1
|
Ke ZB, Chen JY, Xue YT, Lin B, Huang Q, Huang XY, Chen DN, Chen SH, Ye XJ, Zheng QS, Wei Y, Xue XY, Xu N. Mechanical signal modulates prostate cancer immune escape by USP8-mediated ubiquitination-dependent degradation of PD-L1 and MHC-1. Cell Death Dis 2025; 16:413. [PMID: 40410130 PMCID: PMC12102395 DOI: 10.1038/s41419-025-07736-4] [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: 09/14/2024] [Revised: 04/30/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
The tumor environment of prostate cancer (PCa) tissues of high Gleason score has been proved to be more immune suppressive and has higher extracellular matrix (ECM) stiffness, but whether ECM mechanical stiffness is the cause of higher ability of invasiveness and immune escape of PCa with high Gleason score remains uncertain. In this study, we showed that higher polyacrylamide hydrogels (PAAG) stiffness resulted in the progression and immune escape of PCa via integrin β1/FAK/YAP axis. The translocation of YAP into cell nucleus to bind to TEAD2 promoted the transcriptional activation of USP8. NBR1 could be ubiquitinated, and then degraded, via interacting with P62/SQSTM1 and through autophagy-lysosome pathway. Increased expression of USP8 promoted the abundance of NBR1 via K63-linked de-ubiquitination and PD-L1 via K48-linked de-ubiquitination in response to high PAAG stiffness. NBR1-mediated selective autophagy accelerated the degradation of MHC-1 of PCa. The USP8 inhibitor presented a potential application value in sensitizing immunotherapy of PCa. Taken together, we identified a USP8-mediated de-ubiquitination mechanism that involves in the process of high PAAG stiffness-mediated high expression of PD-L1 and low expression of MHC-1 of PCa cells, which provided a rationale of immunotherapy sensitization of PCa via USP8 inhibition.
Collapse
Affiliation(s)
- Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jia-Yin Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Bin Lin
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qi Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xu-Yun Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Jian Ye
- Department of Ultrasonography, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
2
|
Moreira IB, Buettner FFR. Glycosphingolipids as emerging attack points in bladder cancer. Discov Oncol 2025; 16:569. [PMID: 40252176 PMCID: PMC12009261 DOI: 10.1007/s12672-025-02302-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/02/2025] [Indexed: 04/21/2025] Open
Abstract
Bladder cancer is a prevalent malignancy associated with significant morbidity and mortality worldwide. Emerging research highlights the critical role of glycosphingolipids (GSLs) in bladder cancer progression. In this review, we examine GSL expression profiles in bladder cancer and explore their contributions to key cancer hallmarks, including invasion and metastasis, immune evasion, and resistance to cell death. We further discuss the potential of GSLs as therapeutic targets and non-invasive biomarkers, with an emphasis on recent advances in GSL-targeting strategies. Additionally, we highlight our recent discovery of a novel, patented biomarker for bladder cancer diagnosis, identified using cutting-edge glyco-analytical technologies.
Collapse
Affiliation(s)
- Inês B Moreira
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | - Falk F R Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany.
- Proteomics, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Universitätsstrasse 2, 86159, Augsburg, Germany.
| |
Collapse
|
3
|
Sun X, Hu X. Unveiling Matrix Metalloproteinase 13's Dynamic Role in Breast Cancer: A Link to Physical Changes and Prognostic Modulation. Int J Mol Sci 2025; 26:3083. [PMID: 40243781 PMCID: PMC11988641 DOI: 10.3390/ijms26073083] [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: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The biomechanical properties of the extracellular matrix (ECM) including its stiffness, viscoelasticity, collagen architecture, and temperature constitute critical biomechanical cues governing breast cancer progression. Matrix metalloproteinase 13 (MMP13) is an important marker of breast cancer and plays important roles in matrix remodelling and cell metastasis. Emerging evidence highlights MMP13 as a dynamic modulator of the ECM's physical characteristics through dual mechanoregulatory mechanisms. While MMP13-mediated collagen degradation facilitates microenvironmental softening, thus promoting tumour cell invasion, paradoxically, its crosstalk with cancer-associated fibroblasts (CAFs) and tumour-associated macrophages (TAMs) drives pathological stromal stiffening via aberrant matrix deposition and crosslinking. This biomechanical duality is amplified through feedforward loops with an epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) populations, mediated by signalling axes such as TGF-β/Runx2. Intriguingly, MMP13 exhibits context-dependent mechanomodulatory effects, demonstrating anti-fibrotic activity and inhibiting the metastasis of breast cancer. At the same time, angiogenesis and increased metabolism are important mechanisms through which MMP13 promotes a temperature increase in breast cancer. Targeting the spatiotemporal regulation of MMP13's mechanobiological functions may offer novel therapeutic strategies for disrupting the tumour-stroma vicious cycle.
Collapse
Affiliation(s)
- Xiaomeng Sun
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
| | - Xiaojuan Hu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China;
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Xuefu Avenue, Honggutan District, Nanchang 330031, China
| |
Collapse
|
4
|
Ishihara S, Enomoto A, Sakai A, Iida T, Tange S, Kioka N, Nukuda A, Nagasato AI, Yasuda M, Tokino T, Haga H. Stiff extracellular matrix activates the transcription factor ATF5 to promote the proliferation of cancer cells. iScience 2025; 28:112057. [PMID: 40124511 PMCID: PMC11928855 DOI: 10.1016/j.isci.2025.112057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/20/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer tissues are stiffer than normal tissues. Carcinogenesis stiffens the extracellular matrix (ECM) of cancerous tissues, to which cancer cells respond by activating transcription factors, such as YAP/TAZ, Twist1, and β-catenin, which further elevate their malignancy. However, these transcription factors are also expressed in normal tissues. Therefore, inhibiting these factors in order to treat cancer may lead to severe side effects. Here, we show that activating transcription factor 5 (ATF5), highly expressed in tumors, is activated by ECM stiffness and promotes the proliferation of cancer cells, including that of pancreatic cancer cells and lung cancer cells. In addition, ATF5 suppressed the expression of early growth response 1 (EGR1), thereby accelerating cancer cell proliferation. Stiff ECMs trigger the JAK-MYC pathway which activates ATF5. JAK activation was actomyosin independent whereas MYC induction was actomyosin dependent. These results demonstrate the critical role played by ATF5 in the mechanotransduction process seen in cancers.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Akihiro Sakai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550 Japan
| | - Shoichiro Tange
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Akihiro Nukuda
- Transdisciplinary Life Science Course, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810 Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Motoaki Yasuda
- Department of Oral Molecular Microbiology, Division of Oral Pathobiological Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido 060-8586, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| |
Collapse
|
5
|
Saadh MJ, Ahmed HH, Kareem RA, Bishoyi AK, Roopashree R, Shit D, Arya R, Sharma A, Khaitov K, Sameer HN, Yaseen A, Athab ZH, Adil M. Molecular mechanisms of Hippo pathway in tumorigenesis: therapeutic implications. Mol Biol Rep 2025; 52:267. [PMID: 40014178 DOI: 10.1007/s11033-025-10372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
The Hippo signaling pathway is a pivotal regulator of tissue homeostasis, organ size, and cell proliferation. Its dysregulation is profoundly implicated in various forms of cancer, making it a highly promising target for therapeutic intervention. This review extensively evaluates the mechanisms underlying the dysregulation of the Hippo pathway in cancer cells and the molecular processes linking these alterations to tumorigenesis. Under normal physiological conditions, the Hippo pathway is a guardian, ensuring controlled cellular proliferation and programmed cell death. However, numerous mutations and epigenetic modifications can disrupt this equilibrium in cancer cells, leading to unchecked cell proliferation, enhanced survival, and metastatic capabilities. The pathway's interaction with other critical signaling networks, including Wnt/β-catenin, PI3K/Akt, TGF-β/SMAD, and EGFR pathways, further amplifies its oncogenic potential. Central to these disruptions is the activation of YAP and TAZ transcriptional coactivators, which drive the expression of genes that promote oncogenesis. This review delves into the molecular mechanisms responsible for the dysregulation of the Hippo pathway in cancer, elucidating how these disruptions contribute to tumorigenesis. We also explore potential therapeutic strategies, including inhibitors targeting YAP/TAZ activity and modulators of upstream signaling components. Despite significant advancements in understanding the Hippo pathway's role in cancer, numerous questions remain unresolved. Continued research is imperative to unravel the complex interactions within this pathway and to develop innovative and effective therapies for clinical application. In conclusion, the comprehensive understanding of the Hippo pathway's regulatory mechanisms offers significant potential for advancing cancer therapies, regenerative medicine, and treatments for chronic diseases. The translation of these insights into clinical practice will necessitate collaborative efforts from researchers, clinicians, and pharmaceutical developers to bring novel and effective therapies to patients, ultimately improving clinical outcomes and advancing the field of oncology.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Abhishek Sharma
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Kakhramon Khaitov
- Department of Dermatovenerology, Pediatric Dermatovenerology and AIDS, Tashkent Pediatric Medical Institute, Bogishamol Street 223, Tashkent, 100140, Uzbekistan
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
6
|
Huang T, Fan L, Tang J, Chen S, Du G, Zhang N. Advances in research on the carcinogenic mechanisms and therapeutic potential of YAP1 in bladder cancer (Review). Oncol Rep 2025; 53:10. [PMID: 39540392 PMCID: PMC11599795 DOI: 10.3892/or.2024.8843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Bladder cancer is the most common malignant tumor of the urinary system with high morbidity and no clear pathogenesis. The Hippo signaling pathway is an evolutionarily conserved pathway that regulates organ size and maintains tissue homeostasis. Yes‑associated protein 1 (YAP1) is a key effector of this pathway and regulates downstream target genes by binding to transcriptional co‑activators with PDZ binding sequences (TAZ). Several studies have demonstrated that YAP1 is overexpressed in bladder cancer and is involved in adverse outcomes such as bladder cancer occurrence, progression, resistance to cisplatin and the recurrence of tumours. The present review summarized the involvement of YAP1 in bladder cancer disease onset and progression, and the mechanism of YAP1 involvement in bladder cancer treatment. In addition, this study further explored the potential of YAP1 in the diagnosis and treatment of bladder cancer. This study aimed to explore the potential mechanism of YAP1 in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Tianyu Huang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Longmei Fan
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiajia Tang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Shicheng Chen
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guotu Du
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Neng Zhang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
7
|
Berdiaki A, Neagu M, Tzanakakis P, Spyridaki I, Pérez S, Nikitovic D. Extracellular Matrix Components and Mechanosensing Pathways in Health and Disease. Biomolecules 2024; 14:1186. [PMID: 39334952 PMCID: PMC11430160 DOI: 10.3390/biom14091186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Glycosaminoglycans (GAGs) and proteoglycans (PGs) are essential components of the extracellular matrix (ECM) with pivotal roles in cellular mechanosensing pathways. GAGs, such as heparan sulfate (HS) and chondroitin sulfate (CS), interact with various cell surface receptors, including integrins and receptor tyrosine kinases, to modulate cellular responses to mechanical stimuli. PGs, comprising a core protein with covalently attached GAG chains, serve as dynamic regulators of tissue mechanics and cell behavior, thereby playing a crucial role in maintaining tissue homeostasis. Dysregulation of GAG/PG-mediated mechanosensing pathways is implicated in numerous pathological conditions, including cancer and inflammation. Understanding the intricate mechanisms by which GAGs and PGs modulate cellular responses to mechanical forces holds promise for developing novel therapeutic strategies targeting mechanotransduction pathways in disease. This comprehensive overview underscores the importance of GAGs and PGs as key mediators of mechanosensing in maintaining tissue homeostasis and their potential as therapeutic targets for mitigating mechano-driven pathologies, focusing on cancer and inflammation.
Collapse
Affiliation(s)
- Aikaterini Berdiaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Monica Neagu
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
| | - Petros Tzanakakis
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Ioanna Spyridaki
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| | - Serge Pérez
- Centre de Recherche sur les Macromolécules Végétales (CERMAV), Centre National de la Recherche Scientifique (CNRS), University Grenoble Alpes, 38000 Grenoble, France;
| | - Dragana Nikitovic
- Department of Histology-Embryology, Medical School, University of Crete, 712 03 Heraklion, Greece; (A.B.); (P.T.); (I.S.)
| |
Collapse
|
8
|
Tong T, Huang M, Yan B, Lin B, Yu J, Teng Q, Li P, Pang J. Hippo signaling modulation and its biological implications in urological malignancies. Mol Aspects Med 2024; 98:101280. [PMID: 38870717 DOI: 10.1016/j.mam.2024.101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/15/2024]
Abstract
Although cancer diagnosis and treatment have rapidly advanced in recent decades, urological malignancies, which have high morbidity and mortality rates, are among the most difficult diseases to treat. The Hippo signaling is an evolutionarily conserved pathway in organ size control and tissue homeostasis maintenance. Its downstream effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are key modulators of numerous physiological and pathological processes. Recent work clearly indicates that Hippo signaling is frequently altered in human urological malignancies. In this review, we discuss the disparate viewpoints on the upstream regulators of YAP/TAZ and their downstream targets and systematically summarize the biological implications. More importantly, we highlight the molecular mechanisms involved in Hippo-YAP signaling to improve our understanding of its role in every stage of prostate cancer, bladder cancer and kidney cancer progression. A better understanding of the biological outcomes of YAP/TAZ modulation will contribute to the establishment of future therapeutic approaches.
Collapse
Affiliation(s)
- Tongyu Tong
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Mengjun Huang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Binyuan Yan
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Bingbiao Lin
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, Guangdong, 515041, China
| | - Jiaying Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Qiliang Teng
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China; Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Jun Pang
- Department of Urology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
9
|
Haderk F, Chou YT, Cech L, Fernández-Méndez C, Yu J, Olivas V, Meraz IM, Barbosa Rabago D, Kerr DL, Gomez C, Allegakoen DV, Guan J, Shah KN, Herrington KA, Gbenedio OM, Nanjo S, Majidi M, Tamaki W, Pourmoghadam YK, Rotow JK, McCoach CE, Riess JW, Gutkind JS, Tang TT, Post L, Huang B, Santisteban P, Goodarzi H, Bandyopadhyay S, Kuo CJ, Roose JP, Wu W, Blakely CM, Roth JA, Bivona TG. Focal adhesion kinase-YAP signaling axis drives drug-tolerant persister cells and residual disease in lung cancer. Nat Commun 2024; 15:3741. [PMID: 38702301 PMCID: PMC11068778 DOI: 10.1038/s41467-024-47423-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/18/2024] [Indexed: 05/06/2024] Open
Abstract
Targeted therapy is effective in many tumor types including lung cancer, the leading cause of cancer mortality. Paradigm defining examples are targeted therapies directed against non-small cell lung cancer (NSCLC) subtypes with oncogenic alterations in EGFR, ALK and KRAS. The success of targeted therapy is limited by drug-tolerant persister cells (DTPs) which withstand and adapt to treatment and comprise the residual disease state that is typical during treatment with clinical targeted therapies. Here, we integrate studies in patient-derived and immunocompetent lung cancer models and clinical specimens obtained from patients on targeted therapy to uncover a focal adhesion kinase (FAK)-YAP signaling axis that promotes residual disease during oncogenic EGFR-, ALK-, and KRAS-targeted therapies. FAK-YAP signaling inhibition combined with the primary targeted therapy suppressed residual drug-tolerant cells and enhanced tumor responses. This study unveils a FAK-YAP signaling module that promotes residual disease in lung cancer and mechanism-based therapeutic strategies to improve tumor response.
Collapse
Affiliation(s)
- Franziska Haderk
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Yu-Ting Chou
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Cech
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científícas (CSIC) y Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Johnny Yu
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Victor Olivas
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Ismail M Meraz
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dora Barbosa Rabago
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - D Lucas Kerr
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos Gomez
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - David V Allegakoen
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Juan Guan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Khyati N Shah
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kari A Herrington
- Center for Advanced Light Microscopy, University of California, San Francisco, San Francisco, CA, USA
| | | | - Shigeki Nanjo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Mourad Majidi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Whitney Tamaki
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yashar K Pourmoghadam
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Julia K Rotow
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Caroline E McCoach
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan W Riess
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - J Silvio Gutkind
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Tracy T Tang
- Vivace Therapeutics, Inc., 1500 Fashion Island Blvd., Suite 102, San Mateo, CA, USA
| | - Leonard Post
- Vivace Therapeutics, Inc., 1500 Fashion Island Blvd., Suite 102, San Mateo, CA, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científícas (CSIC) y Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Hani Goodarzi
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Sourav Bandyopadhyay
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeroen P Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Wei Wu
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Rainu SK, Singh N. Dual-Sensitive Fluorescent Nanoprobes for Simultaneously Monitoring In Situ Changes in pH and Matrix Metalloproteinase Expression in Stiffness-Tunable Three-Dimensional In Vitro Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12175-12187. [PMID: 38420964 DOI: 10.1021/acsami.3c16334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
A tumor microenvironment often presents altered physicochemical characteristics of the extracellular matrix (ECM) including changes in matrix composition, stiffness, protein expression, pH, temperature, or the presence of certain stromal and immune cells. Of these, overexpression of matrix metalloproteinases (MMPs) and extracellular acidosis are the two major hallmarks of cancer that can be exploited for tumor detection. The change in matrix stiffness and the release of certain cytokines (TNF-α) in the tumor microenvironment play major roles in inducing MMP-9 expression in cancerous cells. This study highlights the role of mechanical cues in upregulating MMP-9 expression in cancerous cells using stiffness-tunable matrix compositions and dual-sensitive fluorescent nanoprobes. Ionically cross-linked 3D alginate/gelatin (AG) scaffolds with three stiffnesses were chosen to reflect the ECM stiffnesses corresponding to healthy and pathological tissues. Moreover, a dual-sensitive nanoprobe, an MMP-sensitive peptide conjugated to carbon nanoparticles with intrinsic pH fluorescence properties, was utilized for in situ monitoring of the two cancer hallmarks in the 3D scaffolds. This platform was further utilized for designing a 3D core-shell platform for spatially mapping tumor margins and for visualizing TNF-α-induced MMP-9 expression in cancerous cells.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
11
|
Shu J, Deng H, Zhang Y, Wu F, He J. Cancer cell response to extrinsic and intrinsic mechanical cue: opportunities for tumor apoptosis strategies. Regen Biomater 2024; 11:rbae016. [PMID: 38476678 PMCID: PMC10932484 DOI: 10.1093/rb/rbae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024] Open
Abstract
Increasing studies have revealed the importance of mechanical cues in tumor progression, invasiveness and drug resistance. During malignant transformation, changes manifest in either the mechanical properties of the tissue or the cellular ability to sense and respond to mechanical signals. The major focus of the review is the subtle correlation between mechanical cues and apoptosis in tumor cells from a mechanobiology perspective. To begin, we focus on the intracellular force, examining the mechanical properties of the cell interior, and outlining the role that the cytoskeleton and intracellular organelle-mediated intracellular forces play in tumor cell apoptosis. This article also elucidates the mechanisms by which extracellular forces guide tumor cell mechanosensing, ultimately triggering the activation of the mechanotransduction pathway and impacting tumor cell apoptosis. Finally, a comprehensive examination of the present status of the design and development of anti-cancer materials targeting mechanotransduction is presented, emphasizing the underlying design principles. Furthermore, the article underscores the need to address several unresolved inquiries to enhance our comprehension of cancer therapeutics that target mechanotransduction.
Collapse
Affiliation(s)
- Jun Shu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Huan Deng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Yu Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, PR China
| | - Fang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
12
|
Kalwa PL, Walz S, Granai M, Fend F, Stenzl A, Schäffer TE. Differentiation of bladder cancer with water flow elastography (WaFE). J Mech Behav Biomed Mater 2024; 150:106319. [PMID: 38142569 DOI: 10.1016/j.jmbbm.2023.106319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023]
Abstract
Cancer affects the mechanical properties of tissue. Therefore, elastography techniques can be used to differentiate cancerous from healthy tissue. Due to probe size and restricted handling, most elastography techniques are not applicable in minimally invasive surgery (MIS). Established techniques such as endoscopic ultrasound elastography measure under undefined boundary conditions, making the determination of quantitative mechanical properties challenging. Water flow elastography (WaFE) has recently been introduced for application in MIS. Here, we present an improved WaFE measurement method in which the probe attaches itself to the sample with a small suction pressure. This leads to defined boundary conditions, allowing for a quantitative determination of the Young's modulus of tissue. To facilitate fast measurements, we developed a correction model for the hydrodynamic resistance and the fluid inertia of the tubing. We used WaFE for ex vivo measurements on human bladders and found a significantly larger Young's modulus for cancerous vs. healthy tissue. We determined the optimal classification threshold for the Young's modulus to be 8 kPa and found that WaFE can differentiate between cancerous and healthy tissue with a sensitivity of 0.96 and a specificity of 1. Our results underline that WaFE can be a helpful differentiating tool in MIS.
Collapse
Affiliation(s)
- Paul L Kalwa
- Institute of Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany
| | - Simon Walz
- Department of Urology, University of Tübingen Medical Center, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Massimo Granai
- Institute of Pathology and Neuropathology, University Hospital of Tübingen, Liebermeisterstr. 8, 72076 Tübingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital of Tübingen, Liebermeisterstr. 8, 72076 Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen Medical Center, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| |
Collapse
|
13
|
Sleeboom JJF, van Tienderen GS, Schenke-Layland K, van der Laan LJW, Khalil AA, Verstegen MMA. The extracellular matrix as hallmark of cancer and metastasis: From biomechanics to therapeutic targets. Sci Transl Med 2024; 16:eadg3840. [PMID: 38170791 DOI: 10.1126/scitranslmed.adg3840] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
The extracellular matrix (ECM) is essential for cell support during homeostasis and plays a critical role in cancer. Although research often concentrates on the tumor's cellular aspect, attention is growing for the importance of the cancer-associated ECM. Biochemical and physical ECM signals affect tumor formation, invasion, metastasis, and therapy resistance. Examining the tumor microenvironment uncovers intricate ECM dysregulation and interactions with cancer and stromal cells. Anticancer therapies targeting ECM sensors and remodelers, including integrins and matrix metalloproteinases, and ECM-remodeling cells, have seen limited success. This review explores the ECM's role in cancer and discusses potential therapeutic strategies for cell-ECM interactions.
Collapse
Affiliation(s)
- Jelle J F Sleeboom
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Postbox 2040, 3000CA Rotterdam, Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, Mekelweg 2, 2628CD Delft, Netherlands
| | - Gilles S van Tienderen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Postbox 2040, 3000CA Rotterdam, Netherlands
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University Tübingen, 72770 Reutlingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Postbox 2040, 3000CA Rotterdam, Netherlands
| | - Antoine A Khalil
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Postbox 2040, 3000CA Rotterdam, Netherlands
| |
Collapse
|
14
|
Liang R, Song G. Matrix stiffness-driven cancer progression and the targeted therapeutic strategy. MECHANOBIOLOGY IN MEDICINE 2023; 1:100013. [PMID: 40395641 PMCID: PMC12082158 DOI: 10.1016/j.mbm.2023.100013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 05/22/2025]
Abstract
Increased matrix stiffness is a common phenomenon in solid tumor tissue and is regulated by both tumor and mesenchymal cells. The increase in collagen and lysyl oxidase family proteins in the extracellular matrix leads to deposition, contraction, and crosslinking of the stroma, promoting increased matrix stiffness in tumors. Matrix stiffness is critical to the progression of various solid tumors. As a mechanical factor in the tumor microenvironment, matrix stiffness is involved in tumor progression, promoting biological processes such as tumor cell proliferation, invasion, metastasis, angiogenesis, drug resistance, and immune escape. Reducing tissue stiffness can slow down tumor progression. Therefore targeting matrix stiffness is a potential option for tumor therapy. This article reviews the detailed mechanisms of matrix stiffness in different malignant tumor phenotypes and potential tumor therapies targeting matrix stiffness. Understanding the role and mechanisms of matrix stiffness in tumors could provide theoretical insights into the treatment of tumors and assist in the clinical development of new drug therapies.
Collapse
Affiliation(s)
- Rui Liang
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing 400030, China
| |
Collapse
|
15
|
Weston WA, Barr AR. A cell cycle centric view of tumour dormancy. Br J Cancer 2023; 129:1535-1545. [PMID: 37608096 PMCID: PMC10645753 DOI: 10.1038/s41416-023-02401-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
Tumour dormancy and recurrent metastatic cancer remain the greatest clinical challenge for cancer patients. Dormant tumour cells can evade treatment and detection, while retaining proliferative potential, often for years, before relapsing to tumour outgrowth. Cellular quiescence is one mechanism that promotes and maintains tumour dormancy due to its central role in reducing proliferation, elevating cyto-protective mechanisms, and retaining proliferative potential. Quiescence/proliferation decisions are dictated by intrinsic and extrinsic signals, which regulate the activity of cyclin-dependent kinases (CDKs) to modulate cell cycle gene expression. By clarifying the pathways regulating CDK activity and the signals which activate them, we can better understand how cancer cells enter, maintain, and escape from quiescence throughout the progression of dormancy and metastatic disease. Here we review how CDK activity is regulated to modulate cellular quiescence in the context of tumour dormancy and highlight the therapeutic challenges and opportunities it presents.
Collapse
Affiliation(s)
- William A Weston
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Alexis R Barr
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Imperial College London, Du Cane Rd, London, W12 0NN, UK.
| |
Collapse
|
16
|
Safaei S, Sajed R, Shariftabrizi A, Dorafshan S, Saeednejad Zanjani L, Dehghan Manshadi M, Madjd Z, Ghods R. Tumor matrix stiffness provides fertile soil for cancer stem cells. Cancer Cell Int 2023; 23:143. [PMID: 37468874 PMCID: PMC10357884 DOI: 10.1186/s12935-023-02992-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
Matrix stiffness is a mechanical characteristic of the extracellular matrix (ECM) that increases from the tumor core to the tumor periphery in a gradient pattern in a variety of solid tumors and can promote proliferation, invasion, metastasis, drug resistance, and recurrence. Cancer stem cells (CSCs) are a rare subpopulation of tumor cells with self-renewal, asymmetric cell division, and differentiation capabilities. CSCs are thought to be responsible for metastasis, tumor recurrence, chemotherapy resistance, and consequently poor clinical outcomes. Evidence suggests that matrix stiffness can activate receptors and mechanosensor/mechanoregulator proteins such as integrin, FAK, and YAP, modulating the characteristics of tumor cells as well as CSCs through different molecular signaling pathways. A deeper understanding of the effect of matrix stiffness on CSCs characteristics could lead to development of innovative cancer therapies. In this review, we discuss how the stiffness of the ECM is sensed by the cells and how the cells respond to this environmental change as well as the effect of matrix stiffness on CSCs characteristics and also the key malignant processes such as proliferation and EMT. Then, we specifically focus on how increased matrix stiffness affects CSCs in breast, lung, liver, pancreatic, and colorectal cancers. We also discuss how the molecules responsible for increased matrix stiffness and the signaling pathways activated by the enhanced stiffness can be manipulated as a therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Sadegh Safaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Ahmad Shariftabrizi
- Division of Nuclear Medicine, Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Division of Nuclear Medicine, Department of Radiology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shima Dorafshan
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Department of Pathology and Genomic Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Masoumeh Dehghan Manshadi
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14530, Iran.
| |
Collapse
|
17
|
Park S, Chien AL, Brown ID, Chen J. Characterizing viscoelastic properties of human melanoma tissue using Prony series. Front Bioeng Biotechnol 2023; 11:1162880. [PMID: 37091343 PMCID: PMC10117758 DOI: 10.3389/fbioe.2023.1162880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
Melanoma is the most invasive and deadly skin cancer, which causes most of the deaths from skin cancer. It has been demonstrated that the mechanical properties of tumor tissue are significantly altered. However, data about characterizing the mechanical properties of in vivo melanoma tissue are extremely scarce. In addition, the viscoelastic or viscous properties of melanoma tissue are rarely reported. In this study, we measured and quantitated the viscoelastic properties of human melanoma tissues based on the stress relaxation test, using the indentation-based mechanical analyzer that we developed previously. The melanoma tissues from eight patients of different ages (57–95), genders (male and female patients), races (White and Asian), and sites (nose, arm, shoulder, and chest) were excised and tested. The results showed that the elastic property (i.e., shear modulus) of melanoma tissue was elevated compared to normal tissue, while the viscous property (i.e., relaxation time) was reduced. Moreover, the tissue thickness had a significant impact on the viscoelastic properties, probably due to the amount of the adipose layer. Our findings provide new insights into the role of the viscous and elastic properties of melanoma cell mechanics, which may be implicated in the disease state and progression.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical Engineering, University of Nevada, Las Vegas, NV, United States
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Seungman Park,
| | - Anna L. Chien
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Isabelle D. Brown
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, NV, United States
| |
Collapse
|
18
|
Qiwei C, Jiajun S, Cheng L, Shengbo H, Yue K, Shujing W, Liu W, Xinqing Z, Hongyu W, Deyong Y. Comparison between renal pelvic and ureteral tumors in muscle-invasive upper tract urothelial carcinoma. Cancer Sci 2023; 114:984-994. [PMID: 36330561 PMCID: PMC9986090 DOI: 10.1111/cas.15634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Although renal pelvic and ureteral urothelial carcinoma share similarities in their origins, disparities on a genetic and clinical level make them divergent entities. Clinical information from the Surveillance, Epidemiology, and End Results (SEER) database was used to validate the characteristics and molecular subtypes using single-center data, which were compared between the two types of muscle-invasive tumors. Simultaneously, to expand the sample size for further verification, we explored a deep learning algorithm to correctly classify molecular subtypes from H&E histology slides. We suggested that the renal pelvic group might have a proclivity towards luminal and the ureter towards basal and P53-like. Furthermore, we explore the heterogeneity of matrix and immune tumor microenvironment, and the ureteral group had more immune cell infiltration and higher stiffness. Collectively, these results showed that muscle-invasive upper tract urothelial carcinoma exist in distinct properties of clinical characteristics, molecular subtype, and tumor microenvironment.
Collapse
Affiliation(s)
- Chen Qiwei
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shi Jiajun
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Huang Shengbo
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, China
| | - Kuai Yue
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, China
| | - Wang Shujing
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, China
| | - Wenlong Liu
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, China
| | - Zhu Xinqing
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wang Hongyu
- School of Information and Communication Engineering, Dalian University of Technology, Dalian, China
| | - Yang Deyong
- Department of Urology, First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Martinez-Vidal L, Chighizola M, Berardi M, Alchera E, Locatelli I, Pederzoli F, Venegoni C, Lucianò R, Milani P, Bielawski K, Salonia A, Podestà A, Alfano M. Micro-mechanical fingerprints of the rat bladder change in actinic cystitis and tumor presence. Commun Biol 2023; 6:217. [PMID: 36823431 PMCID: PMC9950451 DOI: 10.1038/s42003-023-04572-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Tissue mechanics determines tissue homeostasis, disease development and progression. Bladder strongly relies on its mechanical properties to perform its physiological function, but these are poorly unveiled under normal and pathological conditions. Here we characterize the mechanical fingerprints at the micro-scale level of the three tissue layers which compose the healthy bladder wall, and identify modifications associated with the onset and progression of pathological conditions (i.e., actinic cystitis and bladder cancer). We use two indentation-based instruments (an Atomic Force Microscope and a nanoindenter) and compare the micromechanical maps with a comprehensive histological analysis. We find that the healthy bladder wall is a mechanically inhomogeneous tissue, with a gradient of increasing stiffness from the urothelium to the lamina propria, which gradually decreases when reaching the muscle outer layer. Stiffening in fibrotic tissues correlate with increased deposition of dense extracellular matrix in the lamina propria. An increase in tissue compliance is observed before the onset and invasion of the tumor. By providing high resolution micromechanical investigation of each tissue layer of the bladder, we depict the intrinsic mechanical heterogeneity of the layers of a healthy bladder as compared with the mechanical properties alterations associated with either actinic cystitis or bladder tumor.
Collapse
Affiliation(s)
- Laura Martinez-Vidal
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - M Chighizola
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy
| | - M Berardi
- Optics11, Amsterdam, The Netherlands
- LaserLab, Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands
| | - E Alchera
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - I Locatelli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - F Pederzoli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - C Venegoni
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - R Lucianò
- Pathology Unit, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - P Milani
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy
| | | | - A Salonia
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - A Podestà
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy.
| | - M Alfano
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy.
| |
Collapse
|
20
|
Monteiro-Reis S, Ferreira JPS, Pires RA, Lobo J, Carvalho JA, Reis RL, Jorge RN, Jerónimo C. Bladder Wall Stiffness after Cystectomy in Bladder Cancer Patients: A Preliminary Study. Cancers (Basel) 2023; 15:359. [PMID: 36672309 PMCID: PMC9856862 DOI: 10.3390/cancers15020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 01/08/2023] Open
Abstract
Bladder cancer (BlCa), specifically urothelial carcinomas, is a heterogeneous disease that derives from the urothelial lining. Two main classes of BlCa are acknowledged: the non-muscle invasive BlCa and the muscle-invasive BlCa; the latter constituting an aggressive disease which invades locally and metastasizes systemically. Distinguishing the specific microenvironment that cancer cells experience between mucosa and muscularis propria layers can help elucidate how these cells acquire invasive capacities. In this work, we propose to measure the micromechanical properties of both mucosa and muscularis propria layers of the bladder wall of BlCa patients, using atomic force microscopy (AFM). To do that, two cross-sections of both the macroscopically normal urinary bladder wall and the bladder wall adjacent to the tumor were collected and immediately frozen, prior to AFM samples analysis. The respective "twin" formalin-fixed paraffin-embedded tissue fragments were processed and later evaluated for histopathological examination. H&E staining suggested that tumors promoted the development of muscle-like structures in the mucosa surrounding the neoplastic region. The average Young's modulus (cell stiffness) in tumor-adjacent specimens was significantly higher in the muscularis propria than in the mucosa. Similarly, the tumor-free specimens had significantly higher Young's moduli in the muscularis propria than in the urothelium. Young's moduli were higher in all layers of tumor-adjacent tissues when compared with tumor-free samples. Here we provide insights into the stiffness of the bladder wall layers, and we show that the presence of tumor in the surrounding mucosa leads to an alteration of its smooth muscle content. The quantitative assessment of stiffness range here presented provides essential data for future research on BlCa and for understanding how the biomechanical stimuli can modulate cancer cells' capacity to invade through the different bladder layers.
Collapse
Affiliation(s)
- Sara Monteiro-Reis
- Institute of Science and Innovation in Mechanical and Industrial Engineering (INEGI), 4200-465 Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
| | - João P. S. Ferreira
- Institute of Science and Innovation in Mechanical and Industrial Engineering (INEGI), 4200-465 Porto, Portugal
- Department of Mechanical Engineering, Faculty of Engineering, University of Porto (FEUP), 4200-465 Porto, Portugal
| | - Ricardo A. Pires
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto, Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
| | - João A. Carvalho
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Department of Urology, Portuguese Oncology Institute of Porto, 4200-072 Porto, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Renato Natal Jorge
- Institute of Science and Innovation in Mechanical and Industrial Engineering (INEGI), 4200-465 Porto, Portugal
- Department of Mechanical Engineering, Faculty of Engineering, University of Porto (FEUP), 4200-465 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| |
Collapse
|
21
|
The Roles of miRNAs in Predicting Bladder Cancer Recurrence and Resistance to Treatment. Int J Mol Sci 2023; 24:ijms24020964. [PMID: 36674480 PMCID: PMC9864802 DOI: 10.3390/ijms24020964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Bladder cancer (BCa) is associated with significant morbidity, with development linked to environmental, lifestyle, and genetic causes. Recurrence presents a significant issue and is managed in the clinical setting with intravesical chemotherapy or immunotherapy. In order to address challenges such as a limited supply of BCG and identifying cases likely to recur, it would be advantageous to use molecular biomarkers to determine likelihood of recurrence and treatment response. Here, we review microRNAs (miRNAs) that have shown promise as predictors of BCa recurrence. MiRNAs are also discussed in the context of predicting resistance or susceptibility to BCa treatment.
Collapse
|
22
|
Abstract
Bladder cancer as one of the main comorbid diseases might be more susceptible to develop COVID-19 infection with a higher mortality risk during the COVID-19 pandemic. The European Association of Urology (EAU) recommended a comprehensive panel for bladder cancer diagnosis and treatment during this global health problem. The urgent need for treatments of COVID-19 during the pandemic has persuaded researchers to evaluate the different medications, which may lead to drug shortages. Therefore, in this review paper, we have focused on the least recommendations of EAU about bladder cancer during of COVID-19 pandemic to provide a comprehensive panel for high-risk patients.
Collapse
Affiliation(s)
- Shahede Khansary
- Department of Gynecology, Faculty of Medicine, Hamadan university of Medical Sciences, Hamadan, Iran
| | - Heidar Tavilani
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hadi Ghasemi
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| |
Collapse
|
23
|
Vasudevan J, Jiang K, Fernandez J, Lim CT. Extracellular matrix mechanobiology in cancer cell migration. Acta Biomater 2022; 163:351-364. [PMID: 36243367 DOI: 10.1016/j.actbio.2022.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/01/2022]
Abstract
The extracellular matrix (ECM) is pivotal in modulating tumor progression. Besides chemically stimulating tumor cells, it also offers physical support that orchestrates the sequence of events in the metastatic cascade upon dynamically modulating cell mechanosensation. Understanding this translation between matrix biophysical cues and intracellular signaling has led to rapid growth in the interdisciplinary field of cancer mechanobiology in the last decade. Substantial efforts have been made to develop novel in vitro tumor mimicking platforms to visualize and quantify the mechanical forces within the tissue that dictate tumor cell invasion and metastatic growth. This review highlights recent findings on tumor matrix biophysical cues such as fibrillar arrangement, crosslinking density, confinement, rigidity, topography, and non-linear mechanics and their implications on tumor cell behavior. We also emphasize how perturbations in these cues alter cellular mechanisms of mechanotransduction, consequently enhancing malignancy. Finally, we elucidate engineering techniques to individually emulate the mechanical properties of tumors that could help serve as toolkits for developing and testing ECM-targeted therapeutics on novel bioengineered tumor platforms. STATEMENT OF SIGNIFICANCE: Disrupted ECM mechanics is a driving force for transitioning incipient cells to life-threatening malignant variants. Understanding these ECM changes can be crucial as they may aid in developing several efficacious drugs that not only focus on inducing cytotoxic effects but also target specific matrix mechanical cues that support and enhance tumor invasiveness. Designing and implementing an optimal tumor mimic can allow us to predictively map biophysical cue-modulated cell behaviors and facilitate the design of improved lab-grown tumor models with accurately controlled structural features. This review focuses on the abnormal changes within the ECM during tumorigenesis and its implications on tumor cell-matrix mechanoreciprocity. Additionally, it accentuates engineering approaches to produce ECM features of varying levels of complexity which is critical for improving the efficiency of current engineered tumor tissue models.
Collapse
|
24
|
Shiraishi Y, Maehama T, Nishio M, Otani J, Hikasa H, Mak TW, Sasaki T, Honma T, Kondoh Y, Osada H, Yoshida M, Fujisawa M, Suzuki A. N-(3,4-dimethoxyphenethyl)-6-methyl-2,3,4,9-tetrahydro-1H-carbazol-1-amine inhibits bladder cancer progression by suppressing YAP1/TAZ. Genes Cells 2022; 27:602-612. [PMID: 36054428 DOI: 10.1111/gtc.12979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 12/30/2022]
Abstract
Bladder cancer (BlC) is the fourth most common cancer in males worldwide, but few systemic chemotherapy options for its effective treatment exist. The development of new molecularly-targeted agents against BlC is therefore an urgent issue. The Hippo signaling pathway, with its upstream LATS kinases and downstream transcriptional co-activators YAP1 and TAZ, plays a pivotal role in diverse cell functions, including cell proliferation. Recent studies have shown that overexpression of YAP1 occurs in advanced BlCs and is associated with poor patient prognosis. Accessing data from our previous screening of a chemical library of compounds targeting the Hippo pathway, we identified DMPCA (N-(3,4-dimethoxyphenethyl)-6-methyl-2,3,4,9-tetrahydro-1H-carbazol-1-amine) as an agent able to induce the phosphorylation of LATS1 and YAP1/TAZ in BlC cells, thereby suppressing their viability both in vitro and in mouse xenografts. Our data indicate that DMPCA has a potent anti-tumor effect, and raise the possibility that this agent may represent a new and effective therapeutic option for BlC.
Collapse
Affiliation(s)
- Yusuke Shiraishi
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomohiko Maehama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Miki Nishio
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junji Otani
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Hikasa
- Department of Biochemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Tak Wah Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Pathology, LKS Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Teruki Honma
- RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Yasumitsu Kondoh
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Japan.,Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Minoru Yoshida
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Wako, Japan.,Department of Biotechnology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Masato Fujisawa
- Division of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Suzuki
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
25
|
Zakaria MA, Kiew MC, Rajab NF, Chua EW, Masre SF. Rigid Tissue Increases Cytoplasmic pYAP Expression in Pre-Malignant Stage of Lung Squamous Cell Carcinoma (SCC) In Vivo. Curr Issues Mol Biol 2022; 44:4528-4539. [PMID: 36286025 PMCID: PMC9600365 DOI: 10.3390/cimb44100310] [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: 09/06/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
Increased tissue rigidity is able to activate the Hippo signaling pathway, leading to YAP inactivation by phosphorylation and translocation into the cytoplasm. Accumulating evidence suggests that cytoplasmic pYAP serves as a tumor suppressor and could be a prognostic biomarker for several solid cancers. However, the relationship between tissue rigidity and cytoplasmic pYAP expression in the early stage of lung squamous cell carcinoma (SCC) remains elusive; this was determined in this study by using a mouse model. Female BALB/c mice were assigned into two groups (n = 6; the vehicle (VC) and the pre-malignant (PM) group, which received 70% acetone and 0.04 M N-nitroso-tris-chloroethylurea (NTCU) for 15 weeks, respectively. In this study, the formation of hyperplasia and metaplasia lesions was found in the PM group, indicating the pre-malignant stage of lung SCC. The pre-malignant tissue appeared to be more rigid as characterized by significantly higher (p < 0.05) epithelium thickness, proliferative activity, and collagen content than the VC group. The PM group also had a significantly higher (p < 0.05) cytoplasmic pYAP protein expression than the VC group. In conclusion, increased tissue rigidity may contribute to the upregulation of cytoplasmic pYAP expression, which may act as a tumor suppressor in the early stage of lung SCC.
Collapse
Affiliation(s)
- Muhammad Asyaari Zakaria
- Centre for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - May Chee Kiew
- Centre for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Nor Fadilah Rajab
- Centre for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Eng Wee Chua
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| | - Siti Fathiah Masre
- Centre for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
- Correspondence: ; Tel.: +60-137442907
| |
Collapse
|
26
|
Onwudiwe K, Burchett AA, Datta M. Mechanical and metabolic interplay in the brain metastatic microenvironment. Front Oncol 2022; 12:932285. [PMID: 36059679 PMCID: PMC9436395 DOI: 10.3389/fonc.2022.932285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
In this Perspective, we provide our insights and opinions about the contribution-and potential co-regulation-of mechanics and metabolism in incurable breast cancer brain metastasis. Altered metabolic activity can affect cancer metastasis as high glucose supply and demand in the brain microenvironment favors aerobic glycolysis. Similarly, the altered mechanical properties of disseminating cancer cells facilitate migration to and metastatic seeding of the brain, where local metabolites support their progression. Cancer cells in the brain and the brain tumor microenvironment often possess opposing mechanical and metabolic properties compared to extracranial cancer cells and their microenvironment, which inhibit the ease of extravasation and metastasis of these cells outside the central nervous system. We posit that the brain provides a metabolic microenvironment that mechanically reinforces the cellular structure of cancer cells and supports their metastatic growth while restricting their spread from the brain to external organs.
Collapse
Affiliation(s)
| | | | - Meenal Datta
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
27
|
Millet M, Bollmann E, Ringuette Goulet C, Bernard G, Chabaud S, Huot MÉ, Pouliot F, Bolduc S, Bordeleau F. Cancer-Associated Fibroblasts in a 3D Engineered Tissue Model Induce Tumor-like Matrix Stiffening and EMT Transition. Cancers (Basel) 2022; 14:cancers14153810. [PMID: 35954473 PMCID: PMC9367573 DOI: 10.3390/cancers14153810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The physical properties of a tumor, such as stiffness, are important drivers of tumor progression. However, current in vitro tumor models fail to recapitulate the full range of physical properties observed in solid tumors. Here, we proposed a 3D self-assembly engineered bladder model using cancer-associated fibroblasts in which stromal cells produce their extracellular matrix. We then proceeded to assess how our model recapitulates biological and mechanical features found in tumors. We confirmed that stroma assembled by cancer-associated fibroblasts have increased extracellular matrix content and display increased remodeling and higher stiffness. Moreover, normal urothelial cells seeded on the tumor model displayed a mechanotransduction response, increased cell proliferation, cell infiltration within stroma, and displayed features of the epithelial-to-mesenchymal transition. Altogether, we demonstrated that our cancer-associated fibroblast-derived tumor stroma recapitulates several biological and physical features expected from a tumor-like environment and, thus, provides the basis for more accurate cancer models. Abstract A tumor microenvironment is characterized by its altered mechanical properties. However, most models remain unable to faithfully recreate the mechanical properties of a tumor. Engineered models based on the self-assembly method have the potential to better recapitulate the stroma architecture and composition. Here, we used the self-assembly method based on a bladder tissue model to engineer a tumor-like environment. The tissue-engineered tumor models were reconstituted from stroma-derived healthy primary fibroblasts (HFs) induced into cancer-associated fibroblast cells (iCAFs) along with an urothelium overlay. The iCAFs-derived extracellular matrix (ECM) composition was found to be stiffer, with increased ECM deposition and remodeling. The urothelial cells overlaid on the iCAFs-derived ECM were more contractile, as measured by quantitative polarization microscopy, and displayed increased YAP nuclear translocation. We further showed that the proliferation and expression of epithelial-to-mesenchymal transition (EMT) marker in the urothelial cells correlate with the increased stiffness of the iCAFs-derived ECM. Our data showed an increased expression of EMT markers within the urothelium on the iCAFs-derived ECM. Together, our results demonstrate that our tissue-engineered tumor model can achieve stiffness levels comparable to that of a bladder tumor, while triggering a tumor-like response from the urothelium.
Collapse
Affiliation(s)
- Martial Millet
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Enola Bollmann
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
| | - Cassandra Ringuette Goulet
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division), Quebec City, QC G1V 4G2, Canada
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Quebec City, QC G1J 1Z4, Canada
| | - Geneviève Bernard
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division), Quebec City, QC G1V 4G2, Canada
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Quebec City, QC G1J 1Z4, Canada
| | - Stéphane Chabaud
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division), Quebec City, QC G1V 4G2, Canada
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Quebec City, QC G1J 1Z4, Canada
| | - Marc-Étienne Huot
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Frédéric Pouliot
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
- Department of Surgery, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Stéphane Bolduc
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division), Quebec City, QC G1V 4G2, Canada
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Quebec City, QC G1J 1Z4, Canada
- Department of Surgery, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - François Bordeleau
- CHU de Québec-Université Laval Research Center (Oncology Division) and Université Laval Cancer Research Center, Quebec City, QC G1R 3S3, Canada
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Université Laval, Quebec City, QC G1J 1Z4, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-525-4444 (ext. 15554)
| |
Collapse
|
28
|
Cheng X, Lou K, Ding L, Zou X, Huang R, Xu G, Zou J, Zhang G. Clinical potential of the Hippo-YAP pathway in bladder cancer. Front Oncol 2022; 12:925278. [PMID: 35912245 PMCID: PMC9336529 DOI: 10.3389/fonc.2022.925278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Bladder cancer (BC) is one of the world’s most frequent cancers. Surgery coupled with adjuvant platinum-based chemotherapy is the current standard of therapy for BC. However, a high proportion of patients progressed to chemotherapy-resistant or even neoplasm recurrence. Hence, identifying novel treatment targets is critical for clinical treatment. Current studies indicated that the Hippo-YAP pathway plays a crucial in regulating the survival of cancer stem cells (CSCs), which is related to the progression and reoccurrence of a variety of cancers. In this review, we summarize the evidence that Hippo-YAP mediates the occurrence, progression and chemotherapy resistance in BC, as well as the role of the Hippo-YAP pathway in regulating bladder cancer stem-like cells (BCSCs). Finally, the clinical potential of Hippo-YAP in the treatment of BC was prospected.
Collapse
Affiliation(s)
- Xin Cheng
- First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Kecheng Lou
- First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Liang Ding
- First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Ruohui Huang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Gang Xu
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Junrong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Guoxi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
- *Correspondence: Guoxi Zhang,
| |
Collapse
|
29
|
Xiong Y, Pang M, Du Y, Yu X, Yuan J, Liu W, Wang L, Liu X. The LINC01929/miR-6875-5p/ADAMTS12 Axis in the ceRNA Network Regulates the Development of Advanced Bladder Cancer. Front Oncol 2022; 12:856560. [PMID: 35646642 PMCID: PMC9133480 DOI: 10.3389/fonc.2022.856560] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Considering its speedy development and extremely low 5-year overall survival rate worldwide, bladder cancer (BCa) is one of the most common and highly malignant tumors. Increasing evidence suggests that protein-coding mRNAs and non-coding RNAs, including long non-coding RNAs (lncRNAs) and micro RNAs (miRNAs), play an essential role in regulating the biological processes of cancer. To investigate the molecular regulation associated with poor prognosis during advanced BCa development, we constructed a competitive endogenous RNA (ceRNA) network. Using transcriptome profiles from The Cancer Genome Atlas and Gene Expression Omnibus databases, we performed differential expression (DE) analysis, weighted gene co-expression network analysis, functional enrichment analysis, survival analysis, prediction of miRNA targeting, and Pearson correlation analysis. Through layers of selection, 8 lncRNAs-28 mRNAs and 8 miRNAs-28 mRNAs pairs shared similar expression patterns, constituting a core ceRNA regulatory network related to the invasion, progression, and metastasis of advanced clinical stage (ACS) BCa. Subsequently, we conducted real time qPCR, western blotting, and immunohistochemistry to validate expression trend bioinformatics analysis on 3, 2, and 3 differentially expressed mRNAs, lncRNAs, and miRNAs, respectively. The most significantly differentially expressed LINC01929, miR-6875-5p and ADAMTS12 were selected for in vitro experiments to assess the functional role of the LINC01929/miR-6875-5p/ADAMTS12 axis. RNA pull-down, luciferase assays, and rescue assays were performed to examine the binding of LINC01929 and miR-6875-5p. Increasing trends in COL6A1, CDH11, ADAMTS12, LINC01705, and LINC01929 expression variation were verified as consistent with previous DE analysis results in ACS-BCa, compared with low clinical stage (LCS) BCa. Expression trends in parts of these RNAs, such as hsa-miR-6875-5p, hsa-miR-6784-5p, COL6A1, and CDH11, were measured in accordance with DE analysis in LCS-BCa, compared with normal bladder urothelium. Through experimental validation, the cancer-promoting molecule ADAMST12 was found to play a key role in the development of advanced BCa. Functionally, ADAMTS12 knockdown inhibited the progression of bladder cancer. Overexpression of LINC01929 promoted bladder cancer development, while overexpression of miR-6785-5p inhibited bladder cancer development. Mechanistically, LINC01929 acted as a sponge for miR-6785-5p and partially reversed the role of miR-6785-5p. Our findings provide an elucidation of the molecular mechanism by which advanced bladder cancer highly expressed LINC01929 upregulates ADAMTS12 expression through competitive adsorption of miR-6875-5p. It provides a new target for the prognosis and diagnosis of advanced bladder cancer.
Collapse
Affiliation(s)
- YuFeng Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - MingRui Pang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Du
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - JingPing Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wen Liu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - XiuHeng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Allahyari Z, Casillo SM, Perry SJ, Peredo AP, Gholizadeh S, Gaborski TR. Disrupted Surfaces of Porous Membranes Reduce Nuclear YAP Localization and Enhance Adipogenesis through Morphological Changes. ACS Biomater Sci Eng 2022; 8:1791-1798. [PMID: 35363465 DOI: 10.1021/acsbiomaterials.1c01472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The disrupted surface of porous membranes, commonly used in tissue-chip and cellular coculture systems, is known to weaken cell-substrate interactions. Here, we investigated whether disrupted surfaces of membranes with micron and submicron scale pores affect yes-associated protein (YAP) localization and differentiation of adipose-derived stem cells. We found that these substrates reduce YAP nuclear localization through decreased cell spreading, consistent with reduced cell-substrate interactions, and in turn enhance adipogenesis while decreasing osteogenesis.
Collapse
Affiliation(s)
- Zahra Allahyari
- Department of Microsystems Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States.,Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Stephanie M Casillo
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Spencer J Perry
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Ana P Peredo
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Shayan Gholizadeh
- Department of Microsystems Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States.,Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Thomas R Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, New York 14623, United States
| |
Collapse
|
31
|
Zhang X, Xiong H, Zhao Y, Lin S, Huang X, Lin C, Mao S, Chen D. Circular RNA LONP2 regulates proliferation, invasion, and apoptosis of bladder cancer cells by sponging microRNA-584-5p. Bioengineered 2022; 13:8823-8835. [PMID: 35358000 PMCID: PMC9161836 DOI: 10.1080/21655979.2022.2054753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Bladder cancer (BC) is the most frequent type of urinary tumor and a barely treatable disease. Although extensive efforts have been invested in the research of BC, the underlying etiology and pathophysiology remain unclear. CircLONP2 is a circular RNA implicated in the development of many cancers, and miR-584-5p and YAP1 have been reported to contribute to the progression of BC. In this research, we presented novel evidence supporting circLONP2/miR-584-5p/YAP1 axis as a novel regulatory module in the progression of BC. We analyzed the expression of circLONP2 between precancerous BC samples and normal tissues using a published RNA-seq dataset. The expression of circLONP2 was also validated in clinical samples and cell lines by quantitative RT-PCR. Small interfering RNA (siRNA) and miRNA inhibitor was utilized to modulate the expression of circLONP2 and miR-584-5p and investigate their functions on cell proliferation and invasion. Luciferase reporter assay and RNA pull-down were performed to confirm the functional interactions among circLONP2/miR-584-5p/YAP1. CircLONP2 was significantly upregulated in precancerous BC tissues and BC cells. CircLONP2 depletion inhibited cell viability, proliferation, and invasion of BC cell lines, which could be partially rescued by miR-584-5p inhibitor. Further experiments indicated that miR-584-5p regulates cell viability, proliferation, and invasion via directly targeting YAP1. In summary, our work indicates that circLONP2 plays an oncogenic function in BC by regulating miR-584-5p/YAP1 axis, and its interaction with miR-584-5p provides a potential strategy to target BC.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Hao Xiong
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Yong Zhao
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Shengqiang Lin
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Xiang Huang
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Cheng Lin
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Shihui Mao
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| | - Demin Chen
- Department of Urology, The Affiliated Nanping First Hospital of Fujian Medical University, Nanping, Fujian, China
| |
Collapse
|
32
|
Matrix Stiffness Contributes to Cancer Progression by Regulating Transcription Factors. Cancers (Basel) 2022; 14:cancers14041049. [PMID: 35205794 PMCID: PMC8870363 DOI: 10.3390/cancers14041049] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Matrix stiffness is recognized as a critical factor in cancer progression. Recent studies have shown that matrix stiffening is caused by the accumulation, contraction, and crosslinking of the extracellular matrix by cancer and stromal cells. Cancer and stromal cells respond to matrix stiffness, which determines the phenotypes of these cells. In addition, matrix stiffness activates and/or inactivates specific transcription factors in cancer and stromal cells to regulate cancer progression. In this review, we discuss the mechanisms of cancer stiffening and progression that are regulated by transcription factors responding to matrix stiffness. Abstract Matrix stiffness is critical for the progression of various types of cancers. In solid cancers such as mammary and pancreatic cancers, tumors often contain abnormally stiff tissues, mainly caused by stiff extracellular matrices due to accumulation, contraction, and crosslinking. Stiff extracellular matrices trigger mechanotransduction, the conversion of mechanical cues such as stiffness of the matrix to biochemical signaling in the cells, and as a result determine the cellular phenotypes of cancer and stromal cells in tumors. Transcription factors are key molecules for these processes, as they respond to matrix stiffness and are crucial for cellular behaviors. The Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) is one of the most studied transcription factors that is regulated by matrix stiffness. The YAP/TAZ are activated by a stiff matrix and promotes malignant phenotypes in cancer and stromal cells, including cancer-associated fibroblasts. In addition, other transcription factors such as β-catenin and nuclear factor kappa B (NF-κB) also play key roles in mechanotransduction in cancer tissues. In this review, the mechanisms of stiffening cancer tissues are introduced, and the transcription factors regulated by matrix stiffness in cancer and stromal cells and their roles in cancer progression are shown.
Collapse
|
33
|
Ghassemi H, Hashemnia M, Mousavibahar SH, Mahmoodzadeh Hosseini H, Mirhosseini SA. Positive Association of Matrix Proteins Alteration with TAZ and The Progression of High-Grade Bladder Cancer. CELL JOURNAL 2021; 23:742-749. [PMID: 34979063 PMCID: PMC8753103 DOI: 10.22074/cellj.2021.7661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/31/2020] [Indexed: 11/25/2022]
Abstract
Objective Bladder cancer is the 9th cause of human urologic malignancy and the 13th of death worldwide.
Increased collagen cross-linking, NIDOGEN1 expression and consequently stiffness of extracellular matrix (ECM) may
be responsible for the mechanotransduction and regulation of transcriptional co-activator with PDZ-binding motif (TAZ)
and transforming growth factor β1 (TGF-β1) signaling pathways, resulting in progression of tumorigenesis. The present
study aimed to assess whether type 1 collagen expression is associated with TAZ nuclear localization. Materials and Methods In this case-control study, real-time quantitative reverse transcription polymerase chain
reaction (qRT-PCR) and immunohistochemical analysis were performed to evaluate the activation of the TAZ pathway
in patients with bladder cancer (n=40) and healthy individuals (n=20). The ELISA method was also conducted to
measure the serum concentrations of TGF-β1. Masson’s trichrome staining was carried out to histologically evaluate
the density of type 1 collagen.
Results Our findings that the expression levels of COL1A1, COL1A2, NIDOGEN1, TAZ, and TGF-β1 genes were
overexpressed in patients with bladder cancer, and their expression levels were positively associated with the grade
of bladder cancer. The immunohistochemical analysis demonstrated that the nuclear localization of TAZ was markedly
correlated with high-grade bladder cancer. We also found that TAZ nuclear localization was substantially higher in
cancerous tissues as compared with normal bladder tissues. Masson's trichrome staining showed that the tissue density
of type I collagen was considerably increased in patients with bladder cancer as compared with healthy subjects.
Conclusion According to our findings, it seems the alterations in the expression of type I collagen and NIDOGEN1,
as well as TAZ nuclear localization influence the progression of bladder cancer. The significance of TGF-β1 and TAZ
expression in tumorigenesis and progression to high-grade bladder cancer was also highlighted. However, a possible
relationship between TGF-β1 expression and the Hippo pathway needs further investigations.
Collapse
Affiliation(s)
- Hadi Ghassemi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hashemnia
- Department of Pathobiology, Veterinary Medicine Faculty Razi University, Kermanshah, Iran
| | | | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Fu Y, Wan P, Zhang J, Li X, Xing J, Zou Y, Wang K, Peng H, Zhu Q, Cao L, Zhai X. Targeting Mechanosensitive Piezo1 Alleviated Renal Fibrosis Through p38MAPK-YAP Pathway. Front Cell Dev Biol 2021; 9:741060. [PMID: 34805150 PMCID: PMC8602364 DOI: 10.3389/fcell.2021.741060] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
Renal fibrosis is the most common pathological manifestation of a wide variety of chronic kidney disease. Increased extracellular matrix (ECM) secretion and enhanced microenvironment stiffening aggravate the progression of renal fibrosis. However, the related mechanisms remain unclear. Here, we evaluated the mechanism by which ECM stiffness aggravates renal fibrosis. In the present study, renal mesangial cells (MCs) were cultured on polyacrylamide hydrogels with different stiffness accurately detected by atomic force microscope (AFM), simulating the in vivo growth microenvironment of MCs in normal kidney and renal fibrosis. A series of in vitro knockdown and activation experiments were performed to establish the signaling pathway responsible for mechanics-induced MCs activation. In addition, an animal model of renal fibrosis was established in mice induced by unilateral ureteral obstruction (UUO). Lentiviral particles containing short hairpin RNA (sh RNA) targeting Piezo1 were used to explore the effect of Piezo1 knockdown on matrix stiffness-induced MCs activation and UUO-induced renal fibrosis. An in vitro experiment demonstrated that elevated ECM stiffness triggered the activation of Piezo1, which increased YAP nuclear translocation through the p38MAPK, and consequently led to increased ECM secretion. Furthermore, these consequences have been verified in the animal model of renal fibrosis induced by UUO and Piezo1 knockdown could alleviate UUO-induced fibrosis and improve renal function in vivo. Collectively, our results for the first time demonstrate enhanced matrix stiffness aggravates the progression of renal fibrosis through the Piezo1-p38MAPK-YAP pathway. Targeting mechanosensitive Piezo1 might be a potential therapeutic strategy for delaying the progression of renal fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Pengzhi Wan
- Department of Nephrology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jie Zhang
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Xue Li
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jia Xing
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yu Zou
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Kaiyue Wang
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Hui Peng
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qizhuo Zhu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Liu Cao
- Department of Basic Medical College, China Medical University, Shenyang, China
| | - Xiaoyue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Institute of Nephropathology, China Medical University, Shenyang, China
| |
Collapse
|
35
|
Huang K, Liu J, Chen Q, Feng D, Wu H, Aldanakh A, Jian Y, Xu Z, Wang S, Yang D. The effect of mechanical force in genitourinary malignancies. Expert Rev Anticancer Ther 2021; 22:53-64. [PMID: 34726963 DOI: 10.1080/14737140.2022.2000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mechanical force is attributed to the formation of tumor blood vessels, influences cancer cell invasion and metastasis, and promotes reprogramming of the energy metabolism. Currently, therapy strategies for the tumor microenvironment are being developed progressively. The purpose of this article is to discuss the molecular mechanism, diagnosis, and treatment of mechanical force in urinary tract cancers and outline the medications used in the mechanical microenvironment. AREAS COVERED This review covers the complex mechanical elements in the microenvironment of urinary system malignancies, focusing on mechanical molecular mechanisms for diagnosis and treatment. EXPERT OPINION The classification of various mechanical forces, such as matrix stiffness, shear force, and other forces, is relatively straightforward. However, little is known about the molecular process of mechanical forces in urinary tract malignancies. Because mechanical therapy is still controversial, it is critical to understand the molecular basis of mechanical force before adding mechanical therapy solutions.
Collapse
Affiliation(s)
- Kai Huang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Junqiang Liu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiwei Chen
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China.,School of Information Science and Technology, Dalian Maritime University, Dalian City, China
| | - Dan Feng
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Haotian Wu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Abdullah Aldanakh
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuli Jian
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Zhongyang Xu
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Deyong Yang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
36
|
Zhang W, Zhang S, Zhang W, Yue Y, Qian W, Wang Z. Matrix stiffness and its influence on pancreatic diseases. Biochim Biophys Acta Rev Cancer 2021; 1876:188583. [PMID: 34139274 DOI: 10.1016/j.bbcan.2021.188583] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/09/2021] [Accepted: 06/13/2021] [Indexed: 01/12/2023]
Abstract
The matrix stiffness of the extracellular matrix(ECM), which is the slow elastic force on cells, has gradually become investigated. And a higher stiffness could induce changes in cell biological behaviors and activation of internal signaling pathways. Imbalanced stiffness of ECM is associated with a number of diseases, including pancreatic disease. In this review, we discuss the components of the ECM and the increased stiffness caused by unbalanced ECM changes. Next, we describe how matrix stiffness transmits mechanical signals and what signaling pathways are altered within the cell in detail. Finally, we discuss the effect of ECM on the behavior of pancreatic diseases from the perspective of matrix stiffness.
Collapse
Affiliation(s)
- Weifan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China
| | - Simei Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China
| | - Wunai Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China
| | - Yangyang Yue
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi Province, China.
| |
Collapse
|
37
|
Ghasemi H, Mousavibahar SH, Hashemnia M, Karimi J, Khodadadi I, Tavilani H. Transitional cell carcinoma matrix stiffness regulates the osteopontin and YAP expression in recurrent patients. Mol Biol Rep 2021; 48:4253-4262. [PMID: 34086159 DOI: 10.1007/s11033-021-06440-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Cells translate the mechanosensing of extracellular matrix component dysregulation and stiffness into the signal transduction including Osteopontin (OPN) through the Hippo pathway. But how extracellular matrix (ECM) component dysregulation and stiffness are ultimately linked to transitional cell carcinoma (TCC) development remains poorly understood. This study was aimed to evaluate the possible links between ECM component alteration after cancer surgery and OPN and Yes-associated protein (YAP) expression in TCC and adjacent tissues. In this study, we used 50 TCC (25 newly diagnosed and 25 recurrent) and 50 adjacent tissues to determine the tissue stiffness using atomic force microscopy. The mRNA expression of SPP1, Indian hedgehog (IHH), and YAP was also determined using qRT-PCR. Western blotting and ELISA were performed to assess the tissue and serum levels of OPN, respectively. To assess the glycoproteins and elastic fibers content, Periodic Acid Schiff, and Verhoeff-Van Gieson Staining were performed, respectively. Matrix stiffness was markedly higher in TCCs than adjacent tissues (p < 0.05). Gene expression analysis showed that YAP, SPP1, and IHH genes were upregulated in TCC tissues (p < 0.05). Additionally, the OPN protein overexpression was observed in the tissue and the serum of TCC patients (p < 0.05). We also found that glycoproteins, elastic fibers content of recurrent TCC tissues was remarkably higher as compared to adjacent tissues (p < 0.05). Our results suggest that glycoproteins and elastic fibers content modulation and ECM stiffness may upregulates the expression of YAP, SPP1 and IHH genes, and possibly contribute to the TCC development and relapse.
Collapse
Affiliation(s)
- Hadi Ghasemi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Hashemnia
- Department of Pathobiology, Veterinary Medicine Faculty, Razi University, Kermanshah, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Heidar Tavilani
- Urology & Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
38
|
Cai X, Wang KC, Meng Z. Mechanoregulation of YAP and TAZ in Cellular Homeostasis and Disease Progression. Front Cell Dev Biol 2021; 9:673599. [PMID: 34109179 PMCID: PMC8182050 DOI: 10.3389/fcell.2021.673599] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Biophysical cues, such as mechanical properties, play a critical role in tissue growth and homeostasis. During organ development and tissue injury repair, compressive and tensional forces generated by cell-extracellular matrix or cell-cell interaction are key factors for cell fate determination. In the vascular system, hemodynamic forces, shear stress, and cyclic stretch modulate vascular cell phenotypes and susceptibility to atherosclerosis. Despite that emerging efforts have been made to investigate how mechanotransduction is involved in tuning cell and tissue functions in various contexts, the regulatory mechanisms remain largely unknown. One of the challenges is to understand the signaling cascades that transmit mechanical cues from the plasma membrane to the cytoplasm and then to the nuclei to generate mechanoresponsive transcriptomes. YAP and its homolog TAZ, the Hippo pathway effectors, have been identified as key mechanotransducers that sense mechanical stimuli and relay the signals to control transcriptional programs for cell proliferation, differentiation, and transformation. However, the upstream mechanosensors for YAP/TAZ signaling and downstream transcriptome responses following YAP/TAZ activation or repression have not been well characterized. Moreover, the mechanoregulation of YAP/TAZ in literature is highly context-dependent. In this review, we summarize the biomechanical cues in the tissue microenvironment and provide an update on the roles of YAP/TAZ in mechanotransduction in various physiological and pathological conditions.
Collapse
Affiliation(s)
- Xiaomin Cai
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
39
|
Li Y, Sun C, Tan Y, Zhang H, Li Y, Zou H. ITGB1 enhances the Radioresistance of human Non-small Cell Lung Cancer Cells by modulating the DNA damage response and YAP1-induced Epithelial-mesenchymal Transition. Int J Biol Sci 2021; 17:635-650. [PMID: 33613118 PMCID: PMC7893583 DOI: 10.7150/ijbs.52319] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/27/2020] [Indexed: 12/13/2022] Open
Abstract
Objectives: Radiotherapy has played a limited role in the treatment of non-small cell lung cancer (NSCLC) due to the risk of tumour radioresistance. We previously established the radioresistant non-small cell lung cancer (NSCLC) cell line H460R. In this study, we identified differentially expressed genes between these radioresistant H460R cells and their radiosensitive parent line. We further evaluated the role of a differentially expressed gene, ITGB1, in NSCLC cell radioresistance and as a potential target for improving radiosensitivity. Materials and Methods: The radiosensitivity of NSCLC cells was evaluated by flow cytometry, colony formation assays, immunofluorescence, and Western blotting. Bioinformatics assay was used to identify the effect of ITGB1 and YAP1 expression in NSCLC tissues. Results: ITGB1 mRNA and protein expression levels were higher in H460R than in the parental H460 cells. We observed lower clonogenic survival and cell viability and a higher rate of apoptosis of ITGB1-knockdown A549 and H460R cells than of wild type cells post-irradiation. Transfection with an ITGB1 short hairpin (sh) RNA enhanced radiation-induced DNA damage and G2/M phase arrest. Moreover, ITGB1 induced epithelial-mesenchymal transition (EMT) of NSCLC cells. Silencing ITGB1 suppressed the expression and intracellular translocation of Yes-associated protein 1 (YAP1), a downstream effector of ITGB1. Conclusions: ITGB1 may induce radioresistance via affecting DNA repair and YAP1-induced EMT. Taken together, our data suggest that ITGB1 is an attractive therapeutic target to overcome NSCLC cell radioresistance.
Collapse
Affiliation(s)
- Yuexian Li
- Department of Oncology, Shengjing Hospital affiliated with China Medical University, Shenyang 110004, China
| | - Cheng Sun
- Department of Oncology, Shengjing Hospital affiliated with China Medical University, Shenyang 110004, China
| | - Yonggang Tan
- Department of Oncology, Shengjing Hospital affiliated with China Medical University, Shenyang 110004, China
| | - Heying Zhang
- Department of Oncology, Shengjing Hospital affiliated with China Medical University, Shenyang 110004, China
| | - Yuchao Li
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases
| | - Huawei Zou
- Department of Oncology, Shengjing Hospital affiliated with China Medical University, Shenyang 110004, China
| |
Collapse
|