EVs) were isolated and characterized. Functional studies of BANCR were performed in SKBR-3 and SKBR-3TR cells. Coculturing SKBR-3 cells with SKBR-3TR-EVs assessed changes in cell behavior. A xenograft model in nude mice examined in vivo tumorigenicity and trastuzumab resistance. BANCR was highly expressed in SKBR-3TR cells and EVs, linked to trastuzumab resistance. SKBR-3TR-EVs transferred BANCR to SKBR-3 cells, where BANCR inhibited miR-34a-5p, reducing its expression. High-mobility group A1 (HMGA1) was identified as a miR-34a-5p target. BANCR activated the HMGA1/Wnt/β-catenin pathway by inhibiting miR-34a-5p, promoting resistance. In vivo experiments showed that BANCR inhibition delayed tumorigenesis and reversed trastuzumab resistance. BC cell-derived EVs containing BANCR may enhance resistance to trastuzumab by regulating the miR-34a-5p/HMGA1/Wnt/β-catenin axis, presenting a potential target for BC therapy.
Collapse
Affiliation(s)
- Xinming Song
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shen Liu
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ying Zeng
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yilin Cai
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haiqing Luo
- Department of Head and Neck Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
4
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [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: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
5
|
Zhang J, Chen Y, Gong X, Yang Y, Gu Y, Huang L, Fu J, Zhao M, Huang Y, Li L, Liu W, Wan Y, He X, Ma Z, Zhao W, Zhang M, Tang T, Wang Y, Thiery JP, Zheng X, Chen L. GATA factor TRPS1, a new DNA repair protein, cooperates with reversible PARylation to promote chemoresistance in patients with breast cancer. J Biol Chem 2024; 300:107780. [PMID: 39276941 PMCID: PMC11490888 DOI: 10.1016/j.jbc.2024.107780] [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: 11/19/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Resistance to DNA-damaging agents is a major unsolved challenge for breast cancer patients undergoing chemotherapy. Here, we show that elevated expression of transcriptional repressor GATA binding 1 (TRPS1) is associated with lower drug sensitivity, reduced response rate, and poor prognosis in chemotherapy-treated breast cancer patients. Mechanistically, elevated TRPS1 expression promotes hyperactivity of DNA damage repair (DDR) in breast cancer cells. We provide evidence that TRPS1 dynamically localizes to DNA breaks in a Ku70-and Ku80-dependent manner and that TRPS1 is a new member of the DDR protein family. We also discover that the dynamics of TRPS1 assembly at DNA breaks is regulated by its reversible PARylation in the DDR, and that mutations of the PARylation sites on TRPS1 lead to increased sensitivity to chemotherapeutic drugs. Taken together, our findings provide new mechanistic insights into the DDR and chemoresistance in breast cancer patients and identify TRPS1 as a critical DDR protein. TRPS1 may also be considered as a target to improve chemo-sensitization strategies and, consequently, clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yatao Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xue Gong
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yongfeng Yang
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Yun Gu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Ling Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jianfeng Fu
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China
| | - Menglu Zhao
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yehong Huang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lulu Li
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wenzhuo Liu
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yajie Wan
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xilin He
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhifang Ma
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Weiyong Zhao
- Department of Radiation Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meng Zhang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tao Tang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yuzhi Wang
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China
| | | | - Xiaofeng Zheng
- State Key Lab of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, China.
| | - Liming Chen
- Department of Biochemistry, School of Life Sciences, Nanjing Normal University, Nanjing, China; Jiangsu Institute of Cancer Research, Jiangsu Cancer Hospital, the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Wang B, Yang X, Li C, Yang R, Sun T, Yin Y. The shared molecular mechanism of spinal cord injury and sarcopenia: a comprehensive genomics analysis. Front Neurol 2024; 15:1373605. [PMID: 39281413 PMCID: PMC11392746 DOI: 10.3389/fneur.2024.1373605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024] Open
Abstract
Introduction The occurrence of Spinal cord injury (SCI) brings economic burden and social burden to individuals, families and society, and the complications after SCI greatly affect the rehabilitation and treatment of patients in the later stage.This study focused on the potential biomarkers that co-exist in SCI and sarcopenia, with the expectation to diagnose and prognose patients in the acute phase and rehabilitation phase using comprehensive data analysis. Methods The datasets used in this study were downloaded from Gene Expression Omnibus (GEO) database. Firstly, the datasets were analyzed with the "DEseq2" and "Limma" R package to identify differentially expressed genes (DEGs), which were then visualized using volcano plots. The SCI and sarcopenia DEGs that overlapped were used to construct a protein-protein interaction (PPI) network. Three algorithms were used to obtain a list of the top 10 hub genes. Next, validation of the hub genes was performed using three datasets. According to the results, the top hub genes were DCN, FSTL1, and COL12A1, which subsequently underwent were Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. We also assessed immune cell infiltration with the CIBERSORT algorithm to explore the immune cell landscape. The correlations between the hub genes and age and body mass index were investigated. To illustrate the biological mechanisms of the hub genes more clearly, a single-cell RNA-seq dataset was assessed to determine gene expression when muscle injury occurred. According to our analysis and the role in muscle, we chose the fibro/adipogenic progenitors (FAPs) cluster in the next step of the analysis. In the sub cluster analysis, we use the "Monocle" package to perform the trajectory analysis in different injury time points and different cell states. Results A total of 144 overlapped genes were obtained from two datasets. Following PPI network analysis and validation, we finally identified three hub-genes (DCN, FSTL1, and COL12A1), which were significantly altered in sarcopenic SCI patients both before and after rehabilitation training. The three hub genes were also significantly expressed in the FAPs clusters. Furthermore, following injury, the expression of the hub genes changed with the time points, changing in FAPs cluster. Discussion Our study provides comprehensive insights into how muscle changes after SCI are associated with sarcopenia by moving from RNA-seq to RNA-SEQ, including Immune infiltration landscape, pesudotime change and so on. The three hub genes identified in this study could be used to distinguish the sarcopenia state at the genomic level. Additionally, they may also play a prognostic role in evaluating the efficiency of rehabilitation training.
Collapse
Affiliation(s)
- Binyang Wang
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
- The Affiliated Hospital of Yunnan University, Kunming Medical University, Kunming, China
| | - Xu Yang
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Chuanxiong Li
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Rongxing Yang
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Tong Sun
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Yong Yin
- Department of Rehabilitation, The Affiliated Hospital of Yunnan University, Kunming, China
| |
Collapse
|
7
|
Kudo-Saito C, Matsumura S, Mori T, Honma Y, Yoshimoto S. Prognostic significance of the FSTL1-DIP2A axis in early-stage tongue cancer. Am J Cancer Res 2024; 14:3816-3825. [PMID: 39267678 PMCID: PMC11387867 DOI: 10.62347/rzao3562] [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: 06/18/2024] [Accepted: 07/21/2024] [Indexed: 09/15/2024] Open
Abstract
In tongue cancer, many patients already have metastasis at the time of diagnosis, and such cases are usually unresponsive to treatment, resulting in a poor prognosis. Therefore, there is an urgent need to develop more effective diagnostic and therapeutic methods to cure tongue cancer at the earliest possible stage in clinical practice. Follistatin-like 1 (FSTL1) is known as a negative effector molecule that induces and enhances the refractoriness of cancer cells directly and indirectly via suppressing anti-tumor immunity in various types of cancer. However, the molecular expression, functions, and clinical significance of FSTL1 and its receptor DIP2A in tongue cancer remains to be elucidated. In this study, we revealed that FSTL1, which is highly expressed in tongue cancer cells, plays a key role in its malignancy and is a significant risk factor for recurrence of early-stage tongue cancer. Basic study shows that FSTL1 is abundantly produced from human tongue cancer cell lines, and blocking FSTL1 with specific siRNAs or mAb significantly suppresses cellular functions. Clinical study shows that both FSTL1 and its receptor DIP2A are highly and correlatively expressed in tumor tissues of tongue cancer patients, and high expression levels of both in stage I tumors are significantly associated with shorter relapse-free survival. These suggest that targeting the FSTL1-DIP2A axis may be useful as a biomarker for early prediction of prognosis in tongue cancer patients, and as a therapeutic target for developing new drugs to treat tongue cancer more effectively. This strategy will contribute to improving clinical outcomes in tongue cancer.
Collapse
Affiliation(s)
- Chie Kudo-Saito
- Department of Immune Medicine, National Cancer Center Research Institute Tokyo, Japan
| | - Satoko Matsumura
- Department of Head and Neck Surgery, National Cancer Center Hospital Tokyo, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Center Hospital Tokyo, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital Tokyo, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Hospital Tokyo, Japan
| |
Collapse
|
8
|
Mehrtabar E, Khalaji A, Pandeh M, Farhoudian A, Shafiee N, Shafiee A, Ojaghlou F, Mahdavi P, Soleymani-Goloujeh M. Impact of microRNA variants on PI3K/AKT signaling in triple-negative breast cancer: comprehensive review. Med Oncol 2024; 41:222. [PMID: 39120634 DOI: 10.1007/s12032-024-02469-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
Breast cancer (BC) is a significant cause of cancer-related mortality, and triple-negative breast cancer (TNBC) is a particularly aggressive subtype associated with high mortality rates, especially among younger females. TNBC poses a considerable clinical challenge due to its aggressive tumor behavior and limited therapeutic options. Aberrations within the PI3K/AKT pathway are prevalent in TNBC and correlate with increased therapeutic intervention resistance and poor outcomes. MicroRNAs (miRs) have emerged as crucial PI3K/AKT pathway regulators influencing various cellular processes involved in TNBC pathogenesis. The levels of miRs, including miR-193, miR-4649-5p, and miR-449a, undergo notable changes in TNBC tumor tissues, emphasizing their significance in cancer biology. This review explored the intricate interplay between miR variants and PI3K/AKT signaling in TNBC. The review focused on the molecular mechanisms underlying miR-mediated dysregulation of this pathway and highlighted specific miRs and their targets. In addition, we explore the clinical implications of miR dysregulation in TNBC, particularly its correlation with TNBC prognosis and therapeutic resistance. Elucidating the roles of miRs in modulating the PI3K/AKT signaling pathway will enhance our understanding of TNBC biology and unveil potential therapeutic targets. This comprehensive review aims to discuss current knowledge and open promising avenues for future research, ultimately facilitating the development of precise and effective treatments for patients with TNBC.
Collapse
Affiliation(s)
- Ehsan Mehrtabar
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Khalaji
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Pandeh
- School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Aram Farhoudian
- School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Nadia Shafiee
- Children's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefe Shafiee
- Board-Certified Cardiologist, Rajaie Cardiovascular Medical and Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ojaghlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parinaz Mahdavi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Mehdi Soleymani-Goloujeh
- Diabetes Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
10
|
Wang H, Zhang L, Hu C, Li H, Jiang M. Wnt signaling and tumors (Review). Mol Clin Oncol 2024; 21:45. [PMID: 38798312 PMCID: PMC11117032 DOI: 10.3892/mco.2024.2743] [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: 02/02/2023] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Wnt signaling is a highly conserved evolutionary pathway that plays a key role in regulation of embryonic development, as well as tissue homeostasis and regeneration. Abnormalities in Wnt signaling are associated with tumorigenesis and development, leading to poor prognosis in patients with cancer. However, the pharmacological effects and mechanisms underlying Wnt signaling and its inhibition in cancer treatment remain unclear. In addition, potential side effects of inhibiting this process are not well understood. Therefore, the present review outlines the role of Wnt signaling in tumorigenesis, development, metastasis, cancer stem cells, radiotherapy resistance and tumor immunity. The present review further identifies inhibitors that target Wnt signaling to provide a potential novel direction for cancer treatment. This may facilitate early application of safe and effective drugs targeting Wnt signaling in clinical settings. An in-depth understanding of the mechanisms underlying inhibition of Wnt signaling may improve the prognosis of patients with cancer.
Collapse
Affiliation(s)
- Huaishi Wang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Lihai Zhang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Chao Hu
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Mingyan Jiang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| |
Collapse
|
11
|
Du R, Li K, Guo K, Chen Z, Han L, Bian H. FSTL1: A double-edged sword in cancer development. Gene 2024; 906:148263. [PMID: 38346455 DOI: 10.1016/j.gene.2024.148263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/25/2024] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Flolistatin-related protein 1 (FSTL1), a secreted glycoprotein that is involved in many physiological functions, has attracted much interest and has been implicated in a wide range of diseases, including heart diseases and inflammatory diseases. In recent years, the involvement of FSTL1 in cancer progression has been implicated and researched. FSTL1 plays a contradictory role in cancer, depending on the cancer type as well as the contents of the tumor microenvironment. As reviewed here, the structure and distribution of FSTL1 are first introduced. Subsequently, the expression and clinical significance of FSTL1 in various types of cancer as a tumor enhancer or inhibitor are addressed. Furthermore, we discuss the functional role of FSTL1 in various processes that involve tumor cell proliferation, metastasis, immune responses, stemness, cell apoptosis, and resistance to chemotherapy. FSTL1 expression is tightly controlled in cancer, and a multitude of cancer-related signaling cascades like TGF-β/BMP/Smad signaling, AKT, NF-κB, and Wnt-β-catenin signaling pathways are modulated by FSTL1. Finally, FSTL1 as a therapeutic target using monoclonal antibodies is stated. Herein, we review recent findings showing the double-edged characteristics and mechanisms of FSTL1 in cancer and elaborate on the current understanding of therapeutic approaches targeting FSTL1.
Collapse
Affiliation(s)
- Ruijuan Du
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China
| | - Kai Li
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China
| | - Kelei Guo
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China
| | - Zhiguo Chen
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China
| | - Li Han
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China.
| | - Hua Bian
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, PR China; Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang 473004, Henan Province, PR China.
| |
Collapse
|
12
|
Nasimi Shad A, Moghbeli M. Integrins as the pivotal regulators of cisplatin response in tumor cells. Cell Commun Signal 2024; 22:265. [PMID: 38741195 DOI: 10.1186/s12964-024-01648-0] [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: 01/24/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024] Open
Abstract
Cisplatin (CDDP) is a widely used first-line chemotherapeutic drug in various cancers. However, CDDP resistance is frequently observed in cancer patients. Therefore, it is required to evaluate the molecular mechanisms associated with CDDP resistance to improve prognosis among cancer patients. Integrins are critical factors involved in tumor metastasis that regulate cell-matrix and cell-cell interactions. They modulate several cellular mechanisms including proliferation, invasion, angiogenesis, polarity, and chemo resistance. Modification of integrin expression levels can be associated with both tumor progression and inhibition. Integrins are also involved in drug resistance of various solid tumors through modulation of the tumor cell interactions with interstitial matrix and extracellular matrix (ECM). Therefore, in the present review we discussed the role of integrin protein family in regulation of CDDP response in tumor cells. It has been reported that integrins mainly promoted the CDDP resistance through interaction with PI3K/AKT, MAPK, and WNT signaling pathways. They also regulated the CDDP mediated apoptosis in tumor cells. This review paves the way to suggest the integrins as the reliable therapeutic targets to improve CDDP response in tumor cells.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Choi BM, Cheong JH, Ryu JI, Won YD, Min KW, Han MH. Significant Genes Associated with Mortality and Disease Progression in Grade II and III Glioma. Biomedicines 2024; 12:858. [PMID: 38672212 PMCID: PMC11048596 DOI: 10.3390/biomedicines12040858] [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: 03/25/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The Wnt/β-catenin pathway plays a critical role in the tumorigenesis and maintenance of glioma stem cells. This study aimed to evaluate significant genes associated with the Wnt/β-catenin pathway involved in mortality and disease progression in patients with grade II and III glioma, using the Cancer Genome Atlas (TCGA) database. METHODS We obtained clinicopathological information and mRNA expression data from 515 patients with grade II and III gliomas from the TCGA database. We performed a multivariate Cox regression analysis to identify genes independently associated with glioma prognosis. RESULTS The analysis of 34 genes involved in Wnt/β-catenin signaling demonstrated that four genes (CER1, FRAT1, FSTL1, and RPSA) related to the Wnt/β-catenin pathway were significantly associated with mortality and disease progression in patients with grade II and III glioma. We also identified additional genes related to the four significant genes of the Wnt/β-catenin pathway mentioned above. The higher expression of BMP2, RPL18A, RPL19, and RPS12 is associated with better outcomes in patients with glioma. CONCLUSIONS Using a large-scale open database, we identified significant genes related to the Wnt/β-catenin signaling pathway associated with mortality and disease progression in patients with grade II and III gliomas.
Collapse
Affiliation(s)
- Bo Mi Choi
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Gyeonggi-do, Republic of Korea; (B.M.C.); (J.H.C.); (J.I.R.); (Y.D.W.)
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Gyeonggi-do, Republic of Korea; (B.M.C.); (J.H.C.); (J.I.R.); (Y.D.W.)
| | - Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Gyeonggi-do, Republic of Korea; (B.M.C.); (J.H.C.); (J.I.R.); (Y.D.W.)
| | - Yu Deok Won
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Gyeonggi-do, Republic of Korea; (B.M.C.); (J.H.C.); (J.I.R.); (Y.D.W.)
| | - Kyueng-Whan Min
- Department of Pathology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu 11759, Gyeonggi-do, Republic of Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri 11923, Gyeonggi-do, Republic of Korea; (B.M.C.); (J.H.C.); (J.I.R.); (Y.D.W.)
| |
Collapse
|
14
|
Hu Y, He Y, Luo N, Li X, Guo L, Zhang K. A feedback loop between lncRNA MALAT1 and DNMT1 promotes triple-negative breast cancer stemness and tumorigenesis. Cancer Biol Ther 2023; 24:2235768. [PMID: 37548553 PMCID: PMC10408694 DOI: 10.1080/15384047.2023.2235768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/23/2022] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND The function of long non-coding RNA (lncRNA) MALAT1 in regulating triple-negative breast cancer (TNBC) stemness and tumorigenesis was investigated. METHODS Sphere formation and colony formation assays coupled with flow cytometry were employed to evaluate the percentage of CD44high/CD44low cells, and ALDH+ cells were performed to evaluate the stemness. Bisulfite sequencing PCR (BSP) was employed to detect the methylation level of MALAT1. Tumor xenograft experiment was performed to evaluate tumorigenesis in vivo. Finally, dual-luciferase reporter and RIP assays were employed to verify the binding relationship between MALAT1 and miR-137. RESULTS Our results revealed that MALAT1 and BCL11A were highly expressed in TNBC, while miR-137 and DNMT1 were lowly expressed. Our results proved that MALAT1 positively regulated BCL11A expression through targeting miR-137. Functional experiments revealed that MALAT1 inhibited DNMT1 expression through acting on the miR-137/BCL11A pathway to enhance TNBC stemness and tumorigenesis. We also found that high MALAT1 expression in TNBC was related to the DNMT1-mediated hypomethylation of MALAT1. As expected, DNMT1 overexpression could remarkably inhibit TNBC stemness and tumorigenesis, which was eliminated by MALAT1 overexpression. CONCLUSION MALAT1 downregulated DNMT1 by miR-137/BCL11A pathway to enhance TNBC stemness and tumorigenesis; meanwhile, DNMT1/MALAT1 formed a positive feedback loop to continuously promote TNBC malignant behaviors.
Collapse
Affiliation(s)
- Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| | - Yuqiong He
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| | - Na Luo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| | - Xin Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| | - Lei Guo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| | - Kejing Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- Department of General Surgery, Xiangya Hospital, Central South University, Clinical Research Center for Breast Cancer in Hunan Province, Changsha, Hunan Province, P.R. China
| |
Collapse
|
15
|
Huang L, Zhu Y, Kong Q, Guan X, Lei X, Zhang L, Yang H, Yao X, Liang S, An X, Yu J. Inhibition of Integrin α vβ 3-FAK-MAPK signaling constrains the invasion of T-ALL cells. Cell Adh Migr 2023; 17:1-14. [PMID: 36944577 PMCID: PMC10038045 DOI: 10.1080/19336918.2023.2191913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
The role of adhesion receptor integrin αvβ3 in T-ALL was unclear. Firstly, we performed quantitative real-time PCR to assess medullary expression of integrin β3(ITGB3) in T-ALL patients and high ITGB3 expression was relevant with the central nervous system leukemia(CNSL) incidence. Decreasing of cell invasion was observed in Jurkat and Molt4 treated with integrin αvβ3 specific antibody and inhibitor as well as cells with ITGB3 interference. Further, phosphorylation of FAK, cRAF, MEK and ERK decreased in cells with integrin αvβ3 inhibition or interference. Invasion decreased in T-ALL cells treated with FAK and ERK inhibitors. In conclusion, inhibition of integrin αvβ3 signals significantly limits the cell invasion of T-ALL cells.
Collapse
Affiliation(s)
- Lan Huang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yao Zhu
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qinglin Kong
- Department of Hematology and Oncology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianmin Guan
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xiaoying Lei
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Luying Zhang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Hui Yang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinyuan Yao
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Shaoyan Liang
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xizhou An
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jie Yu
- Department of hematology and oncology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
16
|
Anilkumar KV, Rema LP, John MC, Vanesa John T, George A. miRNAs in the prognosis of triple-negative breast cancer: A review. Life Sci 2023; 333:122183. [PMID: 37858714 DOI: 10.1016/j.lfs.2023.122183] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023]
Abstract
Triple-Negative Breast Cancer (TNBC) is a highly aggressive and invasive type of breast cancer (BC) with high mortality rate wherein effective target medicaments are lacking. It is a very heterogeneous group with several subtypes that account for 10-20% of cancer among women globally, being negative for three most important receptors (estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2)), with an early and high recurrence resulting in poor survival rate. Therefore, a more thorough knowledge on carcinogenesis of TNBC is required for the development of personalized treatment options. miRNAs can either promote or suppress tumorigenesis and have been linked to a number of features of cancer progression, including proliferation, metastasis, apoptosis, and epithelial-mesenchymal transition (EMT). Recent miRNA research shows that there is great potential for the development of novel biomarkers as they have emerged as drivers of tumorigenesis and provide opportunities to target various components involved in TNBC, thus helping to solve this difficult-to-treat disease. In this review, we summarize the most relevant miRNAs that play an essential role in TNBC biology. Their role with regard to molecular mechanisms underlying TNBC progression has been discussed, and their potential use as therapeutic or prognostic markers to unravel the intricacy of TNBC based on the pieces of evidence obtained from various works of literature has been briefly addressed.
Collapse
Affiliation(s)
- Kavya V Anilkumar
- PG and Research Department of Zoology, Maharaja's College, Ernakulam, 682011, India; Cell and Molecular Biology Facility, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - L P Rema
- PG and Research Department of Zoology, Maharaja's College, Ernakulam, 682011, India
| | - Mithun Chacko John
- Department of Medical Oncology, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala 680005, India
| | - T Vanesa John
- Department of Pathology, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India
| | - Alex George
- Cell and Molecular Biology Facility, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 680005, Kerala, India.
| |
Collapse
|
17
|
Nogueras Pérez R, Heredia-Nicolás N, de Lara-Peña L, López de Andrés J, Marchal JA, Jiménez G, Griñán-Lisón C. Unraveling the Potential of miRNAs from CSCs as an Emerging Clinical Tool for Breast Cancer Diagnosis and Prognosis. Int J Mol Sci 2023; 24:16010. [PMID: 37958993 PMCID: PMC10647353 DOI: 10.3390/ijms242116010] [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: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BC) is the most diagnosed cancer in women and the second most common cancer globally. Significant advances in BC research have led to improved early detection and effective therapies. One of the key challenges in BC is the presence of BC stem cells (BCSCs). This small subpopulation within the tumor possesses unique characteristics, including tumor-initiating capabilities, contributes to treatment resistance, and plays a role in cancer recurrence and metastasis. In recent years, microRNAs (miRNAs) have emerged as potential regulators of BCSCs, which can modulate gene expression and influence cellular processes like BCSCs' self-renewal, differentiation, and tumor-promoting pathways. Understanding the miRNA signatures of BCSCs holds great promise for improving BC diagnosis and prognosis. By targeting BCSCs and their associated miRNAs, researchers aim to develop more effective and personalized treatment strategies that may offer better outcomes for BC patients, minimizing tumor recurrence and metastasis. In conclusion, the investigation of miRNAs as regulators of BCSCs opens new directions for advancing BC research through the use of bioinformatics and the development of innovative therapeutic approaches. This review summarizes the most recent and innovative studies and clinical trials on the role of BCSCs miRNAs as potential tools for early diagnosis, prognosis, and resistance.
Collapse
Affiliation(s)
- Raquel Nogueras Pérez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
| | - Noelia Heredia-Nicolás
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
| | - Laura de Lara-Peña
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
- Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
- Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
- Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
- Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18016 Granada, Spain; (R.N.P.); (N.H.-N.); (L.d.L.-P.); (J.L.d.A.); (J.A.M.)
- Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| |
Collapse
|
18
|
Tewari AB, Saini A, Sharma D. Extirpating the cancer stem cell hydra: Differentiation therapy and Hyperthermia therapy for targeting the cancer stem cell hierarchy. Clin Exp Med 2023; 23:3125-3145. [PMID: 37093450 DOI: 10.1007/s10238-023-01066-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/02/2023] [Indexed: 04/25/2023]
Abstract
Ever since the discovery of cancer stem cells (CSCs), they have progressively attracted more attention as a therapeutic target. Like the mythical hydra, this subpopulation of cells seems to contribute to cancer immortality, spawning more cells each time that some components of the cancer cell hierarchy are destroyed. Traditional modalities focusing on cancer treatment have emphasized apoptosis as a route to eliminate the tumor burden. A major problem is that cancer cells are often in varying degrees of dedifferentiation contributing to what is known as the CSCs hierarchy and cells which are known to be resistant to conventional therapy. Differentiation therapy is an experimental therapeutic modality aimed at the conversion of malignant phenotype to a more benign one. Hyperthermia therapy (HT) is a modality exploiting the changes induced in cells by the application of heat produced to aid in cancer therapy. While differentiation therapy has been successfully employed in the treatment of acute myeloid leukemia, it has not been hugely successful for other cancer types. Mounting evidence suggests that hyperthermia therapy may greatly augment the effects of differentiation therapy while simultaneously overcoming many of the hard-to-treat facets of recurrent tumors. This review summarizes the progress made so far in integrating hyperthermia therapy with existing modules of differentiation therapy. The focus is on studies related to the successful application of both hyperthermia and differentiation therapy when used alone or in conjunction for hard-to-treat cancer cell niche with emphasis on combined approaches to target the CSCs hierarchy.
Collapse
Affiliation(s)
- Amit B Tewari
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Anamika Saini
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Deepika Sharma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India.
| |
Collapse
|
19
|
Zhang Y, Li Y, Wei Y, Cong L. Molecular Mechanism of Vitamin D Receptor Modulating Wnt/β-catenin Signaling Pathway in Gastric Cancer. J Cancer 2023; 14:3285-3294. [PMID: 37928423 PMCID: PMC10622995 DOI: 10.7150/jca.81034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 07/27/2023] [Indexed: 11/07/2023] Open
Abstract
Background: Gastric cancer is the most common gastrointestinal cancer worldwide. The latest data showed that it was the fourth leading cause of cancer-related death. The unobvious symptom and the difficulties lying in the early diagnosis largely affect the effect of the treatment. Therefore, it becomes particularly important to investigate the related genes and signal transduction pathways in gastric cancer. Our previous study found that the vitamin D receptor (VDR) gene FokI polymorphism may be associated with susceptibility to gastric cancer in the Chinese Han population. However, the mechanism of VDR affecting gastric cancer is unknown. In this study, we explored the molecular mechanism and the possible signaling pathway of VDR modulating carcinogenesis and progression of gastric cancer. Methods: The expression of VDR in gastric cancer cell lines was interfered by plasmid transfection and RNA interference technology. And then we analyzed the cell viability and invasive ability by MTT assay, colony formation assay, and transwell migration assay, and detected the expression of VDR and several signaling proteins in gastric cancer cells by SDS-PAGE and Western blotting. Results: The overexpression of VDR can significantly inhibit the viability and invasive ability of gastric cancer cells; on the contrary, when VDR siRNA inhibits the expression of VDR, the viability and invasive ability of gastric cancer cells enhanced. VDR expression levels in gastric cancer cells treated with 1,25 (OH) 2D3 showed a time-dependent increased expression; and with the increase of the VDR expression, the expression of β-catenin decreased gradually, but the expression of E-cadherin showed a time-dependent increase (P < 0.05). Compared with the mutant-type VDR gene(ff) cells, the degree of β-catenin decline was significantly enhanced after transfected with homozygous wild-type VDR gene (FF) plasmids (p<0.05). Conclusions: The results of this study indicate that VDR FokI polymorphism plays an important role in the malignant phenotype of gastric cancer cells, such as proliferation, invasion, and clone formation. When the VDR is activated by its ligand, it can prevent the nuclear import of β-catenin, affect the E-cadherin level, inhibit the proliferation of gastric cancer cells, which suggested that VDR FokI gene may play a role of cancer suppressor via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Radiotherapy, Cangzhou Central Hospital, Hebei, China
| | - Yan Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuzheng Wei
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Cong
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
20
|
Yang X, Du Y, Luo L, Xu X, Xiong S, Yang X, Guo L, Liang T. Deciphering the Enigmatic Influence: Non-Coding RNAs Orchestrating Wnt/β-Catenin Signaling Pathway in Tumor Progression. Int J Mol Sci 2023; 24:13909. [PMID: 37762212 PMCID: PMC10530696 DOI: 10.3390/ijms241813909] [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: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Dysregulated expression of specific non-coding RNAs (ncRNAs) has been strongly linked to tumorigenesis, cancer progression, and therapeutic resistance. These ncRNAs can act as either oncogenes or tumor suppressors, thereby serving as valuable diagnostic and prognostic markers. Numerous studies have implicated the participation of ncRNAs in the regulation of diverse signaling pathways, including the pivotal Wnt/β-catenin signaling pathway that is widely acknowledged for its pivotal role in embryogenesis, cellular proliferation, and tumor biology control. Recent emerging evidence has shed light on the capacity of ncRNAs to interact with key components of the Wnt/β-catenin signaling pathway, thereby modulating the expression of Wnt target genes in cancer cells. Notably, the activity of this pathway can reciprocally influence the expression levels of ncRNAs. However, comprehensive analysis investigating the specific ncRNAs associated with the Wnt/β-catenin signaling pathway and their intricate interactions in cancer remains elusive. Based on these noteworthy findings, this review aims to unravel the intricate associations between ncRNAs and the Wnt/β-catenin signaling pathway during cancer initiation, progression, and their potential implications for therapeutic interventions. Additionally, we provide a comprehensive overview of the characteristics of ncRNAs and the Wnt/β-catenin signaling pathway, accompanied by a thorough discussion of their functional roles in tumor biology. Targeting ncRNAs and molecules associated with the Wnt/β-catenin signaling pathway may emerge as a promising and effective therapeutic strategy in future cancer treatments.
Collapse
Affiliation(s)
- Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Lulu Luo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Xinru Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Shizheng Xiong
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Xueni Yang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| |
Collapse
|
21
|
Farhan M. Insights on the Role of Polyphenols in Combating Cancer Drug Resistance. Biomedicines 2023; 11:1709. [PMID: 37371804 PMCID: PMC10296548 DOI: 10.3390/biomedicines11061709] [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: 05/22/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chemotherapy resistance is still a serious problem in the treatment of most cancers. Many cellular and molecular mechanisms contribute to both inherent and acquired drug resistance. They include the use of unaffected growth-signaling pathways, changes in the tumor microenvironment, and the active transport of medicines out of the cell. The antioxidant capacity of polyphenols and their potential to inhibit the activation of procarcinogens, cancer cell proliferation, metastasis, and angiogenesis, as well as to promote the inhibition or downregulation of active drug efflux transporters, have been linked to a reduced risk of cancer in epidemiological studies. Polyphenols also have the ability to alter immunological responses and inflammatory cascades, as well as trigger apoptosis in cancer cells. The discovery of the relationship between abnormal growth signaling and metabolic dysfunction in cancer cells highlights the importance of further investigating the effects of dietary polyphenols, including their ability to boost the efficacy of chemotherapy and avoid multidrug resistance (MDR). Here, it is summarized what is known regarding the effectiveness of natural polyphenolic compounds in counteracting the resistance that might develop to cancer drugs as a result of a variety of different mechanisms.
Collapse
Affiliation(s)
- Mohd Farhan
- Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Ahsa 31982, Saudi Arabia
| |
Collapse
|
22
|
Liu F, Wu Q, Dong Z, Liu K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol Ther 2023:108458. [PMID: 37245545 DOI: 10.1016/j.pharmthera.2023.108458] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Integrins are vital surface adhesion receptors that mediate the interactions between the extracellular matrix (ECM) and cells and are essential for cell migration and the maintenance of tissue homeostasis. Aberrant integrin activation promotes initial tumor formation, growth, and metastasis. Recently, many lines of evidence have indicated that integrins are highly expressed in numerous cancer types and have documented many functions of integrins in tumorigenesis. Thus, integrins have emerged as attractive targets for the development of cancer therapeutics. In this review, we discuss the underlying molecular mechanisms by which integrins contribute to most of the hallmarks of cancer. We focus on recent progress on integrin regulators, binding proteins, and downstream effectors. We highlight the role of integrins in the regulation of tumor metastasis, immune evasion, metabolic reprogramming, and other hallmarks of cancer. In addition, integrin-targeted immunotherapy and other integrin inhibitors that have been used in preclinical and clinical studies are summarized.
Collapse
Affiliation(s)
- Fangfang Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China
| | - Qiong Wu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zigang Dong
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Kangdong Liu
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450008, China; Department of Pathophysiology, School of Basic Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou, Henan 450000, China; Tianjian Advanced Biomedical Laboratory, Zhengzhou University, Zhengzhou, Henan 450001, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan 450000, China.
| |
Collapse
|
23
|
de Azevedo ALK, Carvalho TM, Mara CS, Giner IS, de Oliveira JC, Gradia DF, Cavalli IJ, Ribeiro EMSF. Major regulators of the multi-step metastatic process are potential therapeutic targets for breast cancer management. Funct Integr Genomics 2023; 23:171. [PMID: 37211553 DOI: 10.1007/s10142-023-01097-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
Metastasis is a multi-step process that leads to the dissemination of tumor cells to new sites and, consequently, to multi-organ neoplasia. Although most lethal breast cancer cases are related to metastasis occurrence, little is known about the dysregulation of each step, and clinicians still lack reliable therapeutic targets for metastasis impairment. To fill these gaps, we constructed and analyzed gene regulatory networks for each metastasis step (cell adhesion loss, epithelial-to-mesenchymal transition, and angiogenesis). Through topological analysis, we identified E2F1, EGR1, EZH2, JUN, TP63, and miR-200c-3p as general hub-regulators, FLI1 for cell-adhesion loss specifically, and TRIM28, TCF3, and miR-429 for angiogenesis. Applying the FANMOD algorithm, we identified 60 coherent feed-forward loops regulating metastasis-related genes associated with distant metastasis-free survival prediction. miR-139-5p, miR-200c-3p, miR-454-3p, and miR-1301-3p, among others, were the FFL's mediators. The expression of the regulators and mediators was observed to impact overall survival and to go along with metastasis occurrence. Lastly, we selected 12 key regulators and observed that they are potential therapeutic targets for canonical and candidate antineoplastics and immunomodulatory drugs, like trastuzumab, goserelin, and calcitriol. Our results highlight the relevance of miRNAs in mediating feed-forward loops and regulating the expression of metastasis-related genes. Altogether, our results contribute to understanding the multi-step metastasis complexity and identifying novel therapeutic targets and drugs for breast cancer management.
Collapse
Affiliation(s)
| | | | - Cristiane Sato Mara
- Genetics Department, Federal University of Parana, P.O. Box 19071, CEP, Curitiba, Parana, 81531-990, Brazil
| | - Igor Samesima Giner
- Genetics Department, Federal University of Parana, P.O. Box 19071, CEP, Curitiba, Parana, 81531-990, Brazil
| | | | - Daniela Fiori Gradia
- Genetics Department, Federal University of Parana, P.O. Box 19071, CEP, Curitiba, Parana, 81531-990, Brazil
| | - Iglenir João Cavalli
- Genetics Department, Federal University of Parana, P.O. Box 19071, CEP, Curitiba, Parana, 81531-990, Brazil
| | - Enilze M S F Ribeiro
- Genetics Department, Federal University of Parana, P.O. Box 19071, CEP, Curitiba, Parana, 81531-990, Brazil.
| |
Collapse
|
24
|
Cole AJ, Panesso-Gómez S, Shah JS, Ebai T, Jiang Q, Gumusoglu-Acar E, Bello MG, Vlad A, Modugno F, Edwards RP, Buckanovich RJ. Quiescent Ovarian Cancer Cells Secrete Follistatin to Induce Chemotherapy Resistance in Surrounding Cells in Response to Chemotherapy. Clin Cancer Res 2023; 29:1969-1983. [PMID: 36795892 PMCID: PMC10192102 DOI: 10.1158/1078-0432.ccr-22-2254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
PURPOSE We recently reported that the transcription factor NFATC4, in response to chemotherapy, drives cellular quiescence to increase ovarian cancer chemoresistance. The goal of this work was to better understand the mechanisms of NFATC4-driven ovarian cancer chemoresistance. EXPERIMENTAL DESIGN We used RNA sequencing to identify NFATC4-mediated differential gene expression. CRISPR-Cas9 and FST (follistatin)-neutralizing antibodies were used to assess impact of loss of FST function on cell proliferation and chemoresistance. ELISA was used to quantify FST induction in patient samples and in vitro in response to chemotherapy. RESULTS We found that NFATC4 upregulates FST mRNA and protein expression predominantly in quiescent cells and FST is further upregulated following chemotherapy treatment. FST acts in at least a paracrine manner to induce a p-ATF2-dependent quiescent phenotype and chemoresistance in non-quiescent cells. Consistent with this, CRISPR knockout (KO) of FST in ovarian cancer cells or antibody-mediated neutralization of FST sensitizes ovarian cancer cells to chemotherapy treatment. Similarly, CRISPR KO of FST in tumors increased chemotherapy-mediated tumor eradication in an otherwise chemotherapy-resistant tumor model. Suggesting a role for FST in chemoresistance in patients, FST protein in the abdominal fluid of patients with ovarian cancer significantly increases within 24 hours of chemotherapy exposure. FST levels decline to baseline levels in patients no longer receiving chemotherapy with no evidence of disease. Furthermore, elevated FST expression in patient tumors is correlated with poor progression-free, post-progression-free, and overall survival. CONCLUSIONS FST is a novel therapeutic target to improve ovarian cancer response to chemotherapy and potentially reduce recurrence rates.
Collapse
Affiliation(s)
- Alexander J. Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Santiago Panesso-Gómez
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaynish S. Shah
- Australian Centre for Blood Diseases, Central Clinical School, Monash University and Alfred Health, Melbourne, VIC, Australia
| | - Tonge Ebai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qi Jiang
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Ece Gumusoglu-Acar
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maya G. Bello
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anda Vlad
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francesmary Modugno
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert P. Edwards
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ronald J. Buckanovich
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Hashemi M, Hasani S, Hajimazdarany S, Ghadyani F, Olyaee Y, Khodadadi M, Ziyarani MF, Dehghanpour A, Salehi H, Kakavand A, Goharrizi MASB, Aref AR, Salimimoghadam S, Akbari ME, Taheriazam A, Hushmandi K, Entezari M. Biological functions and molecular interactions of Wnt/β-catenin in breast cancer: Revisiting signaling networks. Int J Biol Macromol 2023; 232:123377. [PMID: 36702226 DOI: 10.1016/j.ijbiomac.2023.123377] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/27/2022] [Accepted: 01/15/2023] [Indexed: 01/24/2023]
Abstract
Changes in lifestyle such as physical activity and eating habits have been one of the main reasons for development of various diseases in modern world, especially cancer. However, role of genetic factors in initiation of cancer cannot be ignored and Wnt/β-catenin signaling is such factor that can affect tumor progression. Breast tumor is the most malignant tumor in females and it causes high mortality and morbidity around the world. The survival and prognosis of patients are not still desirable, although there have been advances in introducing new kinds of therapies and diagnosis. The present review provides an update of Wnt/β-catenin function in breast cancer malignancy. The upregulation of Wnt is commonly observed during progression of breast tumor and confirms that tumor cells are dependent on this pathway Wnt/β-catenin induction prevents apoptosis that is of importance for mediating drug resistance. Furthermore, Wnt/β-catenin signaling induces DNA damage repair in ameliorating radio-resistance. Wnt/β-catenin enhances proliferation and metastasis of breast tumor. Wnt/β-catenin induces EMT and elevates MMP expression. Furthermore, Wnt/β-catenin participates in tumor microenvironment remodeling and due to its tumor-promoting factor, drugs for its suppression have been developed. Different kinds of upstream mediators Wnt/β-catenin signaling in breast cancer have been recognized that their targeting is a therapeutic approach. Finally, Wnt/β-catenin can be considered as a biomarker in clinical trials.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sahar Hasani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shima Hajimazdarany
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Ghadyani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yeganeh Olyaee
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Marzieh Khodadadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Fallah Ziyarani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Dehghanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hasti Salehi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc., 6 Tide Street, Boston, MA 02210, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
26
|
Liu P, Kong L, Liu Y, Li G, Xie J, Lu X. A key driver to promote HCC: Cellular crosstalk in tumor microenvironment. Front Oncol 2023; 13:1135122. [PMID: 37007125 PMCID: PMC10050394 DOI: 10.3389/fonc.2023.1135122] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
Liver cancer is the third greatest cause of cancer-related mortality, which of the major pathological type is hepatocellular carcinoma (HCC) accounting for more than 90%. HCC is characterized by high mortality and is predisposed to metastasis and relapse, leading to a low five-year survival rate and poor clinical prognosis. Numerous crosstalk among tumor parenchymal cells, anti-tumor cells, stroma cells, and immunosuppressive cells contributes to the immunosuppressive tumor microenvironment (TME), in which the function and frequency of anti-tumor cells are reduced with that of associated pro-tumor cells increasing, accordingly resulting in tumor malignant progression. Indeed, sorting out and understanding the signaling pathways and molecular mechanisms of cellular crosstalk in TME is crucial to discover more key targets and specific biomarkers, so that develop more efficient methods for early diagnosis and individualized treatment of liver cancer. This piece of writing offers insight into the recent advances in HCC-TME and reviews various mechanisms that promote HCC malignant progression from the perspective of mutual crosstalk among different types of cells in TME, aiming to assist in identifying the possible research directions and methods in the future for discovering new targets that could prevent HCC malignant progression.
Collapse
Affiliation(s)
- Pengyue Liu
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Lingyu Kong
- Department of Traditional Chinese Medicine, Affiliated Hospital of North China University of Science and Technology, Tangshan, China
| | - Ying Liu
- Department of Clinical Skills Training Center, Tangshan Gongren Hospital, Tangshan, China
| | - Gang Li
- Department of Clinical Laboratory, Tangshan Maternal and Child Health Care Hospital, Tangshan, China
| | - Jianjia Xie
- Department of Clinical Laboratory, Tangshan Maternal and Child Health Care Hospital, Tangshan, China
| | - Xin Lu
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
- Department of Clinical Laboratory, Tangshan Maternal and Child Health Care Hospital, Tangshan, China
| |
Collapse
|
27
|
Zhao C, Chen Z, Zhu L, Miao Y, Guo J, Yuan Z, Wang P, Li L, Ning W. The BMP inhibitor follistatin-like 1 (FSTL1) suppresses cervical carcinogenesis. Front Oncol 2023; 13:1100045. [PMID: 36756161 PMCID: PMC9901576 DOI: 10.3389/fonc.2023.1100045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Follistatin-like 1 (FSTL1) is a cancer-related matricellular secretory protein with contradictory organ-specific roles. Its contribution to the pathogenesis of cervical carcinoma is still not clear. Meanwhile, it is necessary to identify novel candidate genes to understand cervical carcinoma's pathogenesis further and find potential therapeutic targets. We collected cervical carcinoma samples and matched adjacent tissues from patients with the locally-advanced disease and used cervical carcinoma cell lines HeLa and C33A to evaluate the effects of FSTL1 on CC cells. The mRNA transcription and protein expression of FSTL1 in cervical carcinoma tumor biopsy tissues were lower than those of matched adjacent tissues. Patients with a lower ratio of FSTL1 mRNA between the tumor and its matched adjacent tissues showed a correlation with the advanced cervical carcinoma FIGO stages. High expression of FSTL1 markedly inhibited the proliferation, motility, and invasion of HeLa and C33A. Regarding mechanism, FSTL1 plays its role by negatively regulating the BMP4/Smad1/5/9 signaling. Our study has demonstrated the tumor suppressor effect of FSTL1, and these findings suggested a potential therapeutic target and biomarker for cervical carcinoma.
Collapse
Affiliation(s)
- Chenjing Zhao
- State Key Laboratory of Medical Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhongjie Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Li Zhu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yunheng Miao
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jiasen Guo
- State Key Laboratory of Medical Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lian Li
- State Key Laboratory of Medical Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China,*Correspondence: Wen Ning, ; Lian Li,
| | - Wen Ning
- State Key Laboratory of Medical Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China,*Correspondence: Wen Ning, ; Lian Li,
| |
Collapse
|
28
|
Jayaraman H, Anandhapadman A, Ghone NV. In Vitro and In Vivo Comparative Analysis of Differentially Expressed Genes and Signaling Pathways in Breast Cancer Cells on Interaction with Mesenchymal Stem Cells. Appl Biochem Biotechnol 2023; 195:401-431. [PMID: 36087230 DOI: 10.1007/s12010-022-04119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2022] [Indexed: 01/13/2023]
Abstract
The interaction of breast cancer cells (BCC) with mesenchymal stem cells (MSC) plays a vital role in influencing the gene expression in breast cancer cells and thereby its uncontrolled proliferation, metastasis, and drug resistance. The extent of MSC governing the BCC or the extent of BCC influencing the MSC is a complex process, and the interaction strongly depends upon conditions such as the presence or absence of other cell types and in vivo tumor microenvironment or simple in vitro conditions. Hence, understanding this interaction through gene expression profiling may provide key insights about potential genes which can be targeted for breast cancer treatment. In the current study, in vitro microarray dataset and in vivo RNA-seq dataset of BCC on interaction with the MSC were downloaded from NCBI GEO database and analyzed for differentially expressed genes (DEGs), gene ontology (GO) term enrichment, and Reactome pathway analysis. To target the genes which have similar effect on both in vitro and in vivo, a comparative analysis was performed, 24 genes were commonly upregulated in both in vitro and in vivo datasets, while no common downregulated genes were observed. Out of which, 16 significant genes based upon fold change (logFC > 2) are identified for manipulating the interactions between MSC and BCC. Among them, 6 of the identified genes (FSTL1, LOX, SERPINE1, INHBA, FN1, and VEGFA) have already been reported to be upregulated in BCC on interaction with MSC by various studies. Further experiments need to be conducted to understand the role of remaining 10 identified genes (EFEMP1, IGFBP3, EDIL3, IFITM1, IGFBP4, ITGA5, SLC3A2, HRH1, PPP1R15A, and NNMT) in MSC-BCC interaction. In addition to the reported significant genes and its associated pathways, the expression of long non-coding RNA identified in this study may increase our understanding about the way MSC interacts with BCC and accelerate MSC-based treatment strategies for breast cancer.
Collapse
Affiliation(s)
- Hariharan Jayaraman
- Department of Biotechnology, Sri Venkateswara College of Engineering, Post Bag No. 1, Sriperumbudur Taluk, 602117, Kancheepuram, Tamil Nadu, India
| | - Ashwin Anandhapadman
- Department of Biotechnology, Sri Venkateswara College of Engineering, Post Bag No. 1, Sriperumbudur Taluk, 602117, Kancheepuram, Tamil Nadu, India
| | - Nalinkanth Veerabadran Ghone
- Department of Chemical Engineering, Sri Sivasubramaniya Nadar College of Engineering, Rajiv Gandhi Salai (OMR), Kalavakkam, 603110, Tamil Nadu, India.
| |
Collapse
|
29
|
Fu Y, Yang Q, Yang H, Zhang X. New progress in the role of microRNAs in the diagnosis and prognosis of triple negative breast cancer. Front Mol Biosci 2023; 10:1162463. [PMID: 37122564 PMCID: PMC10134903 DOI: 10.3389/fmolb.2023.1162463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Triple negative breast cancer is distinguished by its high malignancy, aggressive invasion, rapid progression, easy recurrence, and distant metastases. Additionally, it has a poor prognosis, a high mortality, and is unresponsive to conventional endocrine and targeted therapy, making it a challenging problem for breast cancer treatment and a hotspot for scientific research. Recent research has revealed that certain miRNA can directly or indirectly affect the occurrence, progress and recurrence of TNBC. Their expression levels have a significant impact on TNBC diagnosis, treatment and prognosis. Some miRNAs can serve as biomarkers for TNBC diagnosis and prognosis. This article summarizes the progress of miRNA research in TNBC, discusses their roles in the occurrence, invasion, metastasis, prognosis, and chemotherapy of TNBC, and proposes a treatment strategy for TNBC by interfering with miRNA expression levels.
Collapse
Affiliation(s)
- Yeqin Fu
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiuhui Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongjian Yang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- *Correspondence: Hongjian Yang, ; Xiping Zhang,
| | - Xiping Zhang
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- *Correspondence: Hongjian Yang, ; Xiping Zhang,
| |
Collapse
|
30
|
MicroRNAs: A Link between Mammary Gland Development and Breast Cancer. Int J Mol Sci 2022; 23:ijms232415978. [PMID: 36555616 PMCID: PMC9786715 DOI: 10.3390/ijms232415978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is among the most common cancers in women, second to skin cancer. Mammary gland development can influence breast cancer development in later life. Processes such as proliferation, invasion, and migration during mammary gland development can often mirror processes found in breast cancer. MicroRNAs (miRNAs), small, non-coding RNAs, can repress post-transcriptional RNA expression and can regulate up to 80% of all genes. Expression of miRNAs play a key role in mammary gland development, and aberrant expression can initiate or promote breast cancer. Here, we review the role of miRNAs in mammary development and breast cancer, and potential parallel roles. A total of 32 miRNAs were found to be expressed in both mammary gland development and breast cancer. These miRNAs are involved in proliferation, metastasis, invasion, and apoptosis in both processes. Some miRNAs were found to have contradictory roles, possibly due to their ability to target many genes at once. Investigation of miRNAs and their role in mammary gland development may inform about their role in breast cancer. In particular, by studying miRNA in development, mechanisms and potential targets for breast cancer treatment may be elucidated.
Collapse
|
31
|
Upregulation of Actin-Related Protein 2 (ACTR2) Exacerbated the Malignancy of Diffuse Large B-Cell Lymphoma through Activating Wnt Signaling. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9351921. [PMID: 36570337 PMCID: PMC9771665 DOI: 10.1155/2022/9351921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/29/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022]
Abstract
This investigation mainly explores the roles of actin-related protein 2 (ACTR2) in diffuse large B-cell lymphoma (DLBCL). We first assessed the level of ACTR2 and its association with the overall survival (OS) of DLBCL. The results indicated that ACTR2 was upregulated in DLBCL and was associated with unfavorable prognosis of DLBCL. Next, the effect of ACTR2 knockdown or overexpression on DLBCL was evaluated in vitro. Our investigation revealed that ACTR2 depletion inhibited the malignant behaviors of DLBCL cells; whereas, ACTR2 abundance promoted those behaviors. Besides, ACTR2 activated the Wnt signaling in DLBCL and exerted its oncogenic influence on DLBCL through Wnt signaling in vitro and in vivo. To summarize, our study implicated that ACTR2 was a promising therapeutic target for DLBCL, which might become a novel direction to improve our understanding on DLBCL.
Collapse
|
32
|
Li Y, Jiang C, Zhang X, Liao Z, Chen L, Li S, Tang S, Fan Z, Zhang Q. Inhibition of ABCC9 by zinc oxide nanoparticles induces ferroptosis and inhibits progression, attenuates doxorubicin resistance in breast cancer. Cancer Nanotechnol 2022; 13:3. [DOI: 10.1186/s12645-021-00109-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 01/21/2023] Open
Abstract
Abstract
Background
Zinc oxide nanoparticles (ZONs) are a type of nanomaterial that has presented anti-cancer properties in breast cancer (BC). However, the function of ABCC9 in BC and its correlation with ZONs are still elusive.
Methods
Here, we identified the crucial role of ABCC9 in modulating ferroptosis and doxorubicin (Dox) resistance in BC and the targeted function of ZONs to ABCC9.
Results
The silencing of ABCC9 significantly repressed the viability of BC cells. The knockdown of ABCC9 decreased the numbers of Edu-positive BC cells. Conversely, BC cell apoptosis was increased by the inhibition of ABCC9. Besides, the silencing of ABCC9 reduced the capability of migration and invasion of BC cells. Significantly, tumorigenicity analysis demonstrated that the tumor growth of BC cells was suppressed by the depletion of ABCC9 in the xenograft model of nude mice. Moreover, the treatment of ferroptosis activator erastin repressed cell viability of BC cells and ABCC9 overexpression rescued the repression. Similarly, the numbers of Edu-positive BC cells were inhibited by erastin and the overexpression of ABCC9 reversed the inhibitory effect of erastin. The levels of GSH were decreased and MDA, lipid ROS, and iron levels were increased by the treatment of erastin, while the ABCC9 overexpression could reverse these results in BC cells. Consistently, erastin suppressed the expression of ferroptosis inhibitory factors, including GPX4 and SLC7A11, in BC cells and the overexpression of ABCC9 rescued the expression. The IC50 value of Dox was reduced by the knockdown of ABCC9 in Dox-resistant BC cells (BC/Dox). The numbers of Edu-positive BC/Dox cells were attenuated by the depletion of ABCC9. Meanwhile, the apoptosis of BC/Dox cells was stimulated by the silencing of ABCC9. Furthermore, the treatment of ZONs attenuated Dox resistance of BC cells. ZONs remarkably repressed the expression of ABCC9 in BC/Dox cells. ZONs inhibited the cell viability of BC/Dox cells and the overexpression of ABCC9 reversed the repression. Moreover, the treatment of ZONs reduced GSH levels and enhanced MDA, lipid ROS, and iron levels in erastin-stimulated BC/Dox cells.
Conclusions
In conclusion, we discovered that the inhibition of ABCC9 by zinc oxide nanoparticles induces ferroptosis and attenuates Dox resistance in BC.
Collapse
|
33
|
Horak M, Fairweather D, Kokkonen P, Bednar D, Bienertova-Vasku J. Follistatin-like 1 and its paralogs in heart development and cardiovascular disease. Heart Fail Rev 2022; 27:2251-2265. [PMID: 35867287 PMCID: PMC11140762 DOI: 10.1007/s10741-022-10262-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 11/29/2022]
Abstract
Cardiovascular diseases (CVDs) are a group of disorders affecting the heart and blood vessels and a leading cause of death worldwide. Thus, there is a need to identify new cardiokines that may protect the heart from damage as reported in GBD 2017 Causes of Death Collaborators (2018) (The Lancet 392:1736-1788). Follistatin-like 1 (FSTL1) is a cardiokine that is highly expressed in the heart and released to the serum after cardiac injury where it is associated with CVD and predicts poor outcome. The action of FSTL1 likely depends not only on the tissue source but also post-translation modifications that are target tissue- and cell-specific. Animal studies examining the effect of FSTL1 in various models of heart disease have exploded over the past 15 years and primarily report a protective effect spanning from inhibiting inflammation via transforming growth factor, preventing remodeling and fibrosis to promoting angiogenesis and hypertrophy. A better understanding of FSTL1 and its homologs is needed to determine whether this protein could be a useful novel biomarker to predict poor outcome and death and whether it has therapeutic potential. The aim of this review is to provide a comprehensive description of the literature for this family of proteins in order to better understand their role in normal physiology and CVD.
Collapse
Affiliation(s)
- Martin Horak
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Piia Kokkonen
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - David Bednar
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Julie Bienertova-Vasku
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| |
Collapse
|
34
|
Wu P, Guo J, Yang H, Yuan D, Wang C, Wang Z. Exosomes Derived from Hypoxic Glioma Cells Reduce the Sensitivity of Glioma Cells to Temozolomide Through Carrying miR-106a-5p. Drug Des Devel Ther 2022; 16:3589-3598. [PMID: 36248244 PMCID: PMC9556335 DOI: 10.2147/dddt.s382690] [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: 07/17/2022] [Accepted: 09/17/2022] [Indexed: 11/06/2022] Open
Abstract
Background Hypoxia is a frequent feature of solid tumors which significantly affects the efficacy of treatments such as chemotherapy. In addition, exosomes from hypoxic cancer cells could contribute to the chemoresistance of tumor cells through carrying miRNAs. It has been shown that miR-106-5p level was upregulated in glioma. However, whether exosomes derived from hypoxic glioma cells could affect temozolomide (TMZ) resistance in glioma through carrying miR-106a-5p remains unexplored. Methods Exosomes were isolated from glioma cells under normoxia or hypoxia condition. EdU staining and flow cytometry assays were used to assess the cell proliferation and cell apoptosis. The relation between miR-106a-5p and PTEN was investigated by dual luciferase assay. Results MiR-106a-5p was enriched in exosomes derived from hypoxic glioma cells compared to exosomes from cells under normoxia condition. Additionally, hypoxic glioma cells were able to transfer exosomes to glioma cells, resulting in a significant increase of miR-106a-5p level in cells. TMZ remarkably suppressed glioma cell proliferation and triggered cell apoptosis. However, hypoxic glioma cell-derived exosomes markedly promoted the proliferation and suppressed the apoptosis in TMZ-treated glioma cells, and miR-106a-5p inhibitor was able to abolish these phenomena. Meanwhile, PTEN was verified to be a direct target of miR-106a-5p. Furthermore, TMZ elevated PTEN and Bax level and reduced p-Akt level in glioma cells, whereas these changes were reversed by hypoxia glioma cell-derived exosomes. Furthermore, hypoxia glioma cell-derived exosomes reduced the sensitivity of glioma cells to TMZ in vivo via downregulating PTEN. Conclusion Collectively, exosomal miR-106a-5p derived from hypoxia glioma cells could reduce the sensitivity of glioma cells to TMZ through downregulating PTEN. Thus, our study might provide new strategies for improving the clinical efficacy of TMZ on glioma.
Collapse
Affiliation(s)
- Peizhang Wu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China,Department of Neurosurgery, Yancheng First People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Jun Guo
- Department of Neurosurgery, Yancheng First People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Hongwei Yang
- Department of Neurosurgery, Yancheng First People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Debin Yuan
- Department of Neurosurgery, Yancheng First People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Chaoxiang Wang
- Department of Neurosurgery, Yancheng First People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China,Correspondence: Zhong Wang, Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, 215006, People’s Republic of China, Email
| |
Collapse
|
35
|
Cancer stem cell markers interplay with chemoresistance in triple negative breast cancer: A therapeutic perspective. Bull Cancer 2022; 109:960-971. [DOI: 10.1016/j.bulcan.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/18/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022]
|
36
|
Yang M, Lu Z, Yu B, Zhao J, Li L, Zhu K, Ma M, Long F, Wu R, Hu G, Huang L, Chou J, Gong N, Yang K, Li X, Zhang Y, Lin C. COL5A1 Promotes the Progression of Gastric Cancer by Acting as a ceRNA of miR-137-3p to Upregulate FSTL1 Expression. Cancers (Basel) 2022; 14:3244. [PMID: 35805015 PMCID: PMC9264898 DOI: 10.3390/cancers14133244] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
MicroRNAs (miRNAs) and their target genes have been shown to play an important role in gastric cancer but have not been fully clarified. Therefore, our goal was to identify the key miRNA-mRNA regulatory network in gastric cancer by utilizing a variety of bioinformatics analyses and experiments. A total of 242 miRNAs and 1080 genes were screened from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), respectively. Then, survival-related differentially expressed miRNAs and their differentially expressed target genes were screened. Twenty hub genes were identified from their protein-protein interaction network. After weighted gene co-expression network analysis was conducted, we selected miR-137-3p and its target gene, COL5A1, for further research. We found that miR-137-3p was significantly downregulated and that overexpression of miR-137-3p suppressed the proliferation, invasion, and migration of gastric cancer cells. Furthermore, we found that its target gene, COL5A1, could regulate the expression of another hub gene, FSTL1, by sponging miR-137-3p, which was confirmed by dual-luciferase reporter assays. Knockdown of COL5A1 inhibited the proliferation, invasion, and migration of gastric cancer cells, which could be rescued by the miR-137-3p inhibitor or overexpression of FSTL1. Ultimately, bioinformatics analyses showed that the expression of FSTL1 was highly correlated with immune infiltration.
Collapse
Affiliation(s)
- Ming Yang
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Zhixing Lu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Bowen Yu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Jiajia Zhao
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Liang Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Kaiyu Zhu
- The Five-Year Program in Clinical Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Min Ma
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Fei Long
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Runliu Wu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Gui Hu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Lihua Huang
- Center for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China;
| | - Jing Chou
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Ni Gong
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Kaiyan Yang
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Xiaorong Li
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Yi Zhang
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, China; (M.Y.); (Z.L.); (B.Y.); (J.Z.); (L.L.); (M.M.); (F.L.); (R.W.); (G.H.); (J.C.); (N.G.); (K.Y.); (X.L.)
| |
Collapse
|
37
|
Hu X, Zhang Q, Xing W, Wang W. Role of microRNA/lncRNA Intertwined With the Wnt/β-Catenin Axis in Regulating the Pathogenesis of Triple-Negative Breast Cancer. Front Pharmacol 2022; 13:814971. [PMID: 35814205 PMCID: PMC9263262 DOI: 10.3389/fphar.2022.814971] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
Objective (s): In this mini-review, we aimed to discuss the Wnt/β-catenin signaling pathway modulation in triple-negative breast cancer, particularly the contribution of lncRNAs and miRNAs in its regulation and their possible entwining role in breast cancer pathogenesis, proliferation, migration, or malignancy.Background: Malignant tumor formation is very high for breast cancer in women and is a leading cause of death all over the globe. Among breast cancer subtypes, triple-negative breast cancer is rife in premenopausal women, most invasive, and prone to metastasis. Complex pathways are involved in this cancer’s pathogenesis, advancement, and malignancy, including the Wnt/β-catenin signaling pathway. This pathway is conserved among vertebrates and is necessary for sustaining cell homeostasis. It is regulated by several elements such as transcription factors, enhancers, non-coding RNAs (lncRNAs and miRNAs), etc.Methods: We evaluated lncRNAs and miRNAs differentially expressed in triple-negative breast cancer (TNBC) from the cDNA microarray data set literature survey. Using in silico analyses combined with a review of the current literature, we anticipated identifying lncRNAs and miRNAs that might modulate the Wnt/β-catenin signaling pathway.Result: The miRNAs and lncRNAs specific to triple-negative breast cancer have been identified based on literature and database searches. Tumorigenesis, metastasis, and EMT were all given special attention. Apart from cross-talk being essential for TNBC tumorigenesis and treatment outcomes, our results indicated eight upregulated and seven downregulated miRNAs and 19 upregulated and three downregulated lncRNAs that can be used as predictive or diagnostic markers. This consolidated information could be useful in the clinic and provide a combined literature resource for TNBC researchers working on the Wnt/β-catenin miRNA/lncRNA axis.Conclusion: In conclusion, because the Wnt pathway and miRNAs/lncRNAs can modulate TNBC, their intertwinement results in a cascade of complex reactions that affect TNBC and related processes. Their function in TNBC pathogenesis has been highlighted in molecular processes underlying the disease progression.
Collapse
Affiliation(s)
- Xue Hu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qiang Zhang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Wanying Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wan Wang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Wan Wang,
| |
Collapse
|
38
|
Zhang Y, Wang Y, Zheng G, Liu Y, Li J, Huang H, Xu C, Zeng Y, Zhang X, Qin J, Dai C, Hambrock HO, Hartmann U, Feng B, Mak KK, Liu Y, Lan HY, Huang Y, Zheng ZH, Xia Y. Follistatin-like 1 (FSTL1) interacts with Wnt ligands and Frizzled receptors to enhance Wnt/β-catenin signaling in obstructed kidneys in vivo. J Biol Chem 2022; 298:102010. [PMID: 35525270 PMCID: PMC9234244 DOI: 10.1016/j.jbc.2022.102010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Follistatin (FS)-like 1 (FSTL1) is a member of the FS-SPARC (secreted protein, acidic and rich in cysteine) family of secreted and extracellular matrix proteins. The functions of FSTL1 have been studied in heart and lung injury as well as in wound healing; however, the role of FSTL1 in the kidney is largely unknown. Here, we show using single-cell RNA-Seq that Fstl1 was enriched in stromal cells in obstructed mouse kidneys. In addition, immunofluorescence demonstrated that FSTL1 expression was induced in fibroblasts during kidney fibrogenesis in mice and human patients. We demonstrate that FSTL1 overexpression increased renal fibrosis and activated the Wnt/β-catenin signaling pathway, known to promote kidney fibrosis, but not the transforming growth factor β (TGF-β), Notch, Hedgehog, or Yes-associated protein (YAP) signaling pathways in obstructed mouse kidneys, whereas inhibition of FSTL1 lowered Wnt/β-catenin signaling. Importantly, we show that FSTL1 interacted with Wnt ligands and the Frizzled (FZD) receptors but not the coreceptor lipoprotein receptor–related protein 6 (LRP6). Specifically, we found FSTL1 interacted with Wnt3a through its extracellular calcium–binding (EC) domain and von Willebrand factor type C–like (VWC) domain, and with FZD4 through its EC domain. Furthermore, we show that FSTL1 increased the association of Wnt3a with FZD4 and promoted Wnt/β-catenin signaling and fibrogenesis. The EC domain interacting with both Wnt3a and FZD4 also enhanced Wnt3a signaling. Therefore, we conclude that FSTL1 is a novel extracellular enhancer of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yu Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Guoxun Zheng
- iHuman Institute, Shanghai Tech University, Shanghai 201210, China
| | - Yang Liu
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinhong Li
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Huihui Huang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Chunhua Xu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yelin Zeng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyi Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinzhong Qin
- The Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Chunsun Dai
- Center for Kidney Disease, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Harald O Hambrock
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Ursula Hartmann
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931 Cologne, Germany
| | - Bo Feng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Kingston Kinglun Mak
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, and The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Zhi-Hua Zheng
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, and The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
39
|
miR-1290 modulates the radioresistance of triple-negative breast cancer by targeting NLRP3-mediated pyroptosis. Clin Transl Oncol 2022; 24:1764-1775. [PMID: 35471684 DOI: 10.1007/s12094-022-02831-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/30/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE To explore the roles and underlying mechanisms of miR-1290 in determining the sensitivity of triple-negative breast cancer (TNBC) to radiation therapy. METHODS ELISA was performed to detect the levels of IL-18 and IL-1β in radiosensitive cells and serum samples. The level of miR-1290 in radiosensitive cells and tissues was assessed by qRT-PCR assay. A luciferase reporter assay was performed to confirm NLRP3 as the target of miR-1290. Functionally, the roles of miR-1290 in TNBC radioresistance were analyzed by transfection of either miR-1290 mimic or miR-1290 inhibitor. Moreover, the involvement of the miR-1290/NLRP3 axis in TNBC radioresistance was analyzed by experiments with a miR-1290 mimic and NLRP3 overexpression. MTT and colony formation assays were used to detect radiation-induced cell viability and proliferation. qRT-PCR and western blot assays were used to detect pyroptosis markers (NLRP3, ACS and caspase-1). RESULTS The results showed that radioresistance in TNBC cells was associated with a reduction in pyroptosis. miR-1290 expression was increased in radioresistant cells, and it had higher expression levels in the radioresistant tumor tissues of TNBC patients compared to the radiosensitive samples. The miR-1290 mimic suppressed radiation-induced pyroptosis and reduced the radiosensitivity of TNBC cells. Moreover, we found that NLRP3 was a potential target of miR-1290. Overexpression of NLRP3 partly reversed the effects of miR-1290 on pyroptosis and radioresistance. The mouse models showed that miR-1290 suppressed tumor size, tumor weight and pyroptosis. CONCLUSIONS miR-1290/NLRP3-mediated pyroptosis may play an important role in determining radioresistance in TNBC and serve as a novel therapeutic option.
Collapse
|
40
|
Tan X, Zhao J, Lou J, Zheng W, Wang P. Hsa_circ_0058129 regulates papillary thyroid cancer development via miR‐873‐5p/follistatin‐like 1 axis. J Clin Lab Anal 2022; 36:e24401. [PMID: 35373391 PMCID: PMC9102651 DOI: 10.1002/jcla.24401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 01/06/2023] Open
Abstract
Background Papillary thyroid cancer (PTC) is an endocrine malignancy with a high incidence. Circular RNAs (circRNAs) participate in regulating PTC. Here, we analyzed the role of hsa_circ_0058129 (circ_0058129) in PTC. Methods The expression of circ_0058129, fibronectin 1 (FN1) mRNA, microRNA‐873‐5p (miR‐873‐5p), and follistatin‐like 1 (FSTL1) was detected by qRT‐PCR and western blot. Cell proliferation was analyzed by CCK‐8, EdU, and flow cytometry analysis assays. Cell migration and invasion were evaluated by Transwell assay. The targeting relationship of miR‐873‐5p and circ_0058129 or FSTL1 was identified through dual‐luciferase reporter assay, RIP assay, and RNA pull‐down assay. Xenograft mouse model assay was implemented to determine the effect of circ_0058129 on tumor formation in vivo. Results The circ_0058129 and FSTL1 abundances were increased, while the miR‐873‐5p content was decreased in PTC tissues and cells compared with control groups. Circ_0058129 shortage inhibited PTC cell proliferation, migration, and invasion. Moreover, miR‐873‐5p repressed PTC cell malignancy by binding to FSTL1. Circ_0058129 targeted miR‐873‐5p to regulate FSTL1. Conclusion Circ_0058129 expedited PTC progression through the miR‐873‐5p/FSTL1 pathway.
Collapse
Affiliation(s)
- Xiangrong Tan
- Head and Neck Surgery The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| | - Jiazheng Zhao
- Head and Neck Surgery The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| | - Jianlin Lou
- Head and Neck Surgery The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| | - Wen Zheng
- Head and Neck Surgery The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| | - Peng Wang
- Head and Neck Surgery The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital) Institute of Basic Medicine and Cancer (IBMC) Chinese Academy of Sciences Hangzhou China
| |
Collapse
|
41
|
Yang X, Cao D, Ma W, Gao S, Wen G, Zhong J. Wnt signaling in triple-negative breast cancers: Its roles in molecular subtyping and cancer cell stemness and its crosstalk with non-coding RNAs. Life Sci 2022; 300:120565. [DOI: 10.1016/j.lfs.2022.120565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/11/2022] [Accepted: 04/16/2022] [Indexed: 12/20/2022]
|
42
|
Liu X, Yin Z, Wu Y, Zhan Q, Huang H, Fan J. Circular RNA lysophosphatidic acid receptor 3 (circ-LPAR3) enhances the cisplatin resistance of ovarian cancer. Bioengineered 2022; 13:3739-3750. [PMID: 35081867 PMCID: PMC8974081 DOI: 10.1080/21655979.2022.2029109] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Circular RNA (circRNA) is considered to be an important regulator that mediates cancer chemoresistance. But whether circ-LPAR3 is involved in ovarian cancer (OC) cisplatin (DDP) resistance is unclear. The circ-LPAR3, miR-634 and pyruvate dehydrogenase kinase 1 (PDK1) expression was measured by quantitative real-time PCR (qRT-PCR). Cell cisplatin resistance and viability were measured by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide (MTT) assay. In addition, cell colony number, apoptosis, and metastasis were assessed by colony formation assay, flow cytometry and transwell assay. Furthermore, in vivo experiments were performed by constructing mice xenograft models. RNA interaction was confirmed by dual-luciferase reporter assay, and PDK1 protein expression was examined by Western blot analysis. Our results showed that circ-LPAR3 was markedly upregulated in DDP-resistant OC tissues and cells. Silencing of circ-LPAR3 enhanced the DDP sensitivity of OC cells and tumors. MiR-634 could interact with circ-LPAR3, and its inhibitor overturned the regulation of si-circ-LPAR3 on cell DDP resistance. Additionally, PDK1 was targeted by miR-634, and its overexpression inverted the effect of miR-634 on cell DDP resistance. To sum up, circ-LPAR3 might contribute to the DDP resistance of OC via the miR-634/PDK1 axis.
Collapse
Affiliation(s)
- Xuemei Liu
- Department of Gynaecology and Obstetrics, Jinan City People's Hospital, Jinan, China
| | - Zhiping Yin
- Department of Laboratory Medicine, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Yanjun Wu
- Department of Gynaecology and Obstetrics, Liaocheng People's Hospital, Liaocheng, China
| | - Qian Zhan
- Department of Laboratory Medicine, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Honghong Huang
- Department of Pharmacy, Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jiangtao Fan
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
43
|
Maleki Dana P, Sadoughi F, Asemi Z, Yousefi B. The role of polyphenols in overcoming cancer drug resistance: a comprehensive review. Cell Mol Biol Lett 2022; 27:1. [PMID: 34979906 PMCID: PMC8903685 DOI: 10.1186/s11658-021-00301-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapeutic drugs are used to treat advanced stages of cancer or following surgery. However, cancers often develop resistance against drugs, leading to failure of treatment and recurrence of the disease. Polyphenols are a family of organic compounds with more than 10,000 members which have a three-membered flavan ring system in common. These natural compounds are known for their beneficial properties, such as free radical scavenging, decreasing oxidative stress, and modulating inflammation. Herein, we discuss the role of polyphenols (mainly curcumin, resveratrol, and epigallocatechin gallate [EGCG]) in different aspects of cancer drug resistance. Increasing drug uptake by tumor cells, decreasing drug metabolism by enzymes (e.g. cytochromes and glutathione-S-transferases), and reducing drug efflux are some of the mechanisms by which polyphenols increase the sensitivity of cancer cells to chemotherapeutic agents. Polyphenols also affect other targets for overcoming chemoresistance in cancer cells, including cell death (i.e. autophagy and apoptosis), EMT, ROS, DNA repair processes, cancer stem cells, and epigenetics (e.g. miRNAs).
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Xia M, Zu X, Chen Z, Wen G, Zhong J. Noncoding RNAs in triple negative breast cancer: Mechanisms for chemoresistance. Cancer Lett 2021; 523:100-110. [PMID: 34601022 DOI: 10.1016/j.canlet.2021.09.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype among breast cancers with high recurrence and this condition is partly due to chemoresistance. Therefore, fully understanding the mechanism of TNBC-resistance is the key to overcoming chemoresistance, which will be an effective strategy for TNBC therapy. Various potential mechanisms involved in the chemoresistance of TNBC have been investigated and indicated that noncoding RNAs (ncRNAs) especially microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) take part in most TNBC resistance. The ncRNA-induced chemoresistance process is involved in the alteration of many activities. here, we mainly summarize the mechanisms of ncRNAs in the chemoresistance of TNBC and discuss the potential clinical application of ncRNAs in the treatment of TNBC, indicating that targeting ncRNAs might be a promising strategy for resensitization to chemotherapies.
Collapse
Affiliation(s)
- Min Xia
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xuyu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Zuyao Chen
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Gebo Wen
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Jing Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
45
|
Thomas-Jardin SE, Kanchwala MS, Dahl H, Liu V, Ahuja R, Soundharrajan R, Roos N, Diep S, Sandhu A, Xing C, Delk NA. Chronic IL-1 Exposed AR + PCa Cell Lines Show Conserved Loss of IL-1 Sensitivity and Evolve Both Conserved and Unique Differential Gene Expression Profiles. JOURNAL OF CELLULAR SIGNALING 2021; 2:248-260. [PMID: 34988553 PMCID: PMC8725614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Inflammation drives prostate cancer (PCa) progression. While inflammation is a cancer hallmark, the underlying mechanisms mediating inflammation-induced PCa are still under investigation. Interleukin-1 (IL-1) is an inflammatory cytokine that promotes cancer progression, including PCa metastasis and castration resistance. We previously found that acute IL-1 exposure represses PCa androgen receptor (AR) expression concomitant with the upregulation of pro-survival proteins, causing de novo accumulation of castration-resistant PCa cells. However, acute inflammation is primarily anti-tumorigenic, while chronic inflammation is pro-tumorigenic. Thus, using the LNCaP PCa cell line as model, we found that PCa cells can evolve insensitivity to chronic IL-1 exposure, restoring AR and AR activity and acquiring castration resistance. In this paper we expanded our chronic IL-1 model to include the MDA-PCa-2b PCa cell line to investigate the response to acute versus chronic IL-1 exposure and to compare the gene expression patterns that evolve in the LNCaP and MDA-PCa-2b cells chronically exposed to IL-1. METHODS We chronically exposed MDA-PCa-2b cells to IL-1α or IL-1β for several months to establish sublines. Once established, we determined subline sensitivity to exogenous IL-1 using cell viability assay, RT-qPCR and western blot. RNA sequencing was performed for parental and subline cells and over representation analysis (ORA) for geneset enrichment of biological process/pathway was performed. RESULTS MDA-PCa-2b cells repress AR and AR activity in response to acute IL-1 exposure and evolve insensitivity to chronic IL-1 exposure. While cell biological and molecular response to acute IL-1 signaling is primarily conserved in LNCaP and MDA-PCa-2b cells, including upregulation of NF-κB signaling and downregulation of cell proliferation, the LNCaP and MDA-PCa-2b cells evolve conserved and unique molecular responses to chronic IL-1 signaling that may promote or support tumor progression. CONCLUSIONS Our chronic IL-1 subline models can be used to identify underlying molecular mechanisms that mediate IL-1-induced PCa progression.
Collapse
Affiliation(s)
- Shayna E. Thomas-Jardin
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Mohammed S. Kanchwala
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haley Dahl
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Vivian Liu
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Rohan Ahuja
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Reshma Soundharrajan
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Nicole Roos
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Sydney Diep
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Amrit Sandhu
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Chao Xing
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA,Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA,Department of Population and Data Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080, USA,Correspondence should be addressed to Nikki A. Delk;
| |
Collapse
|
46
|
Loh JJ, Li TW, Zhou L, Wong TL, Liu X, Ma VWS, Lo CM, Man K, Lee TK, Ning W, Tong M, Ma S. FSTL1 Secreted by Activated Fibroblasts Promotes Hepatocellular Carcinoma Metastasis and Stemness. Cancer Res 2021; 81:5692-5705. [PMID: 34551961 DOI: 10.1158/0008-5472.can-20-4226] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/26/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022]
Abstract
The tumor microenvironment plays a critical role in maintaining the immature phenotype of tumor-initiating cells (TIC) to promote cancer. Hepatocellular carcinoma (HCC) is a unique disease in that it develops in the setting of fibrosis and cirrhosis. This pathologic state commonly shows an enrichment of stromal myofibroblasts, which constitute the bulk of the tumor microenvironment and contribute to disease progression. Follistatin-like 1 (FSTL1) has been widely reported as a proinflammatory mediator in different fibrosis-related and inflammatory diseases. Here we show FSTL1 expression to be closely correlated with activated fibroblasts and to be elevated in regenerative, fibrotic, and disease liver states in various mouse models. Consistently, FSTL1 lineage cells gave rise to myofibroblasts in a CCL4-induced hepatic fibrosis mouse model. Clinically, high FSTL1 in fibroblast activation protein-positive (FAP+) fibroblasts were significantly correlated with more advanced tumors in patients with HCC. Although FSTL1 was expressed in primary fibroblasts derived from patients with HCC, it was barely detectable in HCC cell lines. Functional investigations revealed that treatment of HCC cells and patient-derived 3D organoids with recombinant FSTL1 or with conditioned medium collected from hepatic stellate cells or from cells overexpressing FSTL1 could promote HCC growth and metastasis. FSTL1 bound to TLR4 receptor, resulting in activation of AKT/mTOR/4EBP1 signaling. In a preclinical mouse model, blockade of FSTL1 mitigated HCC malignancy and metastasis, sensitized HCC tumors to sorafenib, prolonged survival, and eradicated the TIC subset. Collectively, these data suggest that FSTL1 may serve as an important novel diagnostic/prognostic biomarker and therapeutic target in HCC. SIGNIFICANCE: This study shows that FSTL1 secreted by activated fibroblasts in the liver microenvironment augments hepatocellular carcinoma malignancy, providing a potential new strategy to improve treatment of this aggressive disease.
Collapse
Affiliation(s)
- Jia-Jian Loh
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Tsz-Wai Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lei Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Tin-Lok Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Xue Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Victor W S Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chung-Mau Lo
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,The University of Hong Kong-Shenzhen Hospital, Pok Fu Lam, Hong Kong
| | - Kwan Man
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,The University of Hong Kong-Shenzhen Hospital, Pok Fu Lam, Hong Kong
| | - Terence K Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Wen Ning
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Man Tong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong. .,State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong.,The University of Hong Kong-Shenzhen Hospital, Pok Fu Lam, Hong Kong
| |
Collapse
|
47
|
Inhibition of FSTL3 abates the proliferation and metastasis of renal cell carcinoma via the GSK-3β/β-catenin signaling pathway. Aging (Albany NY) 2021; 13:22528-22543. [PMID: 34555811 PMCID: PMC8507290 DOI: 10.18632/aging.203564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is a lethal malignancy of the genitourinary system. Follistatin-like 3 (FSTL3), which mediates cell differentiation and growth, acts as a biomarker of tumors and participates in cancer development and progression. Presently, the FSTL3’s functions in RCC were investigated. Quantitative reverse transcription PCR (qRT-PCR), Western Blot, and enzyme linked immunosorbent assay (ELISA) were conducted to verify FSTL3 expression in RCC tissues and cell lines. BrdU assay and CCK8 experiment were made to monitor cell proliferation. Transwell was implemented to examine the invasion of the cells. Flow cytometry analyzed cell apoptosis, and Western Blot evaluated the protein levels of E-cadherin, Twist, and Slug. In the meantime, the protein profiles of the GSK-3β, β-catenin, and TGF-β signaling pathways were ascertained. Moreover, the Xenograft tumor model was constructed in nude mice for measuring tumor growth in vivo. The statistics showed that FSTL3 presented an overexpression in RCC, and patients with a lower expression of FSTL3 manifested a better prognosis. Down-regulated FSTL3 hampered the proliferation, invasion, EMT, and tumor growth of RCC cells and caused cell apoptosis. On the contrary, FSTL3 overexpression enhanced the malignant behaviors of RCC cells. Furthermore, FSTL3 knockdown bolstered GSK-3β, suppressed β-catenin, and reduced BMP1-SMAD pathway activation. Inhibited β-catenin substantially mitigated FSTL3-mediated promoting functions in RCC. In short, FSTL3 functions as an oncogene in RCC by modulating the GSK-3β/β-catenin signaling pathway.
Collapse
|
48
|
Viera M, Yip GWC, Shen HM, Baeg GH, Bay BH. Targeting CD82/KAI1 for Precision Therapeutics in Surmounting Metastatic Potential in Breast Cancer. Cancers (Basel) 2021; 13:4486. [PMID: 34503296 PMCID: PMC8431267 DOI: 10.3390/cancers13174486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis is the main cause of mortality in breast cancer patients. There is an unmet need to develop therapies that can impede metastatic spread. Precision oncology has shown great promise for the treatment of cancers, as the therapeutic approach is tailored to a specific group of patients who are likely to benefit from the treatment, rather than the traditional approach of "one size fits all". CD82, also known as KAI1, a glycoprotein belonging to the tetraspanin family and an established metastasis suppressor, could potentially be exploited to hinder metastases in breast cancer. This review explores the prospect of targeting CD82 as an innovative therapeutic approach in precision medicine for breast cancer patients, with the goal of preventing cancer progression and metastasis. Such an approach would entail the selection of a subset of breast cancer patients with low levels of CD82, and instituting an appropriate treatment scheme tailored towards restoring the levels of CD82 in this group of patients. Proposed precision treatment regimens include current modalities of treating breast cancer, in combination with either clinically approved drugs that could restore the levels of CD82, CD82 peptide mimics or non-coding RNA-based therapeutics.
Collapse
Affiliation(s)
- Maximillian Viera
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - George Wai Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Gyeong Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (M.V.); (G.W.C.Y.)
| |
Collapse
|
49
|
Shi J, Wang S, He Q, Liu K, Zhao W, Xie Q, Cheng L. TNF-α induces up-regulation of MicroRNA-27a via the P38 signalling pathway, which inhibits intervertebral disc degeneration by targeting FSTL1. J Cell Mol Med 2021; 25:7146-7156. [PMID: 34190406 PMCID: PMC8335690 DOI: 10.1111/jcmm.16745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/16/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
The mechanism of intervertebral disc degeneration is still unclear, and there are no effective therapeutic strategies for treating this condition. miRNAs are naturally occurring macromolecules in the human body and have many biological functions. Therefore, we hope to elucidate whether miRNAs are associated with intervertebral disc degeneration and the underlying mechanisms involved. In our study, differentially expressed miRNAs were predicted by the GEO database and then confirmed by qPCR and in situ hybridization. Apoptosis of nucleus pulposus cells was detected by flow cytometry and Bcl2, Bax and caspase 3. Deposition of extracellular matrix was assessed by Alcian blue staining, and the expression of COX2 and MMP13 was detected by immunofluorescence, Western blot and qPCR. Moreover, qPCR was used to detect the expression of miR27a and its precursors. The results showed that miR27a was rarely expressed in healthy intervertebral discs but showed increased expression in degenerated intervertebral discs. Ectopic miR27a expression inhibited apoptosis, suppressed the inflammatory response and attenuated the catabolism of the extracellular matrix by targeting FSTL1. Furthermore, it seems that the expression of miR27a was up-regulated by TNF-α via the P38 signalling pathway. So we conclude that TNF-α and FSTL1 engage in a positive feedback loop to promote intervertebral disc degeneration. At the same time, miR27a is up-regulated by TNF-α via the P38 signalling pathway, which ameliorates inflammation, apoptosis and matrix degradation by targeting FSTL1. Thus, this negative feedback mechanism might contribute to the maintenance of a low degeneration load and would be beneficial to maintain a persistent chronic disc degeneration.
Collapse
Affiliation(s)
- Jie Shi
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
- Cheeloo College of MedicineShandong UniversityJinanChina
- NHC key Laboratory of OtorhinolaryngologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Shaoyi Wang
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
- Cheeloo College of MedicineShandong UniversityJinanChina
| | - Qiting He
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
- Cheeloo College of MedicineShandong UniversityJinanChina
- NHC key Laboratory of OtorhinolaryngologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Kaiwen Liu
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
- Cheeloo College of MedicineShandong UniversityJinanChina
| | - Wei Zhao
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
- Cheeloo College of MedicineShandong UniversityJinanChina
| | - Qing Xie
- Department of PharmacyQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
| | - Lei Cheng
- Department of OrthopaedicQilu HospitalCheeloo College of Medicine of Shandong UniversityJinanChina
| |
Collapse
|
50
|
Wu HJ, Chu PY. Epigenetic Regulation of Breast Cancer Stem Cells Contributing to Carcinogenesis and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22158113. [PMID: 34360879 PMCID: PMC8348144 DOI: 10.3390/ijms22158113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Globally, breast cancer has remained the most commonly diagnosed cancer and the leading cause of cancer death among women. Breast cancer is a highly heterogeneous and phenotypically diverse group of diseases, which require different selection of treatments. Breast cancer stem cells (BCSCs), a small subset of cancer cells with stem cell-like properties, play essential roles in breast cancer progression, recurrence, metastasis, chemoresistance and treatments. Epigenetics is defined as inheritable changes in gene expression without alteration in DNA sequence. Epigenetic regulation includes DNA methylation and demethylation, as well as histone modifications. Aberrant epigenetic regulation results in carcinogenesis. In this review, the mechanism of epigenetic regulation involved in carcinogenesis, therapeutic resistance and metastasis of BCSCs will be discussed, and finally, the therapies targeting these biomarkers will be presented.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975611855; Fax: +886-47227116
| |
Collapse
|