1
|
Isogai T, Hirosawa KM, Kanno M, Sho A, Kasai RS, Komura N, Ando H, Furukawa K, Ohmi Y, Furukawa K, Yokota Y, Suzuki KG. Extracellular vesicles adhere to cells primarily by interactions of integrins and GM1 with laminin. J Cell Biol 2025; 224:e202404064. [PMID: 40304687 PMCID: PMC12042775 DOI: 10.1083/jcb.202404064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-derived extracellular vesicles (EVs) have attracted significant attention, yet the molecular mechanisms that govern their specific binding to recipient cells remain elusive. Our in vitro study utilizing single-particle tracking demonstrated that integrin heterodimers comprising α6β4 and α6β1 and ganglioside, GM1, are responsible for the binding of small EV (sEV) subtypes to laminin. EVs derived from four distinct tumor cell lines, regardless of size, exhibited high binding affinities for laminin but not for fibronectin, although fibronectin receptors are abundant in EVs and have functional roles in EV-secreting cells. Our findings revealed that integrins in EVs bind to laminin via the conventional molecular interface, facilitated by CD151 rather than by inside-out signaling of talin-1 and kindlin-2. Super-resolution movie observation revealed that sEV integrins bind only to laminin on living recipient cells. Furthermore, sEVs bound to HUVEC and induced cell branching morphogenesis in a laminin-dependent manner. Thus, we demonstrated that EVs predominantly bind to laminin on recipient cells, which is indispensable for cell responses.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | | | - Miki Kanno
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Ayano Sho
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Rinshi S. Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hiromune Ando
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yasunari Yokota
- Department of Information Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Kenichi G.N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
2
|
Takai Y, Naito S, Ito H, Horie S, Ushijima M, Narisawa T, Yagi M, Ichiyanagi O, Tsuchiya N. Ankrd1 Promotes Lamellipodia Formation and Cell Motility via Interaction with Talin-1 in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2025; 26:4232. [PMID: 40362467 PMCID: PMC12072362 DOI: 10.3390/ijms26094232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Ankyrin repeat domain 1 (Ankrd1), a transcriptional target of Yes-associated protein (YAP), is linked to cardiomyopathy. However, its role in cancer, particularly in clear cell renal cell carcinoma (ccRCC), remains vague. In this study, we examined the expression, regulation, and function of Ankrd1 in ccRCC. High Ankrd1 expression was related to poor prognosis in patients with ccRCC in The Cancer Genome Atlas cohort. Ankrd1 expression was regulated by YAP in all ccRCC cell lines examined and also by ERK5 in a subset of ccRCC cell lines. Moreover, silencing of Ankrd1 in ccRCC cell lines resulted in decreased cell motility, whereas its overexpression increased the cell motility. Ankrd1 colocalized with F-actin in lamellipodia upon phorbol ester stimulation. Ankrd1 silencing resulted in alterations in the shape of RCC cells and caused a decrease in lamellipodia formation. Ankrd1 also colocalized with talin-1 in lamellipodia. Ankrd1 depletion repressed talin-1-mediated activation of the integrin pathway. Immunohistochemical examination of surgical specimens revealed high expression of Ankrd1 in metastatic RCC tissues compared with that in primary RCC tissues from the same patients. Collectively, these findings suggest that Ankrd1 plays a critical role in the motility of ccRCC cells through lamellipodia formation.
Collapse
Affiliation(s)
- Yuki Takai
- Department of Urology, Faculty of Medicine, Yamagata University, 2-2-2 Iida-nishi, Yamagata 990-9585, Japan; (S.N.); (H.I.); (S.H.); (M.U.); (T.N.); (M.Y.); (O.I.); (N.T.)
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Guo X, Li S. Bone metastases of prostate cancer: Molecular mechanisms, targeted diagnosis and targeted therapy (Review). Oncol Rep 2025; 53:46. [PMID: 39981932 PMCID: PMC11865881 DOI: 10.3892/or.2025.8879] [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: 05/18/2024] [Accepted: 09/12/2024] [Indexed: 02/22/2025] Open
Abstract
Prostate cancer (PCa) is second only to lung cancer in terms of death among men worldwide. Advanced PCa frequently results in bone metastases, which occur in ~90% of patients and frequently result in severe skeleton‑related events. Currently, the treatment for this disease is limited to alleviating its clinical symptoms and cannot provide a complete cure. Therefore, the development of novel treatment strategies is particularly important. In recent years, numerous novel strategies for the diagnosis and treatment of PCa have emerged, resulting in good clinical efficacy. For example, strategies targeting prostate specific membrane antigen, poly ADP‑ribose polymerase and programmed cell death protein 1 have been applied to PCa‑induced bone metastasis, and have shown initial efficacy and great potential. Therefore, understanding the molecular mechanisms underlying the formation of bone metastases in patients with PCa is of importance for the effective management of this disease. The purpose of the present review is to comprehensively outline the roles of protein‑coding genes and non‑coding RNAs in the development of bone metastases of PCa to elucidate their significance in the management of PCa. The aim is to offer clinicians and researchers a comprehensive understanding of this topic.
Collapse
Affiliation(s)
- Xutang Guo
- Department of Urology, Gansu Province Maternity and Child Health Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Shaojun Li
- Department of Urology, Gansu Province Maternity and Child Health Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
4
|
He Z, Sun J, Wang M, Chen S, Mao G, Yang L. Talin1 Ser425 phosphorylation promotes colorectal cancer progression and metastasis. Transl Cancer Res 2025; 14:796-807. [PMID: 40104703 PMCID: PMC11912083 DOI: 10.21037/tcr-24-1283] [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: 07/24/2024] [Accepted: 12/04/2024] [Indexed: 03/20/2025]
Abstract
Background Talin1 serves as a crucial element within the multiprotein adhesion complexes that facilitate processes such as cell migration, adhesion, and integrin signaling. This study aimed to explore the underlying role of Talin1 Ser425 phosphorylation in the development of colorectal cancer (CRC). Methods Blank plasmids, non-phosphorylatable mutant Talin1 S425A plasmids, and phosphorylation-mimetic mutant Talin1 S425D plasmids were constructed and used for transfection of CRC cells. The expression of mRNA and protein in CRC cells or tumor tissues was assessed by The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and UALCAN databases, immunohistochemistry (IHC), and Western blot (WB). Cell proliferation was assessed via 5-ethynyl-2-deoxyuridine (EDU) proliferation assay and colony formation assay. Cell migration and invasion were detected by wound healing assay and transwell assay. Cell apoptosis was assessed by flow cytometry. The Kaplan-Meier Plotter was used to evaluate the prognostic value of mRNA in CRC. Results TLN1 was markedly downregulated in CRC tissues while the level of Talin1 Ser425 phosphorylation in CRC tissues and aggressive CRC cells was relatively higher. The S425A mutant inhibited CRC cell proliferation, migration, and invasion, whereas the S425D mutant promoted these processes. Flow cytometry assay showed that cell apoptosis was induced by S425A mutant and suppressed by S425D mutant in RKO cells. Further investigation suggested that CDK5 might be responsible for Talin1 phosphorylation. Conclusions Talin1 Ser425 phosphorylation is of great importance in CRC development and Talin1 is supposed to be a potential tumor marker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhengxiu He
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Gastroenterology, Dongtai People’s Hospital, Yancheng, China
| | - Jian Sun
- Department of Respiratory Medicine, Shanghai Jiading District Anting Hospital, Shanghai, China
| | - Mengmeng Wang
- Department of Chinese Medicine Oncology, Shanghai Jiading District Anting Hospital, Shanghai, China
| | - Shanshan Chen
- Cancer Research Center, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Guoxin Mao
- Department of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Li Yang
- Department of Respiratory Medicine, Shanghai Jiading District Anting Hospital, Shanghai, China
| |
Collapse
|
5
|
Baster Z, Russell L, Rajfur Z. A Review of Talin- and Integrin-Dependent Molecular Mechanisms in Cancer Invasion and Metastasis. Int J Mol Sci 2025; 26:1798. [PMID: 40076426 PMCID: PMC11899650 DOI: 10.3390/ijms26051798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer is the second most common cause of death in the world, representing one of the main economic burdens in health care and research. The effort of research has mainly focused on limiting the growth of a localized tumor, but most recently, there has been more attention focused on restricting the spreading of the cancer via invasion and metastasis. The signaling pathways behind these two processes share many molecules with physiological pathways regulating cell adhesion and migration, and, moreover, adhesion and migration processes themselves underlie tumor potential for invasion. In this work, we reviewed the latest literature about cancer development and invasion and their regulation by cell migration- and adhesion-related proteins, with a specific focus on talins and integrins. We also summarized the most recent developments and approaches to anti-cancer therapies, concentrating on cell migration-related therapies.
Collapse
Affiliation(s)
- Zbigniew Baster
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Laboratory for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lindsay Russell
- Undergraduate Program, Barnard College of Columbia University, New York, NY 10027, USA;
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, 30-348 Kraków, Poland
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348 Kraków, Poland
| |
Collapse
|
6
|
Hashemi F, Tajik F, Saeednejad Zanjani L, Dehghan Manshadi M, Safaei S, Babaheidarian P, Fattahi F, Ghods R, Madjd Z. Clinical significance of Talin-1 and HER-2 status in different types of gastric carcinoma. Biomarkers 2024; 29:539-556. [PMID: 39466840 DOI: 10.1080/1354750x.2024.2423270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Talin-1 (TLN1) is crucial in cell migration, metastasis, and cancer development. This study evaluated Talin-1 expression and its clinical significance in gastric cancer (GC), along with human epidermal growth factor receptor-2 (HER-2) expression and its correlation with Talin-1. METHODS Bioinformatics analysis assessed the potential prognostic value of Talin-1 and HER-2 in GC patients. The study included 223 GC patients (Signet Ring Cells and Intestinal subtypes) and 29 non-malignant tissue samples. Immunohistochemistry (IHC) on tissue microarray slides evaluated Talin-1 and HER-2 expression and clinical significance. Receiver operating characteristic (ROC) curves assessed their diagnostic value. RESULTS Bioinformatics identified Talin-1 as a potential prognostic factor and HER-2 as an oncogene in GC. Talin-1 and HER-2 expression increased in SRC-type GC samples compared to non-malignant tissues. High cytoplasmic Talin-1 expression inversely correlated with tumor expansion and invasion in SRC-type GC. Increased HER-2 expression positively correlated with metastasis. ROC curves showed significant diagnostic values for both proteins. CONCLUSIONS Higher cytoplasmic Talin-1 expression is associated with less invasive tumor behavior, while increased membranous HER-2 expression is associated with metastasis in SRC-type GC. These findings suggest potential use in assessing diagnosis and screening high-risk cancer patients, particularly those with SRC-type GC.
Collapse
Affiliation(s)
- Farideh Hashemi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Masoumeh Dehghan Manshadi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran Iran
| | - Sadegh Safaei
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran Iran
| | | | - Fahimeh Fattahi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran Iran
- Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Li S, Chen A, Gui J, Zhou H, Zhu L, Mi Y. TLN1: an oncogene associated with tumorigenesis and progression. Discov Oncol 2024; 15:716. [PMID: 39589610 PMCID: PMC11599537 DOI: 10.1007/s12672-024-01593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Talin-1 (TLN1), encoded by the TLN1 gene, is a focal adhesion-related protein capable of binding various proteins in the cytoskeleton. It is also expressed at high levels in many cancers wherein it influences cellular adhesion and the activation of integrins. TLN1 is also capable of promoting tumor cell invasivity, proliferation, and metastatic progression, in addition to being a relevant biomarker and therapeutic target in certain cancers. The present review offers a comprehensive overview of current knowledge regarding TLN1 with respect to its structural properties, functions, and role in tumor development.
Collapse
Affiliation(s)
- Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
8
|
Zhang Y, Shi X, Shi M, Li J, Liu Q. Androgens and androgen receptor directly induce the thickening, folding, and vascularization of the seahorse abdominal dermal layer into a placenta-like structure responsible for male pregnancy via multiple signaling pathways. Int J Biol Macromol 2024; 279:135039. [PMID: 39197609 DOI: 10.1016/j.ijbiomac.2024.135039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Seahorses exhibit the unique characteristic of male pregnancy, which incubates numerous embryos in a brood pouch that plays an essential role in enhancing offspring survivability. The pot-belly seahorse (Hippocampus abdominalis) possesses the largest body size among seahorses and is a significant species in Chinese aquaculture. In this study, we revealed the cytological and morphological characteristics, as well as regulatory mechanisms, throughout the entire brood pouch development in H. abdominalis. The brood pouch originated from the abdominal dermis, extending towards the ventral midline. As the dermal layers thicken, the inner epithelium folds, the stroma loosens, and vascularization occurs, culminating in the formation of the brood pouch. Furthermore, through transcriptomic analysis of brood pouches at various developmental stages, 8 key genes (tgfb3, fgf2, wnt7a, pgf, mycn, tln2, jund, ccn4) closely related to the development of brood pouch were identified in the MAPK, Rap1, TGF-β, and Wnt signaling pathways. These genes were highly expressed in the pseudoplacenta and dermal layers at the newly formed stage as examined by in situ hybridization (ISH). The angiogenesis, densification of collagen fibers, and proliferation of fibroblasts and endothelial cells in seahorse brood pouch formation may be regulated by these genes and pathways. Additionally, the expression of the androgen receptor gene (ar) was significantly upregulated during the formation of the brood pouch, and ISH confirmed the expression of the ar gene in the dermis and pseudoplacenta of the brood pouch, highlighting its role in the developmental process. Androgen and flutamide (androgen receptor antagonist) treatments significantly accelerated the formation of the brood pouch and completely inhibited its occurrence respectively, concomitant to the upregulated expression of differentially expressed genes involved above signaling pathways. These findings demonstrated that formation of the brood pouch is determined by androgen and the androgen receptor activates the above signaling pathways in the brood pouch through the regulation of fgf2, tgfb3, pgf, and wnt7a. Interestingly, androgen even induced the formation of the brood pouch in females. We firstly elucidated the formation of the seahorse brood pouch, demonstrating that androgens and their receptors directly induce the thickening, folding, and vascularization of the abdominal dermal layer into a placenta-like structure through multiple signaling pathways. These findings provide foundational insights to further exploring the evolution of male pregnancy and adaptive convergence in viviparity across vertebrates.
Collapse
Affiliation(s)
- Yichao Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266000, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Xuehui Shi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Meilun Shi
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266000, China
| | - Jun Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
| | - Qinghua Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
| |
Collapse
|
9
|
Xu Y, Zhang G, Liu Y, Liu Y, Tian A, Che J, Zhang Z. Molecular mechanisms and targeted therapy for the metastasis of prostate cancer to the bones (Review). Int J Oncol 2024; 65:104. [PMID: 39301646 PMCID: PMC11419411 DOI: 10.3892/ijo.2024.5692] [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: 05/08/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024] Open
Abstract
The incidence of prostate cancer (PCa) is increasing, making it one of the prevalent malignancies among men. Metastasis of PCa to the bones poses the greatest danger to patients, potentially resulting in treatment ineffectiveness and mortality. At present, the management of patients with bone metastasis focuses primarily on providing palliative care. Research has indicated that the spread of PCa to the bones occurs through the participation of numerous molecules and their respective pathways. Gaining knowledge regarding the molecular processes involved in bone metastasis may result in the development of innovative and well‑tolerated therapies, ultimately enhancing the quality of life and prognosis of patients. The present article provides the latest overview of the molecular mechanisms involved in the formation of bone metastatic tumors from PCa. Additionally, the clinical outcomes of targeted drug therapies for bone metastasis are thoroughly analyzed. Finally, the benefits and difficulties of targeted therapy for bone metastasis of PCa are discussed, aiming to offer fresh perspectives for treatment.
Collapse
Affiliation(s)
- Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Gang Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Jizhong Che
- Correspondence to: Professor Zhengchao Zhang or Professor Jizhong Che, Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, 717, Jinbu Street, Muping, Yantai, Shandong 264100, P.R. China, E-mail: , E-mail:
| | - Zhengchao Zhang
- Correspondence to: Professor Zhengchao Zhang or Professor Jizhong Che, Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, 717, Jinbu Street, Muping, Yantai, Shandong 264100, P.R. China, E-mail: , E-mail:
| |
Collapse
|
10
|
Liang S, Zhou S, Tang Y, Xiao M, Ye K. CREB1 promotes cholangiocarcinoma metastasis through transcriptional regulation of the LAYN-mediated TLN1/β1 integrin axis: CREB1 promotes cholangiocarcinoma metastasis through regulating LAYN/TLN1/β1 integrin axis. Heliyon 2024; 10:e36595. [PMID: 39286102 PMCID: PMC11402943 DOI: 10.1016/j.heliyon.2024.e36595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Background Layilin (LAYN) plays an important role in tumor progression, invasion, and metastasis; however, its role in cholangiocarcinoma (CHOL) has not been elucidated. Methods We utilized the GEPIA, STRING, and hTFtarget databases for bioinformatics analysis. Overexpression or knockdown cell lines were constructed by transfecting the cells with different plasmids. Western blot (WB) was performed to detect LAYN, TLN1, and CREB1 expression. Cell proliferation, migration, and invasiveness were assessed using CCK-8 and Transwell assays. Immunofluorescence and WB were used to detect epithelial-mesenchymal transition (EMT) markers. The CHOL metastasis model was established by injecting RBE cells into the tail veins of nude mice. Metastatic lesions were identified using hematoxylin and eosin staining. Co-immunoprecipitation and Chromatin immunoprecipitation were used to validate the interactions. Results LAYN was highly expressed in the CHOL cells. Knockdown of LAYN significantly inhibited proliferation, migration, invasion, and EMT in both QBC-939 and RBE human CHOL cells, while overexpression of LAYN had the opposite effect. Furthermore, in a CHOL metastasis model using nude mice, knocking down LAYN expression markedly suppressed CHOL liver and lung metastases. LAYN interacts with TLN1, and CREB1 binds to the LAYN promoter, with all three showing a positive correlation. Additionally, bioinformatics analysis revealed high expression of both TLN1 and CREB1 in CHOL. Knockdown of TLN1 or CREB1 in QBC-939 and RBE cells inhibited cell proliferation, migration, invasion, and EMT, reversing the effects of LAYN overexpression. Moreover, knockdown of TLN1 or CREB1 also suppressed the expression of ITGB1 and the phosphorylation levels of c-Jun, p38 MAPK, and ERK, further reversing the effects of LAYN overexpression. Conclusion Our results suggest that CREB1 promotes CHOL metastasis through transcriptional regulation of the LAYN-mediated TLN1/β1 integrin axis.
Collapse
Affiliation(s)
- Shuai Liang
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuhua Zhou
- Comprehensive Surgery, Xiangya Boai Rehabilitation Hospital, Changsha, 410100, China
| | - Yangshuo Tang
- Department of Ultrasound Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Moyan Xiao
- Department of Hepatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ke Ye
- Department of Hepatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
11
|
Liu J, Lu Y, Zheng B, Huang D, Song J, Wang B, Zheng S. Talin1 promotes HCC progression by regulating NRG1/PI3K/AKT pathway. Discov Oncol 2024; 15:360. [PMID: 39162903 PMCID: PMC11335986 DOI: 10.1007/s12672-024-01243-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 08/14/2024] [Indexed: 08/21/2024] Open
Abstract
OBJECTIVE OF THE STUDY Hepatocellular carcinoma (HCC) stands as the third leading cause of cancer-related mortality globally. Metastasis, responsible for treatment failures, underscores the urgency to comprehend molecular drivers of invasion and migration. Central to the invasive and migratory processes underlying metastasis is the protein Talin1. However, the role and underlying mechanisms governing Talin1's involvement in HCC have remained elusive. METHODS A total of 100 HCC specimens were collected from patients who underwent hepatectomy in our center. The expression level of talin1 was measured to evaluate the correlationship of talin1 and the development of HCC. In vitro and in vivo experiments were conducted to verify the characteristic of talin1 in HCC. RNA-seq and bioinformatics analysis were performed to identify the downstream signal pathway of talin1 and their impact on HCC development. RESULTS Here, we reported elevated levels of Talin1 mRNA and protein in HCC tissues. Meanwhile, downregulation of Talin1 significantly reduced the HCC cell proliferation and metastasis in vitro and in vivo. Furthermore, elevating NRG-1, a downstream target of Talin1, enhanced metastasis of HCC cells. More importantly, attenuation of Talin1 inhibited HCC progression through decreasing the stabilization of NRG1 mRNA, consequently regulating the expression of NRG1 and its involvement in mediating the PI3K/AKT pathway. CONCLUSION Taken together, Talin1 regulates cellular proliferation, metastasis, and invasiveness by modulating NRG1/PI3K/AKT axis, suggesting that Talin1 emerges as a promising candidate for treating HCC.
Collapse
Affiliation(s)
- Jialong Liu
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yao Lu
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Bowen Zheng
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Deng Huang
- Department of Hepatobiliary, General Hospital of Tibet Military Command Area, Lhasa, Tibet, China
| | - Juxian Song
- Department of Hepatobiliary Surgery, The 925th Hospital of the PLA, Guiyang, Guizhou, China
| | - Baolin Wang
- Department of Surgery, The 63650th Troop Hospital of the Chinese People's Liberation Army, Korla, Xinjiang, China
| | - Shuguo Zheng
- Insititute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
12
|
Samaržija I, Lukiyanchuk V, Lončarić M, Rac-Justament A, Stojanović N, Gorodetska I, Kahya U, Humphries JD, Fatima M, Humphries MJ, Fröbe A, Dubrovska A, Ambriović-Ristov A. The extracellular matrix component perlecan/HSPG2 regulates radioresistance in prostate cancer cells. Front Cell Dev Biol 2024; 12:1452463. [PMID: 39149513 PMCID: PMC11325029 DOI: 10.3389/fcell.2024.1452463] [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: 06/20/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024] Open
Abstract
Radiotherapy of prostate cancer (PC) can lead to the acquisition of radioresistance through molecular mechanisms that involve, in part, cell adhesion-mediated signaling. To define these mechanisms, we employed a DU145 PC model to conduct a comparative mass spectrometry-based proteomic analysis of the purified integrin nexus, i.e., the cell-matrix junction where integrins bridge assembled extracellular matrix (matrisome components) to adhesion signaling complexes (adhesome components). When parental and radioresistant cells were compared, the expression of integrins was not changed, but cell radioresistance was associated with extensive matrix remodeling and changes in the complement of adhesion signaling proteins. Out of 72 proteins differentially expressed in the parental and radioresistant cells, four proteins were selected for functional validation based on their correlation with biochemical recurrence-free survival. Perlecan/heparan sulfate proteoglycan 2 (HSPG2) and lysyl-like oxidase-like 2 (LOXL2) were upregulated, while sushi repeat-containing protein X-linked (SRPX) and laminin subunit beta 3 (LAMB3) were downregulated in radioresistant DU145 cells. Knockdown of perlecan/HSPG2 sensitized radioresistant DU145 RR cells to irradiation while the sensitivity of DU145 parental cells did not change, indicating a potential role for perlecan/HSPG2 and its associated proteins in suppressing tumor radioresistance. Validation in androgen-sensitive parental and radioresistant LNCaP cells further supported perlecan/HSPG2 as a regulator of cell radiosensitivity. These findings extend our understanding of the interplay between extracellular matrix remodeling and PC radioresistance and signpost perlecan/HSPG2 as a potential therapeutic target and biomarker for PC.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Vasyl Lukiyanchuk
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Marija Lončarić
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Anja Rac-Justament
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Nikolina Stojanović
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ielizaveta Gorodetska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Uğur Kahya
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jonathan D Humphries
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mahak Fatima
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Ana Fröbe
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Anna Dubrovska
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium, Partner Site Dresden and German Cancer Research Center, Heidelberg, Germany
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
13
|
Pan T, Liu F, Hao X, Wang S, Wasi M, Song JH, Lewis VO, Lin PP, Moon B, Bird JE, Panaretakis T, Lin SH, Wu D, Farach-Carson MC, Wang L, Zhang N, An Z, Zhang XHF, Satcher RL. BIGH3 mediates apoptosis and gap junction failure in osteocytes during renal cell carcinoma bone metastasis progression. Cancer Lett 2024; 596:217009. [PMID: 38849015 PMCID: PMC11964150 DOI: 10.1016/j.canlet.2024.217009] [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: 03/17/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
Renal cell carcinoma (RCC) bone metastatis progression is driven by crosstalk between tumor cells and the bone microenvironment, which includes osteoblasts, osteoclasts, and osteocytes. RCC bone metastases (RCCBM) are predominantly osteolytic and resistant to antiresorptive therapy. The molecular mechanisms underlying pathologic osteolysis and disruption of bone homeostasis remain incompletely understood. We previously reported that BIGH3/TGFBI (transforming growth factor-beta-induced protein ig-h3, shortened to BIGH3 henceforth) secreted by colonizing RCC cells drives osteolysis by inhibiting osteoblast differentiation, impairing healing of osteolytic lesions, which is reversible with osteoanabolic agents. Here, we report that BIGH3 induces osteocyte apoptosis in both human RCCBM tissue specimens and in a preclinical mouse model. We also demonstrate that BIGH3 reduces Cx43 expression, blocking gap junction (GJ) function and osteocyte network communication. BIGH3-mediated GJ inhibition is blocked by the lysosomal inhibitor hydroxychloroquine (HCQ), but not osteoanabolic agents. Our results broaden the understanding of pathologic osteolysis in RCCBM and indicate that targeting the BIGH3 mechanism could be a combinational strategy for the treatment of RCCBM-induced bone disease that overcomes the limited efficacy of antiresorptives that target osteoclasts.
Collapse
Affiliation(s)
- Tianhong Pan
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fengshuo Liu
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoxin Hao
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Shubo Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Jian H Song
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valerae O Lewis
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick P Lin
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Moon
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justin E Bird
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Theocharis Panaretakis
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sue-Hwa Lin
- Departments of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Departments of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA; Departments of Bioengineering, Rice University, Houston, TX, USA
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA; Departments of BioSciences, Rice University, Houston, TX, USA; Departments of Bioengineering, Rice University, Houston, TX, USA
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Ningyan Zhang
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, USA
| | - Zhiqiang An
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, USA
| | - Xiang H-F Zhang
- Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Departments of Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA; Departments of Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA; Departments of McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Robert L Satcher
- Departments of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
15
|
Wang Y, Huang H, Weng H, Jia C, Liao B, Long Y, Yu F, Nie Y. Talin mechanotransduction in disease. Int J Biochem Cell Biol 2024; 166:106490. [PMID: 37914021 DOI: 10.1016/j.biocel.2023.106490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
Talin protein (Talin 1/2) is a mechanosensitive cytoskeleton protein. The unique structure of the Talin plays a vital role in transmitting mechanical forces. Talin proteins connect the extracellular matrix to the cytoskeleton by linking to integrins and actin, thereby mediating the conversion of mechanical signals into biochemical signals and influencing disease progression as potential diagnostic indicators, therapeutic targets, and prognostic indicators of various diseases. Most studies in recent years have confirmed that mechanical forces also have a crucial role in the development of disease, and Talin has been found to play a role in several diseases. Still, more studies need to be done on how Talin is involved in mechanical signaling in disease. This review focuses on the mechanical signaling of Talin in disease, aiming to summarize the mechanisms by which Talin plays a role in disease and to provide references for further studies.
Collapse
Affiliation(s)
- Yingzi Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Haozhong Huang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Huimin Weng
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Chunsen Jia
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Sichuan Clinical Research Center for Nephropathy, Luzhou, China
| | - Fengxu Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China
| | - Yongmei Nie
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, China; Key Laboratory of Cardiovascular Remodeling and Dysfunction, Luzhou, China.
| |
Collapse
|
16
|
Samaržija I. The Potential of Extracellular Matrix- and Integrin Adhesion Complex-Related Molecules for Prostate Cancer Biomarker Discovery. Biomedicines 2023; 12:79. [PMID: 38255186 PMCID: PMC10813710 DOI: 10.3390/biomedicines12010079] [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: 11/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Prostate cancer is among the top five cancer types according to incidence and mortality. One of the main obstacles in prostate cancer management is the inability to foresee its course, which ranges from slow growth throughout years that requires minimum or no intervention to highly aggressive disease that spreads quickly and resists treatment. Therefore, it is not surprising that numerous studies have attempted to find biomarkers of prostate cancer occurrence, risk stratification, therapy response, and patient outcome. However, only a few prostate cancer biomarkers are used in clinics, which shows how difficult it is to find a novel biomarker. Cell adhesion to the extracellular matrix (ECM) through integrins is among the essential processes that govern its fate. Upon activation and ligation, integrins form multi-protein intracellular structures called integrin adhesion complexes (IACs). In this review article, the focus is put on the biomarker potential of the ECM- and IAC-related molecules stemming from both body fluids and prostate cancer tissue. The processes that they are involved in, such as tumor stiffening, bone turnover, and communication via exosomes, and their biomarker potential are also reviewed.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
17
|
Nikhil K, Shah K. CDK5: an oncogene or an anti-oncogene: location location location. Mol Cancer 2023; 22:186. [PMID: 37993880 PMCID: PMC10666462 DOI: 10.1186/s12943-023-01895-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023] Open
Abstract
Recent studies have uncovered various physiological functions of CDK5 in many nonneuronal tissues. Upregulation of CDK5 and/or its activator p35 in neurons promotes healthy neuronal functions, but their overexpression in nonneuronal tissues is causally linked to cancer of many origins. This review focuses on the molecular mechanisms by which CDK5 recruits diverse tissue-specific substrates to elicit distinct phenotypes in sixteen different human cancers. The emerging theme suggests that CDK5's role as an oncogene or anti-oncogene depends upon its subcellular localization. CDK5 mostly acts as an oncogene, but in gastric cancer, it is a tumor suppressor due to its unique nuclear localization. This indicates that CDK5's access to certain nuclear substrates converts it into an anti-oncogenic kinase. While acting as a bonafide oncogene, CDK5 also activates a few cancer-suppressive pathways in some cancers, presumably due to the mislocalization of nuclear substrates in the cytoplasm. Therefore, directing CDK5 to the nucleus or exporting tumor-suppressive nuclear substrates to the cytoplasm may be promising approaches to combat CDK5-induced oncogenicity, analogous to neurotoxicity triggered by nuclear CDK5. Furthermore, while p35 overexpression is oncogenic, hyperactivation of CDK5 by inducing p25 formation results in apoptosis, which could be exploited to selectively kill cancer cells by dialing up CDK5 activity, instead of inhibiting it. CDK5 thus acts as a molecular rheostat, with different activity levels eliciting distinct functional outcomes. Finally, as CDK5's role is defined by its substrates, targeting them individually or in conjunction with CDK5 should create potentially valuable new clinical opportunities.
Collapse
Affiliation(s)
- Kumar Nikhil
- Department of Chemistry, Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, India
| | - Kavita Shah
- Department of Chemistry, Purdue University Center for Cancer Research, 560 Oval Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
18
|
Xu C, Yang K, Xuan Z, Li J, Liu Y, Zhao Y, Zheng Z, Bai Y, Shi Z, Shao C, Zhang L, Sun H. BCKDK regulates breast cancer cell adhesion and tumor metastasis by inhibiting TRIM21 ubiquitinate talin1. Cell Death Dis 2023; 14:445. [PMID: 37460470 PMCID: PMC10352378 DOI: 10.1038/s41419-023-05944-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023]
Abstract
Breast cancer is the most common malignant cancer in women worldwide. Cancer metastasis is the major cause of cancer-related deaths. BCKDK is associated with various diseases, including proliferation, migration, and invasion in multiple types of human cancers. However, the relevance of BCKDK to the development and progression of breast cancers and its function is unclear. This study found that BCKDK was overexpressed in breast cancer, associated with poor prognosis, and implicated in tumor metastasis. The downregulation of BCKDK expression inhibited the migration of human breast cancer cells in vitro and diminished lung metastasis in vivo. BCKDK perturbed the cadherin-catenin complex at the adherens junctions (AJs) and assembled focal adhesions (FAs) onto the extracellular matrix, thereby promoting the directed migration of breast cancer cells. We observed that BCKDK acted as a conserved regulator of the ubiquitination of cytoskeletal protein talin1 and the activation of the FAK/MAPK pathway. Further studies revealed that BCKDK inhibited the binding of talin1 to E3 ubiquitin ligase-TRIM21, leading to the decreased ubiquitination/degradation of talin1. In conclusion, identifying BCKDK as a biomarker for breast cancer metastasis facilitated further research on diagnostic biomarkers. Elucidating the mechanism by which BCKDK exerted its biological effect could provide a new theoretical basis for developing new markers for breast cancer metastasis and contribute to developing new therapies for the clinical treatment of breast cancer patients.
Collapse
Affiliation(s)
- Chunlan Xu
- School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Kunao Yang
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Zuodong Xuan
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Jinxin Li
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Yankuo Liu
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Yue Zhao
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Zeyuan Zheng
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Yang Bai
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Zhiyuan Shi
- School of Medicine, Xiamen University, 361102, Xiamen, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Chen Shao
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Lei Zhang
- School of Public Health, Xiamen University, Xiamen, China
| | - Huimin Sun
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361101, Xiamen, China.
| |
Collapse
|
19
|
Zhang P, Jiang H, Yang M, Bi C, Zhang K, Liu D, Wei M, Jiang Z, Lv K, Fang C, Liu J, Zhang T, Xu Y, Zhang J. AGK Potentiates Arterial Thrombosis by Affecting Talin-1 and αIIbβ3-Mediated Bidirectional Signaling Pathway. Arterioscler Thromb Vasc Biol 2023; 43:1015-1030. [PMID: 37051931 DOI: 10.1161/atvbaha.122.318647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/22/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND AGK (acylglycerol kinase) was first identified as a mitochondrial transmembrane protein that exhibits a lipid kinase function. Recent studies have established that AGK promotes cancer growth and metastasis, enhances glycolytic metabolism and function fitness of CD8+ T cells, or regulates megakaryocyte differentiation. However, the role of AGK in platelet activation and arterial thrombosis remains to be elaborated. METHODS We performed hematologic analysis using automated hematology analyzer and investigated platelets morphology by transmission electron microscope. We explored the role of AGK in platelet activation and arterial thrombosis utilizing transgenic mice, platelet functional experiments in vitro, and thrombosis models in vivo. We revealed the regulation effect of AGK on Talin-1 by coimmunoprecipitation, mass spectrometry, immunofluorescence, and Western blot. We tested the role of AGK on lipid synthesis of phosphatidic acid/lysophosphatidic acid and thrombin generation by specific Elisa kits. RESULTS In this study, we found that AGK depletion or AGK mutation had no effect on the platelet average volumes, the platelet microstructures, or the expression levels of the major platelet membrane receptors. However, AGK deficiency or AGK mutation conspicuously decreased multiple aspects of platelet activation, including agonists-induced platelet aggregation, granules secretion, JON/A binding, spreading on Fg (fibrinogen), and clot retraction. AGK deficiency or AGK mutation also obviously delayed arterial thrombus formation but had no effect on tail bleeding time and platelet procoagulant function. Mechanistic investigation revealed that AGK may promote Talin-1Ser425 phosphorylation and affect the αIIbβ3-mediated bidirectional signaling pathway. However, AGK does not affect lipid synthesis of phosphatidic acid/lysophosphatidic acid in platelets. CONCLUSIONS AGK, through its kinase activity, potentiates platelet activation and arterial thrombosis by promoting Talin-1 Ser425 phosphorylation and affecting the αIIbβ3-mediated bidirectional signaling pathway.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Haojie Jiang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Mina Yang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Changlong Bi
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Kandi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Dongsheng Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Meng Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Zheyi Jiang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China (K.L., C.F.)
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China (K.L., C.F.)
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Tiantian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| |
Collapse
|
20
|
Roy A, Prasad S, Chen Y, Chao Y, Liu Y, Zhao J, Wang QJ. Protein Kinase D2 and D3 Promote Prostate Cancer Cell Bone Metastasis by Positively Regulating Runx2 in a MEK/ERK1/2-Dependent Manner. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:624-637. [PMID: 36740185 PMCID: PMC10155267 DOI: 10.1016/j.ajpath.2023.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Advanced-stage prostate tumors metastasize to the bone, often causing death. The protein kinase D (PKD) family has been implicated in prostate cancer development; however, its role in prostate cancer metastasis remains elusive. This study examined the contribution of PKD, particularly PKD2 and PKD3 (PKD2/3), to the metastatic potential of prostate cancer cells and the effect of PKD inhibition on prostate cancer bone metastasis in vivo. Depletion of PKD2/3 by siRNAs or inhibition by the PKD inhibitor CRT0066101 in AR-positive and AR-negative castration-resistant prostate cancer cells potently inhibited colony formation and cell migration. Depletion or inhibition of PKD2/3 significantly blocked tumor cell invasion and suppressed the expression of genes related to bone metastasis in the highly invasive PC3-ML cells. The reduced invasive activity resulting from PKD2/3 depletion was in part mediated by the transcription factor Runx2, as its silencing decreased PKD2/3-mediated metastatic gene expression through the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 signaling axis. Furthermore, inhibition of PKD by CRT0066101 potently decreased the frequency of bone micrometastases in a mouse model of bone metastasis based on intracardiac injection of PC3-ML cells. These results indicate that PKD2/3 plays an important role in the bone metastasis of prostate cancer cells, and its inhibition may be beneficial for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Adhiraj Roy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Sahdeo Prasad
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yu Liu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jinjun Zhao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiming Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
21
|
Mao L, Wang L, Xu J, Zou J. The role of integrin family in bone metabolism and tumor bone metastasis. Cell Death Discov 2023; 9:119. [PMID: 37037822 PMCID: PMC10086008 DOI: 10.1038/s41420-023-01417-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Integrins have been the research focus of cell-extracellular matrix adhesion (ECM) and cytokine receptor signal transduction. They are involved in the regulation of bone metabolism of bone precursor cells, mesenchymal stem cells (MSCs), osteoblasts (OBs), osteoclasts (OCs), and osteocytes. Recent studies expanded and updated the role of integrin in bone metabolism, and a large number of novel cytokines were found to activate bone metabolism pathways through interaction with integrin receptors. Integrins act as transducers that mediate the regulation of bone-related cells by mechanical stress, fluid shear stress (FSS), microgravity, hypergravity, extracellular pressure, and a variety of physical factors. Integrins mediate bone metastasis of breast, prostate, and lung cancer by promoting cancer cell adhesion, migration, and survival. Integrin-mediated targeted therapy showed promising prospects in bone metabolic diseases. This review emphasizes the latest research results of integrins in bone metabolism and bone metastasis and provides a vision for treatment strategies.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Lian Wang
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, WA, 6009, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China.
| |
Collapse
|
22
|
Verhoeff TJ, Holloway AF, Dickinson JL. Non-coding RNA regulation of integrins and their potential as therapeutic targets in cancer. Cell Oncol (Dordr) 2023; 46:239-250. [PMID: 36512308 PMCID: PMC10060301 DOI: 10.1007/s13402-022-00752-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] [Accepted: 11/19/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Integrins are integral to cell signalling and management of the extracellular matrix, and exquisite regulation of their expression is essential for a variety of cell signalling pathways, whilst disordered regulation is a key driver of tumour progression and metastasis. Most recently non-coding RNAs in the form of micro-RNA (miRNA) and long non-coding RNA (lncRNA) have emerged as a key mechanism by which tissue dependent gene expression is controlled. Whilst historically these molecules have been poorly understood, advances in 'omic' technologies and a greater understanding of non-coding regions of the genome have revealed that non-coding RNAs make up a large proportion of the transcriptome. CONCLUSIONS AND PERSPECTIVES This review examines the regulation of integrin genes by ncRNAs, provides and overview of their mechanism of action and highlights how exploitation of these discoveries is informing the development of novel chemotherapeutic agents in the treatment of cancer. MiRNA molecules have been the most extensively characterised and negatively regulate most integrin genes, classically regulating genes through binding to recognition sequences in the mRNA 3'-untranslated regions of gene transcripts. LncRNA mechanisms of action are now being elucidated and appear to be more varied and complex, and may counter miRNA molecules, directly engage integrin mRNA transcripts, and guide or block both transcription factors and epigenetic machinery at integrin promoters or at other points in integrin regulation. Integrins as therapeutic targets are of enormous interest given their roles as oncogenes in a variety of tumours, and emerging therapeutics mimicking ncRNA mechanisms of action are already being trialled.
Collapse
Affiliation(s)
- Tristan Joseph Verhoeff
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia
| | - Adele F Holloway
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia.
| |
Collapse
|
23
|
Koizume S, Kanayama T, Kimura Y, Hirano H, Takahashi T, Ota Y, Miyazaki K, Yoshihara M, Nakamura Y, Yokose T, Kato H, Takenaka K, Sato S, Tadokoro H, Miyagi E, Miyagi Y. Cancer cell-derived CD69 induced under lipid and oxygen starvation promotes ovarian cancer progression through fibronectin. Cancer Sci 2023. [PMID: 36854451 DOI: 10.1111/cas.15774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer tissues generally have molecular oxygen and serum component deficiencies because of poor vascularization. Recently, we revealed that ICAM1 is strongly activated through lipophagy in ovarian clear cell carcinoma (CCC) cells in response to starvation of long-chain fatty acids and oxygen and confers resistance to apoptosis caused by these harsh conditions. CD69 is a glycoprotein that is synthesized in immune cells and is associated with their activation through cellular signaling pathways. However, the expression and function of CD69 in nonhematological cells is unclear. Here, we report that CD69 is induced in CCC cells as in ICAM1. Mass spectrometry analysis of phosphorylated peptides followed by pathway analysis revealed that CD69 augments CCC cell binding to fibronectin (FN) in association with the phosphorylation of multiple cellular signaling molecules including the focal adhesion pathway. Furthermore, CD69 synthesized in CCC cells could facilitate cell survival because the CD69-FN axis can induce epithelial-mesenchymal transition. Experiments with surgically removed tumor samples revealed that CD69 is predominantly expressed in CCC tumor cells compared with other histological subtypes of epithelial ovarian cancer. Overall, our data suggest that cancer cell-derived CD69 can contribute to CCC progression through FN.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Tomohiko Kanayama
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yayoi Kimura
- Advancer Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advancer Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Takahashi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yukihide Ota
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Obstetrics, Gynecology and Molecular Reproductive Science, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kaoru Miyazaki
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Hisamori Kato
- Department of Gynecology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Katsuya Takenaka
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| | - Hiroko Tadokoro
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Etsuko Miyagi
- Department of Obstetrics, Gynecology and Molecular Reproductive Science, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, Yokohama, Japan
| |
Collapse
|
24
|
Masi I, Ottavi F, Del Rio D, Caprara V, Vastarelli C, Giannitelli SM, Fianco G, Mozetic P, Buttarelli M, Ferrandina G, Scambia G, Gallo D, Rainer A, Bagnato A, Spadaro F, Rosanò L. The interaction of β-arrestin1 with talin1 driven by endothelin A receptor as a feature of α5β1 integrin activation in high-grade serous ovarian cancer. Cell Death Dis 2023; 14:73. [PMID: 36717550 PMCID: PMC9886921 DOI: 10.1038/s41419-023-05612-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023]
Abstract
Dissemination of high-grade serous ovarian cancer (HG-SOC) in the omentum and intercalation into a mesothelial cell (MC) monolayer depends on functional α5β1 integrin (Intα5β1) activity. Although the binding of Intα5β1 to fibronectin drives these processes, other molecular mechanisms linked to integrin inside-out signaling might support metastatic dissemination. Here, we report a novel interactive signaling that contributes to Intα5β1 activation and accelerates tumor cells toward invasive disease, involving the protein β-arrestin1 (β-arr1) and the activation of the endothelin A receptor (ETAR) by endothelin-1 (ET-1). As demonstrated in primary HG-SOC cells and SOC cell lines, ET-1 increased Intβ1 and downstream FAK/paxillin activation. Mechanistically, β-arr1 directly interacts with talin1 and Intβ1, promoting talin1 phosphorylation and its recruitment to Intβ1, thus fueling integrin inside-out activation. In 3D spheroids and organotypic models mimicking the omentum, ETAR/β-arr1-driven Intα5β1 signaling promotes the survival of cell clusters, with mesothelium-intercalation capacity and invasive behavior. The treatment with the antagonist of ETAR, Ambrisentan (AMB), and of Intα5β1, ATN161, inhibits ET-1-driven Intα5β1 activity in vitro, and tumor cell adhesion and spreading to intraperitoneal organs and Intβ1 activity in vivo. As a prognostic factor, high EDNRA/ITGB1 expression correlates with poor HG-SOC clinical outcomes. These findings highlight a new role of ETAR/β-arr1 operating an inside-out integrin activation to modulate the metastatic process and suggest that in the new integrin-targeting programs might be considered that ETAR/β-arr1 regulates Intα5β1 functional pathway.
Collapse
Affiliation(s)
- Ilenia Masi
- Institute of Molecular Biology and Pathology, CNR, Rome, 00185, Italy
| | - Flavia Ottavi
- Institute of Molecular Biology and Pathology, CNR, Rome, 00185, Italy
| | - Danila Del Rio
- Institute of Molecular Biology and Pathology, CNR, Rome, 00185, Italy
| | - Valentina Caprara
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS-Regina Elena National Cancer Institute, Rome, 00144, Italy
| | | | - Sara Maria Giannitelli
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, Rome, 00128, Italy
| | - Giulia Fianco
- Institute of Molecular Biology and Pathology, CNR, Rome, 00185, Italy
| | - Pamela Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council (CNR), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
- San Raffaele Hospital, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Via Olgettina, 60, Milan, 20132, Italy
| | - Marianna Buttarelli
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica-Sezione di Ginecologia ed Ostetricia-Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, Rome, 00168, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
| | - Gabriella Ferrandina
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica-Sezione di Ginecologia ed Ostetricia-Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, Rome, 00168, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
| | - Giovanni Scambia
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica-Sezione di Ginecologia ed Ostetricia-Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, Rome, 00168, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
| | - Daniela Gallo
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica-Sezione di Ginecologia ed Ostetricia-Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, Rome, 00168, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, Rome, 00128, Italy
- Institute of Nanotechnology (NANOTEC), National Research Council (CNR), c/o Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Anna Bagnato
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS-Regina Elena National Cancer Institute, Rome, 00144, Italy
| | - Francesca Spadaro
- Confocal Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, Rome, 00161, Italy
| | - Laura Rosanò
- Institute of Molecular Biology and Pathology, CNR, Rome, 00185, Italy.
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS-Regina Elena National Cancer Institute, Rome, 00144, Italy.
| |
Collapse
|
25
|
Wang EJY, Chen IH, Kuo BYT, Yu CC, Lai MT, Lin JT, Lin LYT, Chen CM, Hwang T, Sheu JJC. Alterations of Cytoskeleton Networks in Cell Fate Determination and Cancer Development. Biomolecules 2022; 12:biom12121862. [PMID: 36551290 PMCID: PMC9775460 DOI: 10.3390/biom12121862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/03/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
Cytoskeleton proteins have been long recognized as structural proteins that provide the necessary mechanical architecture for cell development and tissue homeostasis. With the completion of the cancer genome project, scientists were surprised to learn that huge numbers of mutated genes are annotated as cytoskeletal or associated proteins. Although most of these mutations are considered as passenger mutations during cancer development and evolution, some genes show high mutation rates that can even determine clinical outcomes. In addition, (phospho)proteomics study confirms that many cytoskeleton-associated proteins, e.g., β-catenin, PIK3CA, and MB21D2, are important signaling mediators, further suggesting their biofunctional roles in cancer development. With emerging evidence to indicate the involvement of mechanotransduction in stemness formation and cell differentiation, mutations in these key cytoskeleton components may change the physical/mechanical properties of the cells and determine the cell fate during cancer development. In particular, tumor microenvironment remodeling triggered by such alterations has been known to play important roles in autophagy, metabolism, cancer dormancy, and immune evasion. In this review paper, we will highlight the current understanding of how aberrant cytoskeleton networks affect cancer behaviors and cellular functions through mechanotransduction.
Collapse
Affiliation(s)
- Evan Ja-Yang Wang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - I-Hsuan Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung County 907391, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Brian Yu-Ting Kuo
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chia-Cheng Yu
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung County 907391, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Ministry of Health and Welfare, Taichung 403301, Taiwan
| | - Jen-Tai Lin
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813405, Taiwan
| | - Leo Yen-Ting Lin
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chih-Mei Chen
- Human Genetic Center, China Medical University Hospital, Taichung 404327, Taiwan
| | - Tritium Hwang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Correspondence: ; Tel.: +886-7-5252000 (ext. 7102)
| |
Collapse
|
26
|
Braidotti N, Chen SN, Long CS, Cojoc D, Sbaizero O. Piezo1 Channel as a Potential Target for Hindering Cardiac Fibrotic Remodeling. Int J Mol Sci 2022; 23:8065. [PMID: 35897650 PMCID: PMC9330509 DOI: 10.3390/ijms23158065] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Fibrotic tissues share many common features with neoplasms where there is an increased stiffness of the extracellular matrix (ECM). In this review, we present recent discoveries related to the role of the mechanosensitive ion channel Piezo1 in several diseases, especially in regulating tumor progression, and how this can be compared with cardiac mechanobiology. Based on recent findings, Piezo1 could be upregulated in cardiac fibroblasts as a consequence of the mechanical stress and pro-inflammatory stimuli that occurs after myocardial injury, and its increased activity could be responsible for a positive feedback loop that leads to fibrosis progression. The increased Piezo1-mediated calcium flow may play an important role in cytoskeleton reorganization since it induces actin stress fibers formation, a well-known characteristic of fibroblast transdifferentiation into the activated myofibroblast. Moreover, Piezo1 activity stimulates ECM and cytokines production, which in turn promotes the phenoconversion of adjacent fibroblasts into new myofibroblasts, enhancing the invasive character. Thus, by assuming the Piezo1 involvement in the activation of intrinsic fibroblasts, recruitment of new myofibroblasts, and uncontrolled excessive ECM production, a new approach to blocking the fibrotic progression can be predicted. Therefore, targeted therapies against Piezo1 could also be beneficial for cardiac fibrosis.
Collapse
Affiliation(s)
- Nicoletta Braidotti
- Department of Physics, University of Trieste, Via A. Valerio 2, 34127 Trieste, Italy;
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Suet Nee Chen
- CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, 12700 East 19th Ave., Aurora, CO 80045, USA;
| | - Carlin S. Long
- Center for the Prevention of Heart and Vascular Disease, University of California, 555 Mission Bay Blvd South, Rm 352K, San Francisco, CA 94143, USA;
| | - Dan Cojoc
- Institute of Materials, National Research Council of Italy (CNR-IOM), Area Science Park Basovizza, Strada Statale 14, Km 163,5, 34149 Trieste, Italy;
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6/A, 34127 Trieste, Italy
| |
Collapse
|
27
|
Tsaur I, Thomas A, Monecke M, Zugelder M, Rutz J, Grein T, Maxeiner S, Xie H, Chun FKH, Rothweiler F, Cinatl J, Michaelis M, Haferkamp A, Blaheta RA. Amygdalin Exerts Antitumor Activity in Taxane-Resistant Prostate Cancer Cells. Cancers (Basel) 2022; 14:cancers14133111. [PMID: 35804883 PMCID: PMC9265127 DOI: 10.3390/cancers14133111] [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: 05/29/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Despite recent advances in the treatment of metastatic prostate cancer (PCa), resistance development after taxane treatments is inevitable, necessitating effective options to combat drug resistance. Previous studies indicated antitumoral properties of the natural compound amygdalin. However, whether amygdalin acts on drug-resistant tumor cells remains questionable. An in vitro study was performed to investigate the influence of amygdalin (10 mg/mL) on the growth of a panel of therapy-naïve and docetaxel- or cabazitaxel-resistant PCa cell lines (PC3, DU145, and LNCaP cells). Tumor growth, proliferation, clonal growth, and cell cycle progression were investigated. The cell cycle regulating proteins (phospho)cdk1, (phospho)cdk2, cyclin A, cyclin B, p21, and p27 and the mammalian target of rapamycin (mTOR) pathway proteins (phospho)Akt, (phospho)Raptor, and (phospho)Rictor as well as integrin β1 and the cytoskeletal proteins vimentin, ezrin, talin, and cytokeratin 8/18 were assessed. Furthermore, chemotactic activity and adhesion to extracellular matrix components were analyzed. Amygdalin dose-dependently inhibited tumor growth and reduced tumor clones in all (parental and resistant) PCa cell lines, accompanied by a G0/G1 phase accumulation. Cell cycle regulating proteins were significantly altered by amygdalin. A moderate influence of amygdalin on tumor cell adhesion and chemotaxis was observed as well, paralleled by modifications of cytoskeletal proteins and the integrin β1 expression level. Amygdalin may, therefore, block tumor growth and disseminative characteristics of taxane-resistant PCa cells. Further studies are warranted to determine amygdalin’s value as an antitumor drug.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| | - Anita Thomas
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
- Correspondence: ; Tel.: +49-6131-172312; Fax: +49-6131-173827
| | - Michelle Monecke
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Marion Zugelder
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Jochen Rutz
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Timothy Grein
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Sebastian Maxeiner
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Hui Xie
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Felix K.-H. Chun
- Department of Urology, Goethe-University, 60590 Frankfurt am Main, Germany; (M.M.); (M.Z.); (J.R.); (T.G.); (S.M.); (H.X.); (F.K.-H.C.)
| | - Florian Rothweiler
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany; (F.R.)
- Petra Joh-Forschungshaus, 60528 Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany; (F.R.)
- Petra Joh-Forschungshaus, 60528 Frankfurt am Main, Germany
| | - Martin Michaelis
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK;
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| | - Roman A. Blaheta
- Department of Urology and Pediatric Urology, University Medicine Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (I.T.); (A.H.); (R.A.B.)
| |
Collapse
|
28
|
Hua T, Zhao BB, Fan SB, Zhao CF, Kong YH, Tian RQ, Zhang BY. Prognostic implications of PPL expression in ovarian cancer. Discov Oncol 2022; 13:35. [PMID: 35612641 PMCID: PMC9133299 DOI: 10.1007/s12672-022-00496-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022] Open
Abstract
Periplakin (PPL) is a main member in plakin family, which plays important role in cellular adhesion complexes supporting and cytoskeletal integrity supplying. PPL was reported to be a potential biomarker candidate for several types of cancers. However, the biological functions and underlying mechanisms of PPL in ovarian cancer (OV) remain unclear. In the present study, we used GEPIA 2, Human Protein Atlas, Oncomine, LinkedOmics, Kaplan-Meier Plotter, STRING, CytoHubba plug-in and TIMER to determine the associations among PPL expression, prognosis, and immune cell infiltration in OV. RT-qPCR and IHC analysis were conducted to validated the role of PPL in an independent OV cohort. Compared with the normal ovary tissues, the levels of PPL mRNA and protein expression were both obviously higher in OV tumors from multiple datasets (P < 0.05), and a poor survival was observed to be strongly correlated with high PPL expression (P < 0.05). Moreover, the results were further validated by RT-qPCR and IHC analysis in an independent OV cohort. A gene-clinical nomogram was constructed, including PPL mRNA expression and clinical factors in TCGA. Functional network analysis suggested that PPL participates in the important pathways like Wnt signaling pathway, MAPK signaling pathway. Ten hub genes (LAMC2, PXN, LAMA3, LAMB3, LAMA5, ITGA3, TLN1, ACTN4, ACTN1, and ITGB4) were identified to be positively associated with PPL. Furthermore, PPL expression was negatively correlated with infiltrating levels of CD4+ T cell, macrophages, neutrophils, and dendritic cells. In conclusion, PPL may be an unfavorable prognostic biomarker candidate in OV, which was also correlated with immune infiltrating and function in immunotherapy response.
Collapse
Affiliation(s)
- Tian Hua
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China.
| | - Bei-Bei Zhao
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| | - Shao-Bei Fan
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| | - Cai-Fen Zhao
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| | - Yun-Hong Kong
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| | - Rui-Qing Tian
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| | - Bao-Ying Zhang
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medial University, 16 Hongxing Road, Xingtai, 054001, Hebei, People's Republic of China
| |
Collapse
|
29
|
Zhang Y, Sun L, Li H, Ai L, Ma Q, Qiao X, Yang J, Zhang H, Ou X, Wang Y, Chen G, Xue J, Zhu X, Zhao Y, Yang Y, Liu C. Binding blockade between TLN1 and integrin β1 represses triple-negative breast cancer. eLife 2022; 11:e68481. [PMID: 35285795 PMCID: PMC8937232 DOI: 10.7554/elife.68481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Background Integrin family are known as key gears in focal adhesion for triple-negative breast cancer (TNBC) metastasis. However, the integrin independent factor TLN1 remains vague in TNBC. Methods Bioinformatics analysis was performed based on TCGA database and Shengjing Hospital cohort. Western blot and RT-PCR were used to detect the expression of TLN1 and integrin pathway in cells. A small-molecule C67399 was screened for blocking TLN1 and integrin β1 through a novel computational screening approach by targeting the protein-protein binding interface. Drug pharmacodynamics were determined through xenograft assay. Results Upregulation of TLN1 in TNBC samples correlates with metastasis and worse prognosis. Silencing TLN1 in TNBC cells significantly attenuated the migration of tumour cells through interfering the dynamic formation of focal adhesion with integrin β1, thus regulating FAK-AKT signal pathway and epithelial-mesenchymal transformation. Targeting the binding between TLN1 and integrin β1 by C67399 could repress metastasis of TNBC. Conclusions TLN1 overexpression contributes to TNBC metastasis and C67399 targeting TLN1 may hold promise for TNBC treatment. Funding This study was supported by grants from the National Natural Science Foundation of China (No. 81872159, 81902607, 81874301), Liaoning Colleges Innovative Talent Support Program (Name: Cancer Stem Cell Origin and Biological Behaviour), Outstanding Scientific Fund of Shengjing Hospital (201803), and Outstanding Young Scholars of Liaoning Province (2019-YQ-10).
Collapse
Affiliation(s)
- Yixiao Zhang
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Lisha Sun
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning ProvinceShenyangChina
| | - Haonan Li
- School of Bioengineering, Dalian University of TechnologyDalianChina
| | - Liping Ai
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Qingtian Ma
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Xinbo Qiao
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Jie Yang
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Hao Zhang
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Xunyan Ou
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Yining Wang
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Guanglei Chen
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Jinqi Xue
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Xudong Zhu
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
| | - Yu Zhao
- Department of Biochemistry and Molecular Biology, Mayo ClinicRochesterUnited States
| | - Yongliang Yang
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- School of Bioengineering, Dalian University of TechnologyDalianChina
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical UniversityShenyangChina
- Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical UniversityShenyangChina
- Innovative Cancer Drug Research and Development Engineering Center of Liaoning ProvinceShenyangChina
| |
Collapse
|
30
|
Gilardi M, Bersini S, Valtorta S, Proietto M, Crippa M, Boussommier-Calleja A, Labelle M, Moresco RM, Vanoni M, Kamm RD, Moretti M. The driving role of the Cdk5/Tln1/FAK S732 axis in cancer cell extravasation dissected by human vascularized microfluidic models. Biomaterials 2021; 276:120975. [PMID: 34333365 DOI: 10.1016/j.biomaterials.2021.120975] [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: 06/30/2020] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Understanding the molecular mechanisms of metastatic dissemination, the leading cause of death in cancer patients, is required to develop novel, effective therapies. Extravasation, an essential rate-limiting process in the metastatic cascade, includes three tightly coordinated steps: cancer cell adhesion to the endothelium, trans-endothelial migration, and early invasion into the secondary site. Focal adhesion proteins, including Tln1 and FAK, regulate the cytoskeleton dynamics: dysregulation of these proteins is often associated with metastatic progression and poor prognosis. METHODS Here, we studied the previously unexplored role of these targets in each extravasation step using engineered 3D in vitro models, which recapitulate the physiological vascular niche experienced by cancer cells during hematogenous metastasis. RESULTS Human breast cancer and fibrosarcoma cell lines respond to Cdk5/Tln1/FAK axis perturbation, impairing their metastatic potential. Vascular breaching requires actin polymerization-dependent invadopodia formation. Invadopodia generation requires the structural function of FAK and Tln1 rather than their activation through phosphorylation. Our data support that the inhibition of FAKS732 phosphorylation delocalizes ERK from the nucleus, decreasing ERK phosphorylated form. These findings indicate the critical role of these proteins in driving trans-endothelial migration. In fact, both knock-down experiments and chemical inhibition of FAK dramatically reduces lung colonization in vivo and TEM in microfluidic setting. Altogether, these data indicate that engineered 3D in vitro models coupled to in vivo models, genetic, biochemical, and imaging tools represent a powerful weapon to increase our understanding of metastatic progression. CONCLUSIONS These findings point to the need for further analyses of previously overlooked phosphorylation sites of FAK, such as the serine 732, and foster the development of new effective antimetastatic treatments targeting late events of the metastatic cascade.
Collapse
Affiliation(s)
- Mara Gilardi
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; Institute of Pathology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Simone Bersini
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Silvia Valtorta
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Proietto
- Department of Biology-University of California - San Diego, La Jolla, CA, USA.
| | - Martina Crippa
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Laboratory of Biological Structures Mechanics, Chemistry, Material and Chemical Engineering Department "Giulio Natta", Politecnico di Milano, Milan, Italy.
| | - Alexandra Boussommier-Calleja
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Rosa Maria Moresco
- Università Degli Studi di Milano-Bicocca, Department of Medicine and Surgery and Tecnomed Foundation, Monza, Italy; Institute of Bioimaging and Molecular Physiology of National Researches Council (IBFM-CNR), Segrate, Italy.
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126, Milan, Italy; ISBE.IT/ Centre of Systems Biology, Milano, Italy.
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA.
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Euler Institute, Biomedical Sciences Faculty, Università Della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
31
|
Chen S, Liu B, Li J, Liao S, Bi Y, Huang W, Yuan L, Yang Y, Qin A. Talin1 regulates endometrial adhesive capacity through the Ras signaling pathway. Life Sci 2021; 274:119332. [PMID: 33711384 DOI: 10.1016/j.lfs.2021.119332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 01/13/2023]
Abstract
AIMS Blastocyst implantation is mainly depended on the adhesion between cells and cell matrix. Endometrial adhesion plays an important role in establishing embryo implantation, but the underlying mechanisms are remains unclear. Talin1 is a local adhesion complex protein that is necessary for cell adhesion and movement. However, the role and mechanisms of Talin1 in embryo implantation are still unclear. MAIN METHODS The expression of Talin1 and Integrin αvβ3 was measured in the receptive endometrium from the RIF (Recurrent implantation failure) cohort and NC (normal fertile control group) cohort. A JEG-3 trophoblast and endometrial epithelial cell adhesion model and pregnant mouse model were established. The molecular mechanism of Talin1-mediated cell adhesion was explored by RNA sequencing, RT-qPCR, as well as western blotting assays. KEY FINDINGS Talin1 enhances endometrial cell adhesion by regulating the Ras signaling pathway, and ultimately facilitates embryo implantation. SIGNIFICANCE This study revealed the molecular mechanisms of regarding the pathogenesis of RIF caused by endometrial receptivity insufficiency. Further pharmacological research on the Ras signaling pathway would be valuable and might provide new therapeutic targets for RIF patients.
Collapse
Affiliation(s)
- Saiqiong Chen
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China; Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Bo Liu
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jingjing Li
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Shengbin Liao
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yin Bi
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Weiyu Huang
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Lifang Yuan
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yihua Yang
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Aiping Qin
- Center of Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
32
|
Hinz N, Jücker M. AKT in Bone Metastasis of Solid Tumors: A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13102287. [PMID: 34064589 PMCID: PMC8151478 DOI: 10.3390/cancers13102287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastasis is a frequent complication of solid tumors and leads to a reduced overall survival. Although much progress has been made in the field of tumor therapy in the last years, bone metastasis depicts a stage of the disease with a lack of appropriate therapeutical options. Hence, this review aims to present the role of AKT in bone metastasis of solid tumors to place the spotlight on AKT as a possible therapeutical approach for patients with bone metastases. Furthermore, we intended to discuss postulated underlying molecular mechanisms of the bone metastasis-promoting effect of AKT, especially in highly bone-metastatic breast, prostate, and lung cancer. To conclude, this review identified the AKT kinase as a potential therapeutical target in bone metastasis and revealed remaining questions, which need to be addressed in further research projects. Abstract Solid tumors, such as breast cancer and prostate cancer, often form bone metastases in the course of the disease. Patients with bone metastases frequently develop complications, such as pathological fractures or hypercalcemia and exhibit a reduced life expectancy. Thus, it is of vital importance to improve the treatment of bone metastases. A possible approach is to target signaling pathways, such as the PI3K/AKT pathway, which is frequently dysregulated in solid tumors. Therefore, we sought to review the role of the serine/threonine kinase AKT in bone metastasis. In general, activation of AKT signaling was shown to be associated with the formation of bone metastases from solid tumors. More precisely, AKT gets activated in tumor cells by a plethora of bone-derived growth factors and cytokines. Subsequently, AKT promotes the bone-metastatic capacities of tumor cells through distinct signaling pathways and secretion of bone cell-stimulating factors. Within the crosstalk between tumor and bone cells, also known as the vicious cycle, the stimulation of osteoblasts and osteoclasts also causes activation of AKT in these cells. As a consequence, bone metastasis is reduced after experimental inhibition of AKT. In summary, AKT signaling could be a promising therapeutical approach for patients with bone metastases of solid tumors.
Collapse
|
33
|
Crippa M, Bersini S, Gilardi M, Arrigoni C, Gamba S, Falanga A, Candrian C, Dubini G, Vanoni M, Moretti M. A microphysiological early metastatic niche on a chip reveals how heterotypic cell interactions and inhibition of integrin subunit β 3 impact breast cancer cell extravasation. LAB ON A CHIP 2021; 21:1061-1072. [PMID: 33522559 DOI: 10.1039/d0lc01011a] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
During metastatic progression multiple players establish competitive mechanisms, whereby cancer cells (CCs) are exposed to both pro- and anti-metastatic stimuli. The early metastatic niche (EMN) is a transient microenvironment which forms in the circulation during CC dissemination. EMN is characterized by the crosstalk among CCs, platelets, leukocytes and endothelial cells (ECs), increasing CC ability to extravasate and colonize secondary tissues. To better understand this complex crosstalk, we designed a human "EMN-on-a-chip" which involves the presence of blood cells as compared to standard metastases-on-chip models, hence providing a microenvironment more similar to the in vivo situation. We showed that CC transendothelial migration (TEM) was significantly increased in the presence of neutrophils and platelets in the EMN-on-a-chip compared to CC alone. Moreover, exploiting the EMN-on-chip in combination with multi-culture experiments, we showed that platelets increased the expression of epithelial to mesenchymal transition (EMT) markers in CCs and that the addition of a clinically approved antiplatelet drug (eptifibatide, inhibiting integrin β3) impaired platelet aggregation and decreased CC expression of EMT markers. Inhibition of integrin β3 in the co-culture system modulated the activation of the Src-FAK-VE-cadherin signaling axis and partially restored the architecture of inter-endothelial junctions by limiting VE-cadherinY658 phosphorylation and its nuclear localization. These observations correlate with the decreased CC TEM observed in the presence of integrin β3 inhibitor. Our EMN-on-a-chip can be easily implemented for drug repurposing studies and to investigate new candidate molecules counteracting CC extravasation.
Collapse
Affiliation(s)
- Martina Crippa
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Tang X, Li Q, Li L, Jiang J. Expression of Talin-1 in endometriosis and its possible role in pathogenesis. Reprod Biol Endocrinol 2021; 19:42. [PMID: 33750407 PMCID: PMC7942010 DOI: 10.1186/s12958-021-00725-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/23/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Endometriosis is a disease that involves active cell invasion and migration. Talin-1 can promote cell invasion, migration and adhension in various cancer cells, but its role in endometriosis has not been investigated. This study was to investigate the expression level of Talin-1 in endometriosis and the role of Talin-1 in the proliferation, adhesion, migration, and invasion of human endometrial stromal cells (ESCs). METHODS Ectopic and eutopic endometrial tissues were collected from women with endometriosis, and the control endometrial tissues were obtained from patients without endometriosis. The expression level of Talin-1 was detected in each sample using quantitative real-time polymerase chain reaction and immunohistochemistry. The expression of Talin-1 was inhibited using RNA interference in ESCs, and its proliferation, apoptosis, adhesion, migration, and invasion capacity were analyzed. Western blotting was performed to detect the expression of related molecules after the downregulation of Talin-1. RESULTS The results showed that the mRNA and protein expression of Talin-1 were significantly increased in the ectopic endometrium and eutopic endometrial tissues compared with the controls. The knockdown of Talin-1 did not affect the proliferation and apoptosis of ESCs. The results indicated that the downexpression of Talin-1 inhibited the adhesion, invasion, and migration of ESCs. In addition, the expressions of N-cadherin, MMP-2, and integrin β3 were significantly lower after the deregulation of Talin-1, whereas the levels of E-cadherin were significantly increased. CONCLUSIONS The expression of Talin-1 was increased in the ectopic and eutopic endometrial tissues compared with the control endometrium. The downregulation of Talin-1 inhibited the adhesion, invasion, and migration of ESCs.
Collapse
Affiliation(s)
- Xian Tang
- Department of Obstetrics and Gynecology, Loudi Central Hospital of Hunan Province, Loudi, Hunan Province, China
| | - Qing Li
- Department of Gynecology, The Third Xiangya Hospital, Central South University, NO.138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Lijie Li
- Department of Gynecology, The Third Xiangya Hospital, Central South University, NO.138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Jianfa Jiang
- Department of Gynecology, The Third Xiangya Hospital, Central South University, NO.138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
35
|
Ruggiero C, Lalli E. Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev 2021; 40:89-140. [PMID: 33471283 DOI: 10.1007/s10555-020-09936-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Cancer is a pathology characterized by a loss or a perturbation of a number of typical features of normal cell behaviour. Indeed, the acquisition of an inappropriate migratory and invasive phenotype has been reported to be one of the hallmarks of cancer. The cytoskeleton is a complex dynamic network of highly ordered interlinking filaments playing a key role in the control of fundamental cellular processes, like cell shape maintenance, motility, division and intracellular transport. Moreover, deregulation of this complex machinery contributes to cancer progression and malignancy, enabling cells to acquire an invasive and metastatic phenotype. Metastasis accounts for 90% of death from patients affected by solid tumours, while an efficient prevention and suppression of metastatic disease still remains elusive. This results in the lack of effective therapeutic options currently available for patients with advanced disease. In this context, the cytoskeleton with its regulatory and structural proteins emerges as a novel and highly effective target to be exploited for a substantial therapeutic effort toward the development of specific anti-metastatic drugs. Here we provide an overview of the role of cytoskeleton components and interacting proteins in cancer metastasis with a special focus on small molecule compounds interfering with the actin cytoskeleton organization and function. The emerging involvement of microtubules and intermediate filaments in cancer metastasis is also reviewed.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560, Valbonne, France.
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| | - Enzo Lalli
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des Lucioles - Sophia Antipolis, 06560, Valbonne, France
| |
Collapse
|
36
|
Wang Y, Zhou J, Tang C, Yu J, Zhu W, Guo J, Wang Y. Positive effect of Astragaloside IV on neurite outgrowth via talin-dependent integrin signaling and microfilament force. J Cell Physiol 2021; 236:2156-2168. [PMID: 32853433 DOI: 10.1002/jcp.30002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022]
Abstract
Integrin plays a prominent role in neurite outgrowth by transmitting both mechanical and chemical signals. Integrin expression is closely associated with Astragaloside IV (AS-IV), the main component extracted from Astragali radix, which has a positive effect on neural-protection. However, the relationship between AS-IV and neurite outgrowth has not been studied exhaustively to date. The present study investigated the underlying mechanism of AS-IV on neurite outgrowth. Longer neurites have been observed in SH-SY5Y cells or cortical neurons after AS-IV treatment. Furthermore, AS-IV not only increased the expression of integrin β but also activated it. The AS-IV-induced increased integrin activity was attributed to the integrin-activating protein talin. Application of the actin force probe showed that AS-IV led to an increase in intracellular microfilament force during neurite growth. Furthermore, in response to AS-IV, the microfilament force was regulated by talin and integrin activity during neurite growth. These results suggest that AS-IV has the ability to increase intracellular structural force and facilitate neurite elongation by integrin signaling, which highlights its therapeutic potential for neurite outgrowth.
Collapse
Affiliation(s)
- Yifan Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jingwen Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Chuanfeng Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jia Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wen Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jun Guo
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
37
|
Uhlorn JA, Husband NA, Romero‐Aleshire MJ, Moffett C, Lindsey ML, Langlais PR, Brooks HL. CD4 + T Cell-Specific Proteomic Pathways Identified in Progression of Hypertension Across Postmenopausal Transition. J Am Heart Assoc 2021; 10:e018038. [PMID: 33410333 PMCID: PMC7955317 DOI: 10.1161/jaha.120.018038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Background Menopause is associated with an increase in the prevalence and severity of hypertension in women. Although premenopausal females are protected against T cell-dependent immune activation and development of angiotensin II (Ang II) hypertension, this protection is lost in postmenopausal females. Therefore, the current study hypothesized that specific CD4+ T cell pathways are regulated by sex hormones and Ang II to mediate progression from premenopausal protection to postmenopausal hypertension. Methods and Results Menopause was induced in C57BL/6 mice via repeated 4-vinylcyclohexene diepoxide injections, while premenopausal females received sesame oil vehicle. A subset of premenopausal mice and all menopausal mice were infused with Ang II for 14 days (Control, Ang II, Meno/Ang II). Proteomic and phosphoproteomic profiles of CD4+ T cells isolated from spleens were examined. Ang II markedly increased CD4+ T cell protein abundance and phosphorylation associated with DNA and histone methylation in both premenopausal and postmenopausal females. Compared with premenopausal T cells, Ang II infusion in menopausal mice increased T cell phosphorylation of MP2K2, an upstream regulator of ERK, and was associated with upregulated phosphorylation at ERK targeted sites. Additionally, Ang II infusion in menopausal mice decreased T cell phosphorylation of TLN1, a key regulator of IL-2Rα and FOXP3 expression. Conclusions These findings identify novel, distinct T cell pathways that influence T cell-mediated inflammation during postmenopausal hypertension.
Collapse
Affiliation(s)
- Joshua A. Uhlorn
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | | | | | - Caitlin Moffett
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Merry L. Lindsey
- Department of Cellular and Integrative PhysiologyCenter for Heart and Vascular ResearchNebraska‐Western Iowa Health Care SystemUniversity of Nebraska Medical Center and Research ServiceOmahaNE
| | - Paul R. Langlais
- Department of MedicineCollege of MedicineUniversity of ArizonaTucsonAZ
| | - Heddwen L. Brooks
- Department of PhysiologyCollege of MedicineUniversity of ArizonaTucsonAZ
| |
Collapse
|
38
|
Su B, Wu J. Phosphorylation of RIAM Activates Its Adaptor Function in Mediating Integrin Signaling. JOURNAL OF CELLULAR SIGNALING 2021; 2:103-110. [PMID: 35128538 PMCID: PMC8813058 DOI: 10.33696/signaling.2.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrins are cellular receptors that regulate cell adhesion and many other cellular functions. Integrins can be activated via an "inside-out pathway" that is promoted by RAP1 GTPase. RAP1-GTP-Interacting Adaptor Molecular (RIAM) mediates integrin activation by linking RAP1 GTPase to talin, an integrin activator. RIAM's function in integrin signaling is tightly regulated. In this commentary, we review recent studies of the molecular mechanisms underlying RIAM autoinhibition via both intramolecular interaction and oligomer assembly, and the phosphorylation-dependent activation of RIAM.
Collapse
Affiliation(s)
| | - Jinhua Wu
- Correspondence should be addressed to Jinhua Wu;
| |
Collapse
|
39
|
Zhang P, Azizi L, Kukkurainen S, Gao T, Baikoghli M, Jacquier MC, Sun Y, Määttä JAE, Cheng RH, Wehrle-Haller B, Hytönen VP, Wu J. Crystal structure of the FERM-folded talin head reveals the determinants for integrin binding. Proc Natl Acad Sci U S A 2020; 117:32402-32412. [PMID: 33288722 PMCID: PMC7768682 DOI: 10.1073/pnas.2014583117] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Binding of the intracellular adapter proteins talin and its cofactor, kindlin, to the integrin receptors induces integrin activation and clustering. These processes are essential for cell adhesion, migration, and organ development. Although the talin head, the integrin-binding segment in talin, possesses a typical FERM-domain sequence, a truncated form has been crystallized in an unexpected, elongated form. This form, however, lacks a C-terminal fragment and possesses reduced β3-integrin binding. Here, we present a crystal structure of a full-length talin head in complex with the β3-integrin tail. The structure reveals a compact FERM-like conformation and a tightly associated N-P-L-Y motif of β3-integrin. A critical C-terminal poly-lysine motif mediates FERM interdomain contacts and assures the tight association with the β3-integrin cytoplasmic segment. Removal of the poly-lysine motif or disrupting the FERM-folded configuration of the talin head significantly impairs integrin activation and clustering. Therefore, structural characterization of the FERM-folded active talin head provides fundamental understanding of the regulatory mechanism of integrin function.
Collapse
Affiliation(s)
- Pingfeng Zhang
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Sampo Kukkurainen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Tong Gao
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616
| | - Marie-Claude Jacquier
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211 Geneva 4, Switzerland
| | - Yijuan Sun
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Juha A E Määttä
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, 1211 Geneva 4, Switzerland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, FI-33520 Tampere, Finland;
- Department of Clinical Chemistry, Fimlab Laboratories, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA 19111;
| |
Collapse
|
40
|
You S, He X, Wang M, Mao L, Zhang L. Tanshinone IIA Suppresses Glioma Cell Proliferation, Migration and Invasion Both in vitro and in vivo Partially Through miR-16-5p/Talin-1 (TLN1) Axis. Cancer Manag Res 2020; 12:11309-11320. [PMID: 33192091 PMCID: PMC7654526 DOI: 10.2147/cmar.s256347] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/01/2020] [Indexed: 01/13/2023] Open
Abstract
Background Tanshinone IIA (TIIA) is one of the active constituents derived from the rhizome of Danshen, a traditional Chinese herbal. Recently, microRNAs (miRNAs) have been suggested to be associated with the anticancer role of TIIA. However, it remains vague of the interaction between miRNAs and TIIA in glioma, a common aggressive brain tumor in humans. Methods Expression of miRNA (miR)-16-5p and talin-1 (TLN1) was detected using reverse transcription-quantitative polymerase chain reaction and Western blotting. Cell proliferation, migration and invasion were assessed with cell viability assay, transwell assay, Western blotting, and xenograft tumor experiment. The target binding between miR-16-5p and TLN1 was confirmed by dual-luciferase reporter assay and RNA pull-down assay. Results TIIA treatment inhibited cell viability, migration and invasion, and decreased Cyclin D1, matrix metalloproteinase (MMP)-9 and Vimentin expression in glioma T98G and A172 cells both in vitro and in vivo. Thus, TIIA induced anti-glioma role, wherein miR-16-5p was upregulated and TLN1 was downregulated. Moreover, silencing miR-16-5p could abate TIIA-mediated suppression on glioma cell proliferation, migration and invasion in vitro and in vivo. TLN1 overexpression also exerted tumor-promoting effect in TIIA-treated T98G and A172 cells. Mechanically, miR-16-5p could regulate TLN1 expression via target binding, and depleting TLN1 could counteract the inhibitory effect of miR-16-5p knockdown on the curative effect of TIIA in T98G and A172 cells. Conclusion TIIA exerted the anti-proliferation, anti-migration and anti-invasion role in glioma cells both in vitro and in vivo partially through regulating miR-16-5p/TLN1 axis.
Collapse
Affiliation(s)
- Shihao You
- Department of Neurology, Qingdao Fuwai Cardiovascular Hospital, Qingdao, Shandong, People's Republic of China
| | - Xianghui He
- Department of Emergency, Qingdao Fuwai Cardiovascular Hospital, Qingdao, Shandong, People's Republic of China
| | - Mei Wang
- Department of Neurology, Qingdao Fuwai Cardiovascular Hospital, Qingdao, Shandong, People's Republic of China
| | - Lina Mao
- Department of Neurology, Qingdao Fuwai Cardiovascular Hospital, Qingdao, Shandong, People's Republic of China
| | - Lu Zhang
- Department of Peripheral Vascular Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| |
Collapse
|
41
|
Rivello F, Matuła K, Piruska A, Smits M, Mehra N, Huck WTS. Probing single-cell metabolism reveals prognostic value of highly metabolically active circulating stromal cells in prostate cancer. SCIENCE ADVANCES 2020; 6:6/40/eaaz3849. [PMID: 32998889 PMCID: PMC7527228 DOI: 10.1126/sciadv.aaz3849] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 08/06/2020] [Indexed: 05/05/2023]
Abstract
Despite their important role in metastatic disease, no general method to detect circulating stromal cells (CStCs) exists. Here, we present the Metabolic Assay-Chip (MA-Chip) as a label-free, droplet-based microfluidic approach allowing single-cell extracellular pH measurement for the detection and isolation of highly metabolically active cells (hm-cells) from the tumor microenvironment. Single-cell mRNA-sequencing analysis of the hm-cells from metastatic prostate cancer patients revealed that approximately 10% were canonical EpCAM+ hm-CTCs, 3% were EpCAM- hm-CTCs with up-regulation of prostate-related genes, and 87% were hm-CStCs with profiles characteristic for cancer-associated fibroblasts, mesenchymal stem cells, and endothelial cells. Kaplan-Meier analysis shows that metastatic prostate cancer patients with more than five hm-cells have a significantly poorer survival probability than those with zero to five hm-cells. Thus, prevalence of hm-cells is a prognosticator of poor outcome in prostate cancer, and a potentially predictive and therapy response biomarker for agents cotargeting stromal components and preventing epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Francesca Rivello
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Kinga Matuła
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Aigars Piruska
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Minke Smits
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands.
| |
Collapse
|
42
|
Lv Z, Li W, Wei X. S100A9 promotes prostate cancer cell invasion by activating TLR4/NF-κB/integrin β1/FAK signaling. Onco Targets Ther 2020; 13:6443-6452. [PMID: 32884282 PMCID: PMC7435298 DOI: 10.2147/ott.s192250] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 04/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background S100A9, which is expressed in prostate cancer, has been reported in association with prostate cancer progression. However, the role of S100A9 in prostate cancer metastasis is largely unknown. The aim of this study was to investigate the effect of S100A9 on prostate cancer cell invasion and the involved mechanisms. Materials and methods Integrin β1 expression in PC-3 and DU-145 cells was determined by quantitative real-time polymerase chain reaction (PCR) (qRT-PCR) and Western blot. Cellular invasion was measured by transwell invasion assay. Western blot was used to determine protein expression. Concentrations of S100A9 and fibronectin were analyzed by enzyme-linked immunosorbent assay. The protein interaction was detected by immunoprecipitation. The NF-κB activity was measured by luciferase reporter assay. The DU-145 cells metastasis in vivo was determined in mice xenograft models after S100A9 overexpression. Results S100A9 promoted prostate cancer cells invasion, integrin β1 expression and fibronectin secretion. Further investigation evidenced that S100A9 interacted with Toll-like receptor 4 (TLR4) and activated NF-κB, which was responsible for tumor cell invasion, integrin β1 up-regulation and focal adhesion kinase (FAK) phosphorylation. Furthermore, integrin β1 inhibition led to decreased FAK phosphorylation and reduced tumor cell invasion. Overexpression of S100A9 increased xenograft tumor micro-metastases, integrin β1 expression and induced NF-κB and FAK activation in vivo. Conclusion Our study demonstrated that S100A9 promotes prostate cancer cell invasion, and one of the underlying molecular mechanisms is that S100A9 activates integrin β1/FAK through TLR4/NF-κB signaling leading to metastasis of prostate cancer cell.
Collapse
Affiliation(s)
- Zhonghua Lv
- Department of Urology, Jining First People's Hospital, Jining, Shandong 272011, People's Republic of China
| | - Wenlin Li
- Department of Urology, Rizhao Traditional Chinese Medicine Hospital, Rizhao, Shandong 276800, People's Republic of China
| | - Xichao Wei
- Department of Urology, Jining Traditional Chinese Medicine Hospital, Jining, Shandong 272000, People's Republic of China
| |
Collapse
|
43
|
Baster Z, Li L, Rajfur Z, Huang C. Talin2 mediates secretion and trafficking of matrix metallopeptidase 9 during invadopodium formation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118693. [DOI: 10.1016/j.bbamcr.2020.118693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/18/2022]
|
44
|
Rangarajan ES, Primi MC, Colgan LA, Chinthalapudi K, Yasuda R, Izard T. A distinct talin2 structure directs isoform specificity in cell adhesion. J Biol Chem 2020; 295:12885-12899. [PMID: 32605925 DOI: 10.1074/jbc.ra119.010789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 06/23/2020] [Indexed: 01/25/2023] Open
Abstract
Integrin receptors regulate normal cellular processes such as signaling, cell migration, adhesion to the extracellular matrix, and leukocyte function. Talin recruitment to the membrane is necessary for its binding to and activation of integrin. Vertebrates have two highly conserved talin homologs that differ in their expression patterns. The F1-F3 FERM subdomains of cytoskeletal proteins resemble a cloverleaf, but in talin1, its F1 subdomain and additional F0 subdomain align more linearly with its F2 and F3 subdomains. Here, we present the talin2 crystal structure, revealing that its F0-F1 di-subdomain displays another unprecedented constellation, whereby the F0-F1-F2 adopts a new cloverleaf-like arrangement. Using multiangle light scattering (MALS), fluorescence lifetime imaging (FLIM), and FRET analyses, we found that substituting the corresponding residues in talin2 that abolish lipid binding in talin1 disrupt the binding of talin to the membrane, focal adhesion formation, and cell spreading. Our results provide the molecular details of the functions of specific talin isoforms in cell adhesion.
Collapse
Affiliation(s)
- Erumbi S Rangarajan
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Marina C Primi
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Lesley A Colgan
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| | - Krishna Chinthalapudi
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, Florida, USA
| | - Tina Izard
- Cell Adhesion Laboratory, Department of Integrative Structural and Computational Biology, The Scripps Research Institute, Jupiter, Florida, USA.
| |
Collapse
|
45
|
Clark DJ, Schnaubelt M, Hoti N, Hu Y, Zhou Y, Gooya M, Zhang H. Impact of Increased FUT8 Expression on the Extracellular Vesicle Proteome in Prostate Cancer Cells. J Proteome Res 2020; 19:2195-2205. [PMID: 32378902 DOI: 10.1021/acs.jproteome.9b00578] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Extracellular vesicles (EVs) are involved in intercellular communication, transporting proteins and nucleic acids to proximal and distal regions. There is evidence of glycosylation influencing protein routing into EVs; however, the impact of aberrant cellular glycotransferase expression on EV protein profiles has yet to be evaluated. In this study, we paired extracellular vesicle characterization and quantitative proteomics to determine the systemic impact of altered α(1,6)fucosyltranferase (FUT8) expression on prostate cancer-derived EVs. Our results showed that increased cellular expression of FUT8 could reduce the number of vesicles secreted by prostate cancer cells as well as increase the abundance of proteins associated with cell motility and prostate cancer metastasis. In addition, overexpression of FUT8 resulted in altered glycans on select EV-derived glycoproteins. This study presents the first evidence of altered cellular glycosylation impacting EV protein profiles and provides further rationale for exploring the functional role of glycosylation in EV biogenesis and biology.
Collapse
Affiliation(s)
- David J Clark
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Michael Schnaubelt
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Naseruddin Hoti
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Yingwei Hu
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Yangying Zhou
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Mahta Gooya
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore 21231, Maryland, United States
| |
Collapse
|
46
|
Floren M, Restrepo Cruz S, Termini CM, Marjon KD, Lidke KA, Gillette JM. Tetraspanin CD82 drives acute myeloid leukemia chemoresistance by modulating protein kinase C alpha and β1 integrin activation. Oncogene 2020; 39:3910-3925. [PMID: 32203165 PMCID: PMC7210072 DOI: 10.1038/s41388-020-1261-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
A principal challenge in treating acute myeloid leukemia (AML) is chemotherapy refractory disease. As such, there remains a critical need to identify key regulators of chemotherapy resistance in AML. In this study, we demonstrate that the membrane scaffold, CD82, contributes to the chemoresistant phenotype of AML. Using an RNA-seq approach, we identified the increased expression of the tetraspanin family member, CD82, in response to the chemotherapeutic, daunorubicin. Analysis of the TARGET and BEAT AML databases identifies a correlation between CD82 expression and overall survival of AML patients. Moreover, using a combination of cell lines and patient samples, we find that CD82 overexpression results in significantly reduced cell death in response to chemotherapy. Investigation of the mechanism by which CD82 promotes AML survival in response to chemotherapy identified a crucial role for enhanced protein kinase c alpha (PKCα) signaling and downstream activation of the β1 integrin. In addition, analysis of β1 integrin clustering by super-resolution imaging demonstrates that CD82 expression promotes the formation of dense β1 integrin membrane clusters. Lastly, evaluation of survival signaling following daunorubicin treatment identified robust activation of p38 mitogen-activated protein kinase (MAPK) downstream of PKCα and β1 integrin signaling when CD82 is overexpressed. Together, these data propose a mechanism where CD82 promotes chemoresistance by increasing PKCα activation and downstream activation/clustering of β1 integrin, leading to AML cell survival via activation of p38 MAPK. These observations suggest that the CD82-PKCα signaling axis may be a potential therapeutic target for attenuating chemoresistance signaling in AML.
Collapse
Affiliation(s)
- Muskan Floren
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Sebastian Restrepo Cruz
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Christina M Termini
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Kristopher D Marjon
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Keith A Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, USA
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.
| |
Collapse
|
47
|
Mohamed AA, El-Toukhy N, Ghaith DM, Badawy I, Abdo SM, Elkadeem M, Mahrous MN, Abd-Elsalam S. Talin-1 Gene Expression as a Tumor Marker in Hepatocellular Carcinoma Patients: A Pilot Study. THE OPEN BIOMARKERS JOURNAL 2020; 10:15-22. [DOI: 10.2174/1875318302010010015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/29/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
Background & Aims:
Hepatocellular Carcinoma (HCC) is the most common primary liver tumor. It is the second most common cancer in men and the sixth in women in Egypt. One of the proteins participating in the trans-endothelial migration is Talin-1. It also has a role in the formation and metastasis of different types of cancer. This study aimed to evaluate the diagnostic impact of Talin-1 gene expression in HCC Egyptian patients.
Methods:
Our study included forty HCC patients, thirty liver cirrhosis patients without HCC and thirty healthy subjects. For all groups, clinical and biochemical parameters were investigated. Tumor characteristics were assessed and tumor staging was done using Okuda, CLIP, VISUM and Tokyo staging systems. In addition, Serum Alpha-Fetoprotein (AFP) levels were assayed using Enzyme Immunoassay (EIA) and Talin-1 gene expression was assessed in the Peripheral Blood Mononuclear Cells (PBMCs) via quantitative real-time Polymerase Chain Reaction (PCR).
Results:
Talin-1 gene expression was significantly upregulated in HCC patients in comparison to cirrhotic and control subjects. The Receiver Operating Characteristic (ROC) analysis indicated that Talin-1 gene expression surpasses serum levels of AFP in the diagnosis of HCC. In particular, the cut off value of 9.5 (2-∆∆Ct) recorded an AUC of 85.7% with a sensitivity of 93.3% and specificity of 80%.
Conclusion:
Our data confirmed an évident diagnostic role of Talin-1 gene expression for HCC detection.
Collapse
|
48
|
Long Noncoding RNA Lnc-TLN2-4:1 Suppresses Gastric Cancer Metastasis and Is Associated with Patient Survival. JOURNAL OF ONCOLOGY 2020; 2020:8681361. [PMID: 32256587 PMCID: PMC7086451 DOI: 10.1155/2020/8681361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/30/2020] [Accepted: 02/08/2020] [Indexed: 01/23/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide, and the tumor metastasis leads to poor outcomes of GC patients. Long noncoding RNAs (lncRNAs) have emerged as new regulatory molecules that play a crucial role in tumor metastasis. However, the biological function and underlying mechanism of numerous lncRNAs in GC metastasis remain largely unclear. Here, we report a novel lncRNA, lnc-TLN2-4:1, whose expression is decreased in GC tissue versus matched normal tissue, and its low expression is involved in the lymph node and distant metastases of GC, as well as poor overall survival rates of GC patients. We further found that lnc-TLN2-4:1 inhibits the ability of GC cells to migrate and invade but does not influence GC cell proliferation and confirmed that lnc-TLN2-4:1 is mainly located in the cytoplasm of GC cells. We then found that lnc-TLN2-4:1 increases the mRNA and protein expression of TLN2 in GC cells and there is a positive correlation between the expression of lnc-TLN2-4:1 and TLN2 mRNA in GC tissue. Collectively, we identified a novel lncRNA, lnc-TLN2-4:1, in GC, where lnc-TLN2-4:1 represses cell migration and invasion. The low expression of lnc-TLN2-4:1 is associated with poor overall survival rates of GC patients. These suggest that lnc-TLN2-4:1 may be a tumor suppressor during GC metastasis.
Collapse
|
49
|
Talin1 regulates the endometrial epithelial cell adhesive capacity by interacting with LASP1 and Vitronectin. Reprod Biol 2020; 20:229-236. [PMID: 32113856 DOI: 10.1016/j.repbio.2020.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/27/2020] [Accepted: 02/14/2020] [Indexed: 01/13/2023]
Abstract
The endometrium is a highly complex tissue that is vulnerable to subtle gene expression changes and is the first point of contact for an implanting blastocyst. Talin1 has previously been identified to regulate cytoskeleton and cell motility, however it has not been investigated in association with infertility. Herein, we presented that Talin1 dysregulation in the missed abortion endometrium would negatively influence endometrial adhesive capacity. Mechanistically, intracellular Talin1 inhibited the nuclear transportation of LIM and SH3 protein 1 (LASP1) and restored the expression of adhesion-associated protein. Moreover, extracellular Talin1 enforces endometrial epithelial cell adhesive capacity by interacting with Vitronectin (VTN) and activating the FAK/Src/ERK signalling pathway. This finding provides a novel insight into the potential use of Talin1 for managing endometrial epithelia cell adhesion. This study represents the first demonstration of Talin1 function in endometrial epithelial cell adhesion and endometrial receptivity. Our findings indicate that re-expression of Talin1 might represent a useful strategy for preventing and treating early pregnancy failure and infertility.
Collapse
|
50
|
Baster Z, Li L, Kukkurainen S, Chen J, Pentikäinen O, Győrffy B, Hytönen VP, Zhu H, Rajfur Z, Huang C. Cyanidin-3-glucoside binds to talin and modulates colon cancer cell adhesions and 3D growth. FASEB J 2020; 34:2227-2237. [PMID: 31916632 DOI: 10.1096/fj.201900945r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/21/2019] [Accepted: 06/25/2019] [Indexed: 12/20/2022]
Abstract
Cyanidin-3-glucoside (C3G) is a natural pigment, found in many colorful fruits and vegetables. It has many health benefits, including anti-inflammation, cancer prevention, and anti-diabetes. Although C3G is assumed to be an antioxidant, it has been reported to affect cell-matrix adhesions. However, the underlying molecular mechanism is unknown. Here, we show that the expression of talin1, a key regulator of integrins and cell adhesions, negatively correlated with the survival rate of colon cancer patients and that depletion of talin1 inhibited 3D spheroid growth in colon cancer cells. Interestingly, C3G bound to talin and promoted the interaction of talin with β1A-integrin. Molecular docking analysis shows that C3G binds to the interface of the talin-β-integrin complex, acting as an allosteric regulator and altering the interaction between talin and integrin. Moreover, C3G promoted colon cancer cell attachment to fibronectin. While C3G had no significant effect on colon cancer cell proliferation, it significantly inhibited 3D spheroid growth in fibrin gel assays. Since C3G has no or very low toxicity, it could be potentially used for colon cancer prevention or therapy.
Collapse
Affiliation(s)
- Zbigniew Baster
- Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.,Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Liqing Li
- Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Sampo Kukkurainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, FL, USA
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Olli Pentikäinen
- Institute of Biomedicine, Integrative Physiology and Pharmacology, University of Turku, Turku, FL, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, FL, USA.,Fimlab Laboratories, Tampere, FL, USA
| | - Haining Zhu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Zenon Rajfur
- Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Cai Huang
- Markey Cancer Center, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|