1
|
Chen D, Lu S, Huang K, Pearson JD, Pacal M, Peidis P, McCurdy S, Yu T, Sangwan M, Nguyen A, Monnier PP, Schramek D, Zhu L, Santamaria D, Barbacid M, Akeno N, Wikenheiser-Brokamp KA, Bremner R. Cell cycle duration determines oncogenic transformation capacity. Nature 2025; 641:1309-1318. [PMID: 40307557 PMCID: PMC12119354 DOI: 10.1038/s41586-025-08935-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
Oncogenic mutations are widespread in normal human tissues1. Similarly, in murine chimeras, cells carrying an oncogenic lesion contribute normal cells to adult tissues without causing cancer2-4. How lineages that escape cancer via normal development differ from the minority that succumb is unclear. Tumours exhibit characteristic cancer hallmarks; we therefore searched for hallmarks that differentiate cancer-prone lineages from resistant lineages. Here we show that total cell cycle duration (Tc) predicts transformation susceptibility across multiple tumour types. Cancer-prone Rb- and p107-deficient retina (Rb is also known as Rb1 and p107 is also known as Rbl1) exhibited defects in apoptosis, senescence, immune surveillance, angiogenesis, DNA repair, polarity and proliferation. Perturbing the SKP2-p27-CDK2/CDK1 axis could block cancer without affecting these hallmarks. Thus, cancer requires more than the presence of its hallmarks. Notably, every tumour-suppressive mutation that we tested increased Tc, and the Tc of the cell of origin of retinoblastoma cells was half that of resistant lineages. Tc also differentiated the cell of origin in Rb-/- pituitary cancer. In lung, loss of Rb and p53 (also known as Trp53) transforms neuroendocrine cells, whereas KrasG12D or BrafV600E mutations transform alveolar type 2 cells5-7. The shortest Tc consistently identified the cell of origin, regardless of mutation timing. Thus, relative Tc is a hallmark of initiation that distinguishes cancer-prone from cancer-resistant lineages in several settings, explaining how mutated cells escape transformation without inducing apoptosis, senescence or immune surveillance.
Collapse
Affiliation(s)
- Danian Chen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology, Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Suying Lu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Katherine Huang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Joel D Pearson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- CancerCare Manitoba Research Institute and Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marek Pacal
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Phillipos Peidis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Sean McCurdy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Tao Yu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Monika Sangwan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Angela Nguyen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Philippe P Monnier
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Liang Zhu
- Department of Developmental and Molecular Biology, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David Santamaria
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca, Spain
| | - Mariano Barbacid
- Molecular Oncology Program, National Center for Cancer Research (CNIO), Madrid, Spain
| | - Nagako Akeno
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathryn A Wikenheiser-Brokamp
- The Perinatal Institute Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada.
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Kofler M, Venugopal S, Gill G, Di Ciano-Oliveira C, Kapus A. M-Motif, a potential non-conventional NLS in YAP/TAZ and other cellular and viral proteins that inhibits classic protein import. iScience 2025; 28:112105. [PMID: 40224012 PMCID: PMC11986988 DOI: 10.1016/j.isci.2025.112105] [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: 03/12/2024] [Revised: 08/22/2024] [Accepted: 02/21/2025] [Indexed: 04/15/2025] Open
Abstract
Multiple mechanisms were proposed to mediate the nuclear import of TAZ/YAP, transcriptional co-activators regulating organ growth and regeneration. Our earlier observations showed that TAZ/YAP harbor a C-terminal, unconventional nuclear localization signal (NLS). Here, we show that this sequence, necessary and sufficient for basal, ATP-independent nuclear import, contains an indispensable central methionine flanked by negatively charged residues. Based on these features, we define the M-motif and propose that it is a new class of NLS, also present and import-competent in other cellular (STAT1 and cyclin B1) and viral (ORF6 of SARS-CoV2, VSV-M) proteins. Accordingly, ORF6 SARS-Cov2 competitively inhibits TAZ/YAP uptake, while TAZ abrogates STAT1 import. Similar to viral M-motif proteins, TAZ binds RAE1 and inhibits classic nuclear protein import, including that of antiviral factors (IRF3 and NF-κB). However, RAE1 is dispensable for TAZ import itself. Thus, the TAZ/YAP NLS has a dual function: it mediates unconventional nuclear import and inhibits classic import, contributing to the suppression of antiviral responses.
Collapse
Affiliation(s)
- Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | - Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | - Gary Gill
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | | | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
- Department Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
3
|
Zhang S, Hong HI, Mak VCY, Zhou Y, Lu Y, Zhuang G, Cheung LWT. Vertical inhibition of p110α/AKT and N-cadherin enhances treatment efficacy in PIK3CA-aberrated ovarian cancer cells. Mol Oncol 2025; 19:1132-1154. [PMID: 39543937 PMCID: PMC11977650 DOI: 10.1002/1878-0261.13761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha [PIK3CA, encoding PI3Kalpha (also known as p110α)] is one of the most commonly aberrated genes in human cancers. In serous ovarian cancer, PIK3CA amplification is highly frequent but PIK3CA point mutation is rare. However, whether PIK3CA amplification and PIK3CA driver mutations have the same functional impact in the disease is unclear. Here, we report that both PIK3CA amplification and E545K mutation are tumorigenic. While the protein kinase B (AKT) signaling axis was activated in both E545K knock-in cells and PIK3CA-overexpressing cells, the mitogen-activated protein kinase 3/1 (ERK1/2) pathway was induced selectively by E545K mutation but not PIK3CA amplification. Intriguingly, AKT signaling in these PIK3CA-aberrated cells increased transcriptional coactivator YAP1 (YAP) Ser127 phosphorylation and thereby cytoplasmic YAP levels, which in turn increased cell migration through Ras-related C3 botulinum toxin substrate 1 (RAC1) activation. In addition to the altered YAP signaling, AKT upregulated N-cadherin expression, which also contributed to cell migration. Pharmacological inhibition of N-cadherin reduced cell migratory potential. Importantly, co-targeting N-cadherin and p110α/AKT caused additive reduction in cell migration in vitro and metastases formation in vivo. Together, this study reveals the molecular pathways driven by the PIK3CA aberrations and the exploitable vulnerabilities in PIK3CA-aberrated serous ovarian cancer cells.
Collapse
Affiliation(s)
- Shibo Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
- Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouChina
| | - Hei Ip Hong
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Victor C. Y. Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Yiling Lu
- Division of Cancer Medicine, Department of Genomic MedicineUT MD Anderson Cancer CentreHoustonTXUSA
| | - Guanglei Zhuang
- State Key Laboratory of Systems Medicine for Cancer, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer InstituteShanghai Jiao Tong University School of MedicineChina
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji HospitalShanghai Jiao Tong University School of MedicineChina
| | - Lydia W. T. Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| |
Collapse
|
4
|
Chan SW, Ong C, Hong W. The recent advances and implications in cancer therapy for the hippo pathway. Curr Opin Cell Biol 2025; 93:102476. [PMID: 39908768 DOI: 10.1016/j.ceb.2025.102476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/07/2025]
Abstract
The Hippo pathway is a highly conserved signaling network integrating diverse intracellular, intercellular and extracellular biological cues to regulate complex physiological processes such as organ size, tissue development, homeostasis and regeneration. These cues include cytoskeletal organization, mechanical force, cell-cell interaction, cell polarity, cell-extracellular matrix interaction to govern cell proliferation, differentiation, apoptosis, stem cell property and tissue microenvironment. In this review, we discuss how the emerging role of biomolecular condensates regulates the activity of the pathway components, and how dysregulation of the pathway leads to cancer. Lastly, we highlight the therapeutic modalities which target YAP/TAZ-TEAD interaction for cancer therapy.
Collapse
Affiliation(s)
- Siew Wee Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Camellia Ong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.
| |
Collapse
|
5
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
6
|
Su JX, Zhou HX, Zhang ZJ, Zhou XF, Zou QM, Li SJ, Zhuang XS, Lai JQ, Yang SY, Cui K, Liu YQ, Yuan RJ, Pan HX, Li ZS, Tu HY, Cheng M, Yan Y, Qi Q, Zhang YB. Gracillin suppresses cancer progression through inducing Merlin/LATS protein-protein interaction and activating Hippo signaling pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01514-w. [PMID: 40055528 DOI: 10.1038/s41401-025-01514-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/13/2025] [Indexed: 03/12/2025]
Abstract
Gene therapy, epigenetic therapies, natural compounds targeted therapy, photodynamic therapy, nanoparticles, and precision medicines are becoming available to diagnose and treat cancer. Gracillin, a natural steroidal saponin extracted from herbs, has shown potent efficacy against a range of malignancies. In this study, we investigated the molecular anticancer mechanisms of gracillin. We showed that gracillin dose-dependently suppressed proliferation, migration, and invasion in breast cancer, liver cancer, and glioblastoma cells with IC50 values around 1 μM, which were associated with MST-independent activation of Hippo signaling pathway and subsequent decreased YAP activity. We demonstrated that gracillin activated the Hippo signaling by inducing Merlin/LATS protein-protein interaction (PPI). A competitive inhibitory peptide (SP) derived from the binding interface of the PPI, disrupted the interaction, abolishing the anticancer activity of gracillin. In nude mice bearing MDA-MB-231, HCCLM3, or U87MG xenograft tumor, administration of gracillin (5, 10 mg·kg-1·d-1, i.g. for 21 days) dose-dependently suppressed the tumor growth, associated with the induced Merlin/LATS PPI, activated Hippo signaling, as well as decreased YAP activity in tumor tissues. Our data demonstrate that gracillin is an efficacious therapeutic agent for cancer treatment, induction of Merlin/LATS PPI might provide proof-of-concept in developing therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Jin-Xuan Su
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Hai-Xia Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhi-Jing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Qiu-Ming Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Si-Jia Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiao-Song Zhuang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jian-Qin Lai
- Department of Colorectal & Anal Surgery, Guangzhou First People's Hospital, Guangzhou, 510632, China
| | - Si-Yu Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Kai Cui
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yong-Qi Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Respiratory and Critical Care, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Rui-Jie Yuan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Heng-Xin Pan
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Zi-Sheng Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Han-Yun Tu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Mei Cheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Yu Yan
- Functional Experimental Teaching Center, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Yu-Bo Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
7
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
8
|
Kaarijärvi R, Kaljunen H, Niemi O, Räsänen M, Paakinaho V, Ketola K. Matrix stiffness modulates androgen response genes and chromatin state in prostate cancer. NAR Cancer 2025; 7:zcaf010. [PMID: 40115748 PMCID: PMC11923743 DOI: 10.1093/narcan/zcaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 02/10/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025] Open
Abstract
The interplay between the extracellular matrix (ECM) and prostate cancer has been shown to increase ECM stiffness, correlating with more aggressive disease forms. However, the impact of ECM stiffness on the androgen receptor (AR), a key target in prostate cancer treatment, remains elusive. Here, we investigated whether matrix stiffness influences prostate cancer progression, transcriptional regulation, chromatin state, and AR function in AR-positive prostate cancer cells under varying ECM stiffness conditions. We utilized ATAC-seq (assay for transposase-accessible chromatin with sequencing) and RNA sequencing under different ECM conditions, along with the SUC2 metastatic prostate adenocarcinoma patient dataset, to investigate the role of ECM stiffness in chromatin state and androgen response genes, as well as its impact on prostate cancer progression. Results demonstrated that increased ECM stiffness elevated the expression of genes related to proliferation and differentiation. In contrast, androgen response genes were most highly induced in soft ECM conditions. Integrating chromatin accessibility with transcriptomic data revealed that androgen response genes were more transcriptionally available in soft ECM conditions. Additionally, increased ECM stiffness upregulated genes associated with low overall survival in the SUC2 dataset. Taken together, our results indicate that high expression of hard matrix stiffness genes may promote prostate cancer progression, leading to more aggressive disease forms associated with poor survival.
Collapse
Affiliation(s)
- Roosa Kaarijärvi
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Heidi Kaljunen
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Onni Niemi
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Merja Räsänen
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Ville Paakinaho
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Kirsi Ketola
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| |
Collapse
|
9
|
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L, Li W. Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 2025; 16:1477. [PMID: 39929832 PMCID: PMC11811181 DOI: 10.1038/s41467-024-55068-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Post-operative recurrence rates of stage I non-small cell lung cancer (NSCLC) range from 20% to 40%. Nonetheless, the molecular mechanisms underlying recurrence hitherto remain largely elusive. Here, we generate genomic, epigenomic and transcriptomic profiles of paired tumors and adjacent tissues from 122 stage I NSCLC patients, among which 57 patients develop recurrence after surgery during follow-up. Integrated analyses illustrate that the presence of predominantly solid or micropapillary histological subtypes, increased genomic instability, and APOBEC-related signature are associated with recurrence. Furthermore, TP53 missense mutation in DNA-binding domain could contribute to shorter time to recurrence. DNA hypomethylation is pronounced in recurrent NSCLC, and PRAME is the significantly hypomethylated and overexpressed gene in recurrent lung adenocarcinoma (LUAD). Mechanistically, hypomethylation at TEAD1 binding site facilitates the transcriptional activation of PRAME. Inhibition of PRAME restrains the tumor metastasis via downregulation of epithelial-mesenchymal transition-related genes. We also identify that enrichment of AT2 cells with higher copy number variation burden, exhausted CD8 + T cells and Macro_SPP1, along with the reduced interaction between AT2 and immune cells, is essential for the formation of ecosystem in recurrent LUAD. Finally, multi-omics clustering could stratify the NSCLC patients into 4 subclusters with varying recurrence risk and subcluster-specific therapeutic vulnerabilities. Collectively, this study constitutes a promising resource enabling insights into the biological mechanisms and clinical management for post-operative recurrence of stage I NSCLC.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyao Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lujia Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changshu Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Qiu
- Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Fan L, Guo X, Washington MK, Shi J, Ness RM, Liu Q, Wen W, Huang S, Liu X, Cai Q, Zheng W, Coffey RJ, Shrubsole MJ, Su T. Yes-associated protein plays oncogenic roles in human sporadic colorectal adenomas. Carcinogenesis 2025; 46:bgaf007. [PMID: 39977302 PMCID: PMC11923420 DOI: 10.1093/carcin/bgaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 02/22/2025] Open
Abstract
The role of Hippo-Yes-associated protein (YAP) in human colorectal cancer (CRC) presents contradictory results. We examined the function of YAP in the early stages of CRC by quantitatively measuring the expression of phospho-YAPS127 (p-YAP) and five APC-related proteins in 145 sporadic adenomas from the Tennessee Colorectal Polyp Study, conducting APC sequencing for 114 adenomas, and analyzing YAP-correlated cancer pathways using gene expression data from 326 adenomas obtained from Gene Expression Omnibus. The p-YAP expression was significantly correlated with YAP expression (r = 0.53, P < .0001) and nuclear β-catenin (r = 0.26, P = .0018) in adenoma tissues. Both p-YAP and nuclear β-catenin were associated with APC mutations (P = .05). A strong association was observed between p-YAP overexpression and advanced adenoma odds (OR = 12.62, 95% CI = 4.57-34.86, P trend < .001), which persisted after adjusting for covariates and biomarkers (OR = 12.31, 95% CI = 3.78-40.10, P trend < .0001). P-YAP exhibited a sensitivity of 77.4% and specificity of 78.2% in defining advanced versus nonadvanced adenomas. Additionally, synergistic interaction was noted between p-YAP positivity and nuclear β-catenin on advanced adenomas (OR = 16.82, 95% CI = 4.41-64.08, P < .0001). YAP-correlated genes were significantly enriched in autophagy, unfolded protein response, and sirtuin pathways showing predominantly pro-tumorigenic alterations. Collectively, YAP plays an oncogenic role in interacting with Wnt as well as other cancer pathways within human sporadic adenomas. P-YAP could be a potential biomarker for human high-risk sporadic adenomas.
Collapse
Affiliation(s)
- Lei Fan
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
| | - Xingyi Guo
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Mary K Washington
- Department of Pathology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Jiajun Shi
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Reid M Ness
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Qi Liu
- Center for Quantitative Sciences and Department of Biostatistics, Vanderbilt University School of Medicine, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Shuya Huang
- Department of Breast Surgery, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, Shandong 250031, China
| | - Xiao Liu
- Center for Quantitative Sciences and Department of Biostatistics, Vanderbilt University School of Medicine, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Robert J Coffey
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Cell and Development Biology, Vanderbilt University, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Martha J Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| | - Timothy Su
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, United States
- GRECC, Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, TN 37212, United States
| |
Collapse
|
11
|
Joshi A, Bhaskar N, Pearson JD. Neuroendocrine Transformation as a Mechanism of Resistance to Targeted Lung Cancer Therapies: Emerging Mechanisms and Their Therapeutic Implications. Cancers (Basel) 2025; 17:260. [PMID: 39858043 PMCID: PMC11763869 DOI: 10.3390/cancers17020260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, highlighting a major clinical challenge. Lung cancer is broadly classified into two histologically distinct subtypes, termed small cell lung cancer (SCLC) or non-small cell lung cancer (NSCLC). Identification of various oncogenic drivers of NSCLC has facilitated the development of targeted therapies that have dramatically improved patient outcomes. However, acquired resistance to these targeted therapies is common, which ultimately results in patient relapse. Several on-target and off-target resistance mechanisms have been described for targeted therapies in NSCLC. One common off-target mechanism of resistance to these therapies is histological transformation of the initial NSCLC into SCLC, a highly aggressive form of lung cancer that exhibits neuroendocrine histology. This mechanism of resistance presents a significant clinical challenge, since there are very few treatments available for these relapsed patients. Although the phenomenon of NSCLC-to-SCLC transformation was described almost 20 years ago, only recently have we begun to understand the mechanisms underlying this therapy-driven response. These recent discoveries will be key to identifying novel biomarkers and therapeutic strategies to improve outcomes of patients that undergo NSCLC-to-SCLC transformation. Here, we highlight these recent advances and discuss the potential therapeutic strategies that they have uncovered to target this mechanism of resistance.
Collapse
Affiliation(s)
- Asim Joshi
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Nivitha Bhaskar
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Joel D. Pearson
- Department of Pharmacology & Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 0T6, Canada; (A.J.); (N.B.)
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
12
|
Duan J, Li H, Zhang J, Xu H, Gao J, Cai M, Pan Y, Shi Y, Wang H. PIEZO1 Affects Cell Growth and Migration via Microfilament-Mediated YAP trans-Latitudinal Regulation. Anal Chem 2025; 97:147-156. [PMID: 39729436 DOI: 10.1021/acs.analchem.4c03420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
Environmental mechanical forces, such as cell membrane stress, cell extrusion, and stretch, have been proven to affect cell growth and migration. Piezo1, a mechanosensitive channel protein, responds directly to endogenous or exogenous mechanical stimuli. Here, we explored the Piezo1 distribution and microfilament morphological changes induced by mechanical forces in the tumor and normal cells. In addition, Piezo1 activation in tumor cells resulted in the nuclear accumulation of YAP, whereas nuclear export of YAP and microfilament depolymerization occurred with the prolonged activation, while a removal stimulation restored the YAP distribution and microfilament polymerization. Combining the morphological changes of the microfilament under Piezo1 activation and the function of YAP in regulating cell growth and development, we suggest that Piezo1 senses changes in environmental mechanical forces and regulates YAP distribution through the microfilament cytoskeleton network, which in turn affects the growth and migration more obviously in tumor cells rather than normal cells. Our results are essential for understanding the trans-latitudinal transmission of mechanical forces and exploring the role of environmental mechanical forces in tumor therapy.
Collapse
Affiliation(s)
- Jiawei Duan
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongru Li
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yangang Pan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yan Shi
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongda Wang
- University of Science and Technology of China, Hefei, Anhui 230026, China
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| |
Collapse
|
13
|
Peng H, Chao Z, Wang Z, Hao X, Xi Z, Ma S, Guo X, Zhang J, Zhou Q, Qu G, Gao Y, Luo J, Wang Z, Wang J, Li L. Biomechanics in the tumor microenvironment: from biological functions to potential clinical applications. Exp Hematol Oncol 2025; 14:4. [PMID: 39799341 PMCID: PMC11724500 DOI: 10.1186/s40164-024-00591-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Immune checkpoint therapies have spearheaded drug innovation over the last decade, propelling cancer treatments toward a new era of precision therapies. Nonetheless, the challenges of low response rates and prevalent drug resistance underscore the imperative for a deeper understanding of the tumor microenvironment (TME) and the pursuit of novel targets. Recent findings have revealed the profound impacts of biomechanical forces within the tumor microenvironment on immune surveillance and tumor progression in both murine models and clinical settings. Furthermore, the pharmacological or genetic manipulation of mechanical checkpoints, such as PIEZO1, DDR1, YAP/TAZ, and TRPV4, has shown remarkable potential in immune activation and eradication of tumors. In this review, we delved into the underlying biomechanical mechanisms and the resulting intricate biological meaning in the TME, focusing mainly on the extracellular matrix, the stiffness of cancer cells, and immune synapses. We also summarized the methodologies employed for biomechanical research and the potential clinical translation derived from current evidence. This comprehensive review of biomechanics will enhance the understanding of the functional role of biomechanical forces and provide basic knowledge for the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Hao Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zefeng Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaodong Hao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Zirui Xi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Xiangdong Guo
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, 810001, Qinghai, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
- The Second Clinical School, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
- Taikang Tongji (Wuhan) Hospital, 420060, Wuhan, China.
| | - Jing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430300, China.
| |
Collapse
|
14
|
Rito T, Libby ARG, Demuth M, Domart MC, Cornwall-Scoones J, Briscoe J. Timely TGFβ signalling inhibition induces notochord. Nature 2025; 637:673-682. [PMID: 39695233 PMCID: PMC11735409 DOI: 10.1038/s41586-024-08332-w] [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: 02/27/2023] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
The formation of the vertebrate body involves the coordinated production of trunk tissues from progenitors located in the posterior of the embryo. Although in vitro models using pluripotent stem cells replicate aspects of this process1-10, they lack crucial components, most notably the notochord-a defining feature of chordates that patterns surrounding tissues11. Consequently, cell types dependent on notochord signals are absent from current models of human trunk formation. Here we performed single-cell transcriptomic analysis of chick embryos to map molecularly distinct progenitor populations and their spatial organization. Guided by this map, we investigated how differentiating human pluripotent stem cells develop a stereotypical spatial organization of trunk cell types. We found that YAP inactivation in conjunction with FGF-mediated MAPK signalling facilitated WNT pathway activation and induced expression of TBXT (also known as BRA). In addition, timely inhibition of WNT-induced NODAL and BMP signalling regulated the proportions of different tissue types, including notochordal cells. This enabled us to create a three-dimensional model of human trunk development that undergoes morphogenetic movements, producing elongated structures with a notochord and ventral neural and mesodermal tissues. Our findings provide insights into the mechanisms underlying vertebrate notochord formation and establish a more comprehensive in vitro model of human trunk development. This paves the way for future studies of tissue patterning in a physiologically relevant environment.
Collapse
Affiliation(s)
- Tiago Rito
- The Francis Crick Institute, London, UK.
| | | | | | | | | | | |
Collapse
|
15
|
Yin H, Jiang D, Li Y, Chen W, Zhang J, Yang X, Hu J, Wei H. KDELR1 regulates chondrosarcoma drug resistance and malignant behavior through Intergrin-Hippo-YAP1 axis. Cell Death Dis 2024; 15:928. [PMID: 39715773 DOI: 10.1038/s41419-024-07264-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Chondrosarcoma (CS) is the second most common primary bone malignancy, known for its unique transcriptional landscape that renders most CS subtypes resistant to chemotherapy, including neoadjuvant chemotherapy commonly used in osteosarcoma (OS) treatment. Understanding the transcriptional landscape of CS and the mechanisms by which key genes contribute to chemotherapy resistance could be a crucial step in overcoming this challenge. To address this, we developed a single-cell transcriptional map of CS, comparing it with OS and normal cancellous bone. Our analysis revealed a specific increase in KDEL receptor 1 (KDELR1) expression in CS, which was closely associated with CS prognosis, tumor aggressiveness, and drug resistance. KDELR1 plays a key role in regulating membrane protein processing and secretion, as well as contributing to tumor extracellular matrix (ECM) formation and drug resistance. Further investigation using mass spectrometry proteomics and transcriptomics uncovered KDELR1's involvement in modulating the Hippo-YAP pathway activity in CS cells. The KDELR1-Integrin-PLCγ-YAP1 axis emerges as a critical process mediating drug resistance and malignant behavior in CS, offering novel insights and potential therapeutic targets for CS treatment.
Collapse
Affiliation(s)
- Huabin Yin
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85 Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Dongjie Jiang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Yongai Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85 Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Wenjun Chen
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jie Zhang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Xinghai Yang
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Jinbo Hu
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Haifeng Wei
- Spinal Tumor Center, Department of Orthopaedic Oncology, No.905 Hospital of PLA Navy, Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
16
|
Kotagiri S, Blazanin N, Xi Y, Han Y, Qudratullah M, Liang X, Wang Y, Pandey P, Mazhar H, Lam TN, Singh AK, Wang J, Lissanu Y. Enhancer reprogramming underlies therapeutic utility of a SMARCA2 degrader in SMARCA4 mutant cancer. Cell Chem Biol 2024; 31:2069-2084.e9. [PMID: 39378885 DOI: 10.1016/j.chembiol.2024.09.004] [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: 10/12/2023] [Revised: 07/02/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
Genomic studies have identified frequent mutations in subunits of the SWI/SNF (switch/sucrose non-fermenting) chromatin remodeling complex including SMARCA4 and ARID1A in non-small cell lung cancer (NSCLC). Genetic evidence indicates that the paralog SMARCA2 is synthetic lethal to SMARCA4 suggesting SMARCA2 is a valuable therapeutic target. However, the discovery of selective inhibitors of SMARCA2 has been challenging. Here, we utilized structure-activity relationship (SAR) studies to develop YD23, a potent and selective proteolysis targeting chimera (PROTAC) targeting SMARCA2. Mechanistically, we show that SMARCA2 degradation induces reprogramming of the enhancer landscape in SMARCA4-mutant cells with loss of chromatin accessibility at enhancers of genes involved in cell proliferation. Furthermore, we identified YAP/TEADas key partners to SMARCA2 in driving growth of SMARCA4-mutant cells. Finally, we show that YD23 has potent tumor growth inhibitory activity in SMARCA4-mutant xenografts. These findings provide the mechanistic basis for development of SMARCA2 degraders as synthetic lethal therapeutics against SMARCA4-mutant lung cancers.
Collapse
Affiliation(s)
- Sasikumar Kotagiri
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas Blazanin
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanxin Xi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanyan Han
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Md Qudratullah
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaobing Liang
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yawen Wang
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Poonam Pandey
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hira Mazhar
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Truong Nguyen Lam
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anand Kamal Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yonathan Lissanu
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Guo Y, Dupart M, Irondelle M, Peraldi P, Bost F, Mazure NM. YAP1 modulation of primary cilia-mediated ciliogenesis in 2D and 3D prostate cancer models. FEBS Lett 2024; 598:3071-3086. [PMID: 39424416 DOI: 10.1002/1873-3468.15029] [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: 05/07/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The primary cilium, a non-motile organelle present in most human cells, plays a crucial role in detecting microenvironmental changes and regulating intracellular signaling. Its dysfunction is linked to various diseases, including cancer. We explored the role of ciliated cells in prostate cancer by using Gefitinib and Jasplakinolide compounds to induce ciliated cells in both normal and tumor-like prostate cell lines. We assessed GLI1 and IFT20 expression and investigated YAP1 protein's role, which is implicated in primary cilium regulation. Finally, we examined these compounds in 3D cell models, aiming to simulate in vivo conditions. Our study highlights YAP1 as a potential target for novel genetic models to understand the primary cilium's role in mediating resistance to anticancer treatments.
Collapse
Affiliation(s)
- Yingbo Guo
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Mathilde Dupart
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
- IRCAN, Université Côte d'Azur, Nice Cedex 02, France
| | - Marie Irondelle
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
| | - Pascal Peraldi
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Frederic Bost
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| | - Nathalie M Mazure
- INSERM U1065, C3M, Université Côte d'Azur, Nice Cedex 03, France
- Equipe Labellisée Ligue Contre le Cancer, Xxxxx, France
| |
Collapse
|
18
|
Koroleva OA, Kurkin AV, Shtil AA. The Hippo pathway as an antitumor target: time to focus on. Expert Opin Investig Drugs 2024; 33:1177-1185. [PMID: 39592955 DOI: 10.1080/13543784.2024.2432395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
INTRODUCTION The Hippo signaling governs the expression of genes critically important for cell proliferation and survival. The components of this pathway are considered antitumor drug targets. However, the design of Hippo inhibitors is a challenge given the complexity of the network and redundancy of its elements. AREAS COVERED We review the current state-of-the-art in the structure of the Hippo pathway, the microenvironment-induced extracellular cues, the strategies to design pharmacological instruments for inactivation of the Hippo signaling using small molecular weight modulators, as well as the results of initial clinical trials. EXPERT OPINION One special characteristic of the Hippo signaling is the adverse role of phosphorylation: opposite to classical kinase cascades that activate the transcription factors, the Hippo kinases retain their partners in a transcriptionally inactive state. Therefore, approaches for pharmacological or genetic inhibition of Hippo protein kinases are counterproductive. The developing alternatives such as disruption of protein-protein interactions or PROTAC techniques are straightforward for preventing the Hippo signaling in cancer therapy.
Collapse
Affiliation(s)
- Olga A Koroleva
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander V Kurkin
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Alexander A Shtil
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
- Institute of Carcinogenesis, Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| |
Collapse
|
19
|
Han H, Huang Z, Xu C, Seo G, An J, Yang B, Liu Y, Lan T, Yan J, Ren S, Xu Y, Xiao D, Yan JK, Ahn C, Fishman DA, Meng Z, Guan KL, Qi R, Luo R, Wang W. Functional annotation of the Hippo pathway somatic mutations in human cancers. Nat Commun 2024; 15:10106. [PMID: 39572544 PMCID: PMC11582751 DOI: 10.1038/s41467-024-54480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
The Hippo pathway is commonly altered in cancer initiation and progression; however, exactly how this pathway becomes dysregulated to promote human cancer development remains unclear. Here we analyze the Hippo somatic mutations in the human cancer genome and functionally annotate their roles in targeting the Hippo pathway. We identify a total of 85 loss-of-function (LOF) missense mutations for Hippo pathway genes and elucidate their underlying mechanisms. Interestingly, we reveal zinc-finger domain as an integral structure for MOB1 function, whose LOF mutations in head and neck cancer promote tumor growth. Moreover, the schwannoma/meningioma-derived NF2 LOF mutations not only inhibit its tumor suppressive function in the Hippo pathway, but also gain an oncogenic role for NF2 by activating the VANGL-JNK pathway. Collectively, our study not only offers a rich somatic mutation resource for investigating the Hippo pathway in human cancers, but also provides a molecular basis for Hippo-based cancer therapy.
Collapse
Affiliation(s)
- Han Han
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| | - Zhen Huang
- Chemical and Materials Physics Graduate Program, University of California, Irvine, Irvine, CA, USA
| | - Congsheng Xu
- Department of Chemistry and Shenzhen Grubbs Institute, Guangdong Provincial Key Laboratory of Catalysis, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Gayoung Seo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jeongmin An
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Yuhan Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Tian Lan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jiachen Yan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Shanshan Ren
- Department of Chemistry and Shenzhen Grubbs Institute, Guangdong Provincial Key Laboratory of Catalysis, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yue Xu
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Di Xiao
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Jonathan K Yan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Claire Ahn
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Dmitry A Fishman
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Ruxi Qi
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Ray Luo
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
20
|
Chen H, Cui H, Liu W, Li BW, Tian Z, Zhao YY, Yu GT. Manganese drives ferroptosis of cancer cells via YAP/TAZ phase separation activated ACSL4 in OSCC. Oral Dis 2024; 30:4898-4908. [PMID: 38462885 DOI: 10.1111/odi.14925] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE Ferroptosis has been defined as a novel form of regulated cell death characterized by iron-dependent lipid peroxidation. Manganese has been used to induce ferroptosis in cancer cells recently. This study aims to investigate whether manganese can induce ferroptosis in oral squamous cell carcinoma (OSCC) and the underlying biological mechanisms. MATERIALS AND METHODS Cancer cells with or without manganese treatment were analyzed by RNA-sequencing to identify ferroptosis-related genes. Next, the activation of YAP/TAZ/ACSL4-ferroptosis signaling pathway was detected. Bioinformatic analysis and immunofluorescence assay were used to explore the phase separation of YAP/TAZ. Finally, specimens of OSCC patients were applied to analyze the clinical significance of YAP/TAZ/ACSL4. RESULTS RNA-sequencing analysis showed the ferroptosis-related genes and YAP/TAZ were upregulated after manganese treatment. The results of immunofluorescence, ELISA, western blotting, etc. further confirmed that manganese-induced ferroptosis depends on YAP/TAZ/ACSL4 signaling pathway. Moreover, the activation of ACSL4 was achieved by YAP/TAZ phase separation. The survival analysis in OSCC specimen suggested that the higher level of YAP/TAZ-ACSL4 axis expression indicates longer survival. CONCLUSIONS Manganese induces YAP/TAZ phase separation and subsequent ACSL4 activation via YAP/TAZ nuclear translocation, which facilitates ferroptosis of OSCC. Then YAP/TAZ-ACSL4 axis can be used as a potential prognostic predictor of OSCC patients.
Collapse
Affiliation(s)
- Hao Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo-Wen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yu-Yue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Guang-Tao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Jiang J, Han D, Wang J, Wen W, Zhang R, Qin W. Neuroendocrine transdifferentiation in human cancer: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e761. [PMID: 39372390 PMCID: PMC11450264 DOI: 10.1002/mco2.761] [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: 07/17/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 10/08/2024] Open
Abstract
Neuroendocrine transdifferentiation (NEtD), also commonly referred to as lineage plasticity, emerges as an acquired resistance mechanism to molecular targeted therapies in multiple cancer types, predominately occurs in metastatic epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors and metastatic castration-resistant prostate cancer treated with androgen receptor targeting therapies. NEtD tumors are the lethal cancer histologic subtype with unfavorable prognosis and limited treatment. A comprehensive understanding of molecular mechanism underlying targeted-induced plasticity could greatly facilitate the development of novel therapies. In the past few years, increasingly elegant studies indicated that NEtD tumors share key the convergent genomic and phenotypic characteristics irrespective of their site of origin, but also embrace distinct change and function of molecular mechanisms. In this review, we provide a comprehensive overview of the current understanding of molecular mechanism in regulating the NEtD, including genetic alterations, DNA methylation, histone modifications, dysregulated noncoding RNA, lineage-specific transcription factors regulation, and other proteomic alterations. We also provide the current management of targeted therapies in clinical and preclinical practice.
Collapse
Affiliation(s)
- Jun Jiang
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
- Department of Health Service, Base of Health ServiceAir Force Medical UniversityXi'anChina
| | - Donghui Han
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| | - Jiawei Wang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, and National Translational Science Center for Molecular MedicineAir Force Medical UniversityXi'anChina
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical ResearchNorthwestern Polytechnical UniversityXi'anChina
| | - Rui Zhang
- State Key Laboratory of Cancer BiologyDepartment of ImmunologyAir Force Medical UniversityXi'anChina
| | - Weijun Qin
- Department of UrologyXijing HospitalAir Force Medical UniversityXi'anChina
| |
Collapse
|
22
|
Abegunde SO, Grieve S, Reiman T. TAZ downregulated ANXA1 expression to modulate myeloma cell interactions with bone marrow mesenchymal stromal cells. Exp Hematol 2024; 138:104282. [PMID: 39032857 DOI: 10.1016/j.exphem.2024.104282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/01/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
We and others have previously shown that TAZ plays a tumor suppressive role in multiple myeloma. However, recent reports suggest that molecular crosstalk between the myeloma cells and bone marrow stromal components contributes to the myeloma cell survival and drug resistance. These reports further point to reciprocal interaction via adhesion molecules as the most prominent mechanism of intercellular crosstalk between myeloma cells and bone marrow mesenchymal stromal cells (BM-MSCs). YAP/TAZ silencing/expression has been shown to correlate across all cancers with a set of adhesion/extracellular matrix proteins. Therefore, we hypothesized that TAZ may regulate myeloma cell interaction with BM stromal cells by influencing the expression of distinct cell adhesion signatures. We used previously established TAZ myeloma cell line models, including DELTA47-pLENTI or TAZ knockout DELTA47 cells cocultured with or without BM-MSCs, as our study models. Using RNA sequencing analysis, we performed the first comprehensive screen for cell adhesion-related transcriptional targets of TAZ in multiple myeloma (MM). In doing so, we uncovered an enrichment of cell adhesion-related genes in TAZ knockout DELTA47 cells relatively to pLENTI-DELTA47 cells, including 11 genes with log2 fold change > 2 (p < 0.05), namely, ANXA1, ADGRL2, NCAM1, NCAM2, ADGRL3, CXADR, ALCAM, JAM2, KIRREL1, KIRREL2, and ADGRG7, suggesting possible relationship with TAZ. We validated ANXA1 as a bona fide target of TAZ in MM. We show that TAZ represses myeloma cell migration and interaction with BM-MSCs by transcriptionally downregulating ANXA1 expression via TEAD-dependent mechanism. Our data provide new insights into the understanding of the role of TAZ in the intercellular communication signals between myeloma cells and BM-MSCs. Our findings also suggest that ANXA1 represents a putative cell adhesion target to attenuate BM-MSC driven, tumor-promoting interaction with myeloma cells.
Collapse
Affiliation(s)
- Samuel O Abegunde
- Department of Biology, University of New Brunswick, Saint John, NB, Canada; Dalhousie Medicine NB, Saint John, NB, Canada; Vancouver General Hospital, Vancouver, BC, Canada.
| | | | - Tony Reiman
- Department of Biology, University of New Brunswick, Saint John, NB, Canada; Dalhousie Medicine NB, Saint John, NB, Canada; Saint John Regional Hospital, Saint John, NB, Canada.
| |
Collapse
|
23
|
Niu X, Liu W, Zhang Y, Liu J, Zhang J, Li B, Qiu Y, Zhao P, Wang Z, Wang Z. Cancer plasticity in therapy resistance: Mechanisms and novel strategies. Drug Resist Updat 2024; 76:101114. [PMID: 38924995 DOI: 10.1016/j.drup.2024.101114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Therapy resistance poses a significant obstacle to effective cancer treatment. Recent insights into cell plasticity as a new paradigm for understanding resistance to treatment: as cancer progresses, cancer cells experience phenotypic and molecular alterations, corporately known as cell plasticity. These alterations are caused by microenvironment factors, stochastic genetic and epigenetic changes, and/or selective pressure engendered by treatment, resulting in tumor heterogeneity and therapy resistance. Increasing evidence suggests that cancer cells display remarkable intrinsic plasticity and reversibly adapt to dynamic microenvironment conditions. Dynamic interactions between cell states and with the surrounding microenvironment form a flexible tumor ecosystem, which is able to quickly adapt to external pressure, especially treatment. Here, this review delineates the formation of cancer cell plasticity (CCP) as well as its manipulation of cancer escape from treatment. Furthermore, the intrinsic and extrinsic mechanisms driving CCP that promote the development of therapy resistance is summarized. Novel treatment strategies, e.g., inhibiting or reversing CCP is also proposed. Moreover, the review discusses the multiple lines of ongoing clinical trials globally aimed at ameliorating therapy resistance. Such advances provide directions for the development of new treatment modalities and combination therapies against CCP in the context of therapy resistance.
Collapse
Affiliation(s)
- Xing Niu
- China Medical University, Shenyang, Liaoning 110122, China; Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, 999077, Hong Kong, China
| | - Wenjing Liu
- Medical Oncology Department of Thoracic Cancer (2), Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yinling Zhang
- Department of Oncology Radiotherapy 1, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266042, China
| | - Jing Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Peng Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhongmiao Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Zhe Wang
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
24
|
Pearson JD, Huang K, Dela Pena LG, Ducarouge B, Mehlen P, Bremner R. Netrin-1 and UNC5B Cooperate with Integrins to Mediate YAP-Driven Cytostasis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2374-2383. [PMID: 39172021 PMCID: PMC11384508 DOI: 10.1158/2767-9764.crc-24-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/24/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
Opposite expression and pro- or anti-cancer function of YAP and its paralog TAZ/WWTR1 stratify cancers into binary YAPon and YAPoff classes. These transcriptional coactivators are oncogenic in YAPon cancers. In contrast, YAP/TAZ are silenced epigenetically along with their integrin and extracellular matrix adhesion target genes in neural and neuroendocrine YAPoff cancers (e.g., small cell lung cancer, retinoblastoma). Forced YAP/TAZ expression induces these targets, causing cytostasis in part through Integrin-αV/β5, independent of the integrin-binding RGD ligand. Other effectors of this anticancer YAP function are unknown. Here, using clustered regularly interspaced short palindromic repeats (CRISPR) screens, we link the Netrin receptor UNC5B to YAP-induced cytostasis in YAPoff cancers. Forced YAP expression induces UNC5B through TEAD DNA-binding partners, as either TEAD1/4-loss or a YAP mutation that disrupts TEAD-binding (S94A) blocks, whereas a TEAD-activator fusion (TEAD(DBD)-VP64) promotes UNC5B induction. Ectopic YAP expression also upregulates UNC5B relatives and their netrin ligands in YAPoff cancers. Netrins are considered protumorigenic, but knockout and peptide/decoy receptor blocking assays reveal that in YAPoff cancers, UNC5B and Netrin-1 can cooperate with integrin-αV/β5 to mediate YAP-induced cytostasis. These data pinpoint an unsuspected Netrin-1/UNC5B/integrin-αV/β5 axis as a critical effector of YAP tumor suppressor activity. SIGNIFICANCE Netrins are widely perceived as procancer proteins; however, we uncover an anticancer function for Netrin-1 and its receptor UNC5B.
Collapse
Affiliation(s)
- Joel D. Pearson
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Paul Albrechtsen Research Institute CancerCare Manitoba & Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada.
| | - Katherine Huang
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, Canada.
| | - Louis G. Dela Pena
- Paul Albrechtsen Research Institute CancerCare Manitoba & Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada.
| | | | - Patrick Mehlen
- Netris Pharma, Centre Léon Bérard 28 Rue Laennec, Lyon, France.
- Apoptosis, Cancer and Development Laboratory-Equipe labellisée ‘La Ligue’, LabEX DEVweCAN, Centre de Recherche en Cancérologie de Lyon, Lyon, France.
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
25
|
Zhang P, Li P, Tang M, Gimple RC, Huang L, Yue J, Shen Q, Du Y, Zhang Q, Yang Z, He H, Yang K, Zhao L, Zhou S. The genomic and immunogenomic landscape of mechanics pathway informs clinical prognosis and response to mechanotherapy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1549-1562. [PMID: 39037695 DOI: 10.1007/s11427-024-2622-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/07/2024] [Indexed: 07/23/2024]
Abstract
Mechanics shape cell and tissue plasticity and maintain their homeostasis. In cancers, mechanical signals regulate cancer hallmarks via mechanotransduction pathways, such as proliferation, metastasis and metabolic reprogramming. However, comprehensive characterization of mechanotransduction pathway genes and their clinical relevance across different cancer types remains untouched. Herein, we systematically portrayed the alterations of mechanotransduction pathway genes across 31 cancer types using The Cancer Genome Atlas (TCGA) databases. All the cancer types could be categorized into 6 subtypes based upon the transcriptional pattern of mechanics pathway genes. Each subtype has its own unique molecular expression pattern, mutation landscapes, immune infiltrates, and patient clinical outcome. We further found that the responses of two subtypes of cancers, one with the optimal outcome and the other with the worst prognosis, to a classical mechanotherapeutic agent (Fasudil, RhoA/ROCK inhibitor) were totally different, indicating that our cancer stratification system based upon mechanotransduction pathway genes could inform clinical responses of patients to mechanotherapeutic agents. Collectively, our study provides a novel pan-cancer landscape of the mechanotransduction pathways and underscores its potential clinical significance in the prediction of clinical prognosis and therapeutic responses to mechanotherapy among cancer patients.
Collapse
Affiliation(s)
- Peidong Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Peiwei Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Muya Tang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Ryan C Gimple
- Physician Scientist Training Program, Department of Medicine, Washington University School of Medicine, St. Louis, 63110, USA
| | - Liang Huang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Jing Yue
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Qiuhong Shen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Yiwei Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Zhengnan Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Haihuai He
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, 44106, USA.
| | - Linjie Zhao
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
26
|
Li S, Li X, Yang YB, Wu SF. YAP/TAZ-TEAD activity promotes the malignant transformation of cervical intraepithelial neoplasia through enhancing the characteristics and Warburg effect of cancer stem cells. Apoptosis 2024; 29:1198-1210. [PMID: 38553612 PMCID: PMC11263238 DOI: 10.1007/s10495-023-01935-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2023] [Indexed: 07/23/2024]
Abstract
A number of studies have confirmed that Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ)-transcriptional enhanced associate domain (TEAD) activity is the driver of cancer development. However, the role and mechanism of the YAP/TAZ-TEAD pathway in cervical intraepithelial neoplasia (CIN) remain to be clarified. Therefore, this study was designed to observe the effect of YAP/TAZ-TEAD activity on the development of CIN and provide new ideas for the diagnosis and treatment of CIN. Firstly, cervical tissues were collected from CIN patients in different stages [CIN grade 1 (CIN1) tissue, CIN grade 2/3 (CIN 2/3) and squamous cell carcinoma (SCC)] and healthy volunteers. Next, the expression levels of YAP, TAZ and TEAD in cervical tissues and cells were observed by immunohistochemistry, qRT-PCR and western blot. Besides, Z172 and Z183 cells were transfected with siRNA-YAP/TAZ (si-YAP/TAZ) and YAP/TAZ overexpression vector (YAP-5SA). Also, Z172 cells were co-transfected with YAP-5SA and si-TEAD2/4. Subsequently, the stemness characteristics, glycolysis level and malignant transformation of cells in each group were observed by sphere-formation assay, commercial kit, MTT, Transwell, scratch experiment, xenotransplantation and western blot.The expression of YAP, TAZ and TEAD increased significantly in cervical cancer tissue and cell line at the stage of CIN2/3 and SCC. When YAP/TAZ was knocked down, the stemness characteristics, glycolysis level and malignant transformation of cancer cells were notably inhibited; while activating YAP/TAZ exhibited a completely opposite result. In addition, activating YAP/TAZ and knocking down the TEAD expression at the same time significant weakened the effect of activated YAP/TAZ signal on precancerous cells and reduced inhibitory effect of knocking down TEAD alone. YAP/TAZ-TEAD signal activates the characteristics and Warburg effect of cancer stem cells, thereby promoting the malignant transformation of CIN.
Collapse
MESH Headings
- Humans
- Female
- Transcription Factors/genetics
- Transcription Factors/metabolism
- YAP-Signaling Proteins/metabolism
- YAP-Signaling Proteins/genetics
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Uterine Cervical Neoplasms/genetics
- Uterine Cervical Neoplasms/metabolism
- Uterine Cervical Neoplasms/pathology
- Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Uterine Cervical Dysplasia/pathology
- Uterine Cervical Dysplasia/genetics
- Uterine Cervical Dysplasia/metabolism
- Animals
- Trans-Activators/genetics
- Trans-Activators/metabolism
- TEA Domain Transcription Factors/metabolism
- Cell Line, Tumor
- Mice
- Warburg Effect, Oncologic
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Cell Proliferation/genetics
- Mice, Nude
- Gene Expression Regulation, Neoplastic
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
Collapse
Affiliation(s)
- Shu Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yong-Bin Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Su-Fang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
27
|
Dehghanian F, Ghahnavieh LE, Nilchi AN, Khalilian S, Joonbakhsh R. Breast cancer drug resistance: Decoding the roles of Hippo pathway crosstalk. Gene 2024; 916:148424. [PMID: 38588933 DOI: 10.1016/j.gene.2024.148424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/13/2024] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
The most significant factors that lead to cancer-related death in breast cancer (BC) patients include drug resistance, migration, invasion, and metastasis. Several signaling pathways are involved in the development of BC. The different types of BC are initially sensitive to chemotherapy, and drug resistance can occur through multiple molecular mechanisms. Regardless of developing targeted Therapy, due to the heterogenic nature and complexity of drug resistance, it is a major clinical challenge with the low survival rate in BC patients. The deregulation of several signaling pathways, particularly the Hippo pathway (HP), is one of the most recent findings about the molecular mechanisms of drug resistance in BC, which are summarized in this review. Given that HP is one of the recent cancer research hotspots, this review focuses on its implication in BC drug resistance. Unraveling the different molecular basis of HP through its crosstalk with other signaling pathways, and determining the effectiveness of HP inhibitors can provide new insights into possible therapeutic strategies for overcoming chemoresistance in BC.
Collapse
Affiliation(s)
- Fariba Dehghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran.
| | - Laleh Ebrahimi Ghahnavieh
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| | - Amirhossein Naghsh Nilchi
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| | - Sheyda Khalilian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| | - Rezvan Joonbakhsh
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, HezarJarib Street, Isfahan 81746-73441, Iran
| |
Collapse
|
28
|
Liu J, Fu J, Fu P, Liu M, Liu Z, Song B. Pitavastatin sensitizes the EGFR-TKI associated resistance in lung cancer by inhibiting YAP/AKT/BAD-BCL-2 pathway. Cancer Cell Int 2024; 24:224. [PMID: 38943199 PMCID: PMC11214206 DOI: 10.1186/s12935-024-03416-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 06/22/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as "statins" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated. METHODS Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model. RESULTS PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway. CONCLUSION Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.
Collapse
Affiliation(s)
- Jie Liu
- Cancer Center, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Jialei Fu
- Shandong Academy of Chinese Medicine, Jinan, China
| | - Ping Fu
- Department of Chemotherapy, Jinan Zhangqiu District People's Hospital, Jinan, China
| | - Menghan Liu
- Clinical Medical College, Shandong First Medical University, Jinan, China
| | - Zining Liu
- Department of Nuclear Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China.
| | - Bao Song
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
29
|
Zhu C, Zheng R, Han X, Tang Z, Li F, Hu X, Lin R, Shen J, Pei Q, Wang R, Wei G, Peng Z, Chen W, Liang Z, Zhou Y. Knockout of integrin αvβ6 protects against renal inflammation in chronic kidney disease by reduction of pro-inflammatory macrophages. Cell Death Dis 2024; 15:397. [PMID: 38844455 PMCID: PMC11156928 DOI: 10.1038/s41419-024-06785-5] [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: 02/01/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
Integrin αvβ6 holds promise as a therapeutic target for organ fibrosis, yet targeted therapies are hampered by concerns over inflammatory-related side effects. The role of αvβ6 in renal inflammation remains unknown, and clarifying this issue is crucial for αvβ6-targeted treatment of chronic kidney disease (CKD). Here, we revealed a remarkable positive correlation between overexpressed αvβ6 in proximal tubule cells (PTCs) and renal inflammation in CKD patients and mouse models. Notably, knockout of αvβ6 not only significantly alleviated renal fibrosis but also reduced inflammatory responses in mice, especially the infiltration of pro-inflammatory macrophages. Furthermore, conditional knockout of αvβ6 in PTCs in vivo and co-culture of PTCs with macrophages in vitro showed that depleting αvβ6 in PTCs suppressed the migration and pro-inflammatory differentiation of macrophages. Screening of macrophage activators showed that αvβ6 in PTCs activates macrophages via secreting IL-34. IL-34 produced by PTCs was significantly diminished by αvβ6 silencing, and reintroduction of IL-34 restored macrophage activities, while anti-IL-34 antibody restrained macrophage activities enhanced by αvβ6 overexpression. Moreover, RNA-sequencing of PTCs and verification experiments demonstrated that silencing αvβ6 in PTCs blocked hypoxia-stimulated IL-34 upregulation and secretion by inhibiting YAP expression, dephosphorylation, and nuclear translocation, which resulted in the activation of Hippo signaling. While application of a YAP agonist effectively recurred IL-34 production by PTCs, enhancing the subsequent macrophage migration and activation. Besides, reduced IL-34 expression and YAP activation were also observed in global or PTCs-specific αvβ6-deficient injured kidneys. Collectively, our research elucidates the pro-inflammatory function and YAP/IL-34/macrophage axis-mediated mechanism of αvβ6 in renal inflammation, providing a solid rationale for the use of αvβ6 inhibition to treat kidney inflammation and fibrosis.
Collapse
Affiliation(s)
- Changjian Zhu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Ruilin Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Xu Han
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Ziwen Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Feng Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Xinrong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Ruoni Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Jiani Shen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Qiaoqiao Pei
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Rong Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Guangyan Wei
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhenwei Peng
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| | - Zhou Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
| |
Collapse
|
30
|
Thatikonda V, Supper V, Wachter J, Kaya O, Kombara A, Bilgilier C, Ravichandran MC, Lipp JJ, Sharma R, Badertscher L, Boghossian AS, Rees MG, Ronan MM, Roth JA, Grosche S, Neumüller RA, Mair B, Mauri F, Popa A. Genetic dependencies associated with transcription factor activities in human cancer cell lines. Cell Rep 2024; 43:114175. [PMID: 38691456 DOI: 10.1016/j.celrep.2024.114175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 02/02/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024] Open
Abstract
Transcription factors (TFs) are important mediators of aberrant transcriptional programs in cancer cells. In this study, we focus on TF activity (TFa) as a biomarker for cell-line-selective anti-proliferative effects, in that high TFa predicts sensitivity to loss of function of a given gene (i.e., genetic dependencies [GDs]). Our linear-regression-based framework identifies 3,047 pan-cancer and 3,952 cancer-type-specific candidate TFa-GD associations from cell line data, which are then cross-examined for impact on survival in patient cohorts. One of the most prominent biomarkers is TEAD1 activity, whose associations with its predicted GDs are validated through experimental evidence as proof of concept. Overall, these TFa-GD associations represent an attractive resource for identifying innovative, biomarker-driven hypotheses for drug discovery programs in oncology.
Collapse
Affiliation(s)
- Venu Thatikonda
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria.
| | - Verena Supper
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Johannes Wachter
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Onur Kaya
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Anju Kombara
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Ceren Bilgilier
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | | | - Jesse J Lipp
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Rahul Sharma
- Myllia Biotechnology GmbH, Am Kanal 27, Vienna 1110, Austria
| | | | | | - Matthew G Rees
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah Grosche
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Ralph A Neumüller
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Barbara Mair
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Federico Mauri
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria
| | - Alexandra Popa
- Boehringer Ingelheim RCV GmbH & Co KG, Doktor-Boehringer-Gasse 5-11, Vienna 1120, Austria.
| |
Collapse
|
31
|
Lee DK, Oh J, Park HW, Gee HY. Anchorage Dependence and Cancer Metastasis. J Korean Med Sci 2024; 39:e156. [PMID: 38769921 PMCID: PMC11106561 DOI: 10.3346/jkms.2024.39.e156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
The process of cancer metastasis is dependent on the cancer cells' capacity to detach from the primary tumor, endure in a suspended state, and establish colonies in other locations. Anchorage dependence, which refers to the cells' reliance on attachment to the extracellular matrix (ECM), is a critical determinant of cellular shape, dynamics, behavior, and, ultimately, cell fate in nonmalignant and cancer cells. Anchorage-independent growth is a characteristic feature of cells resistant to anoikis, a programmed cell death process triggered by detachment from the ECM. This ability to grow and survive without attachment to a substrate is a crucial stage in the progression of metastasis. The recently discovered phenomenon named "adherent-to-suspension transition (AST)" alters the requirement for anchoring and enhances survival in a suspended state. AST is controlled by four transcription factors (IKAROS family zinc finger 1, nuclear factor erythroid 2, BTG anti-proliferation factor 2, and interferon regulatory factor 8) and can detach cells without undergoing the typical epithelial-mesenchymal transition. Notably, AST factors are highly expressed in circulating tumor cells compared to their attached counterparts, indicating their crucial role in the spread of cancer. Crucially, the suppression of AST substantially reduces metastasis while sparing primary tumors. These findings open up possibilities for developing targeted therapies that inhibit metastasis and emphasize the importance of AST, leading to a fundamental change in our comprehension of how cancer spreads.
Collapse
Affiliation(s)
- Dong Ki Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Jongwook Oh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul, Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
Abou Nader N, Charrier L, Meisnsohn MC, Banville L, Deffrennes B, St-Jean G, Boerboom D, Zamberlam G, Brind'Amour J, Pépin D, Boyer A. Lats1 and Lats2 regulate YAP and TAZ activity to control the development of mouse Sertoli cells. FASEB J 2024; 38:e23633. [PMID: 38690712 DOI: 10.1096/fj.202400346r] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.
Collapse
Affiliation(s)
- Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Laureline Charrier
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Marie-Charlotte Meisnsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Laurence Banville
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Bérengère Deffrennes
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - Julie Brind'Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
33
|
Liang H, Xu Y, Zhao J, Chen M, Wang M. Hippo pathway in non-small cell lung cancer: mechanisms, potential targets, and biomarkers. Cancer Gene Ther 2024; 31:652-666. [PMID: 38499647 PMCID: PMC11101353 DOI: 10.1038/s41417-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Lung cancer is the primary contributor to cancer-related deaths globally, and non-small cell lung cancer (NSCLC) constitutes around 85% of all lung cancer cases. Recently, the emergence of targeted therapy and immunotherapy revolutionized the treatment of NSCLC and greatly improved patients' survival. However, drug resistance is inevitable, and extensive research has demonstrated that the Hippo pathway plays a crucial role in the development of drug resistance in NSCLC. The Hippo pathway is a highly conserved signaling pathway that is essential for various biological processes, including organ development, maintenance of epithelial balance, tissue regeneration, wound healing, and immune regulation. This pathway exerts its effects through two key transcription factors, namely Yes-associated protein (YAP) and transcriptional co-activator PDZ-binding motif (TAZ). They regulate gene expression by interacting with the transcriptional-enhanced associate domain (TEAD) family. In recent years, this pathway has been extensively studied in NSCLC. The review summarizes a comprehensive overview of the involvement of this pathway in NSCLC, and discusses the mechanisms of drug resistance, potential targets, and biomarkers associated with this pathway in NSCLC.
Collapse
Affiliation(s)
- Hongge Liang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
34
|
Hosohama L, Tifrea DF, Nee K, Park SY, Wu J, Habowski AN, Van C, Seldin MM, Edwards RA, Waterman ML. Colorectal Cancer Stem Cell Subtypes Orchestrate Distinct Tumor Microenvironments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591144. [PMID: 38712298 PMCID: PMC11071458 DOI: 10.1101/2024.04.25.591144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Several classification systems have been developed to define tumor subtypes in colorectal cancer (CRC). One system proposes that tumor heterogeneity derives in part from distinct cancer stem cell populations that co-exist as admixtures of varying proportions. However, the lack of single cell resolution has prohibited a definitive identification of these types of stem cells and therefore any understanding of how each influence tumor phenotypes. Here were report the isolation and characterization of two cancer stem cell subtypes from the SW480 CRC cell line. We find these cancer stem cells are oncogenic versions of the normal Crypt Base Columnar (CBC) and Regenerative Stem Cell (RSC) populations from intestinal crypts and that their gene signatures are consistent with the "Admixture" and other CRC classification systems. Using publicly available single cell RNA sequencing (scRNAseq) data from CRC patients, we determine that RSC and CBC cancer stem cells are commonly co-present in human CRC. To characterize influences on the tumor microenvironment, we develop subtype-specific xenograft models and we define their tumor microenvironments at high resolution via scRNAseq. RSCs create differentiated, inflammatory, slow growing tumors. CBCs create proliferative, undifferentiated, invasive tumors. With this enhanced resolution, we unify current CRC patient classification schema with TME phenotypes and organization.
Collapse
Affiliation(s)
- Linzi Hosohama
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, California
| | - Delia F. Tifrea
- Department of Pathology & Laboratory Medicine, School of Medicine, University of California, Irvine, California
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | - Kevin Nee
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California
| | - Sung Yun Park
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, California
| | - Jie Wu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | - Amber N. Habowski
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, California
| | - Cassandra Van
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California
| | - Marcus M. Seldin
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
- Cancer Research Institute, University of California, Irvine, California
| | - Robert A. Edwards
- Department of Pathology & Laboratory Medicine, School of Medicine, University of California, Irvine, California
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
- Cancer Research Institute, University of California, Irvine, California
| | - Marian L. Waterman
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California, Irvine, California
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
- Cancer Research Institute, University of California, Irvine, California
| |
Collapse
|
35
|
Du Y. The Hippo signalling pathway and its impact on eye diseases. J Cell Mol Med 2024; 28:e18300. [PMID: 38613348 PMCID: PMC11015399 DOI: 10.1111/jcmm.18300] [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: 10/28/2023] [Revised: 02/26/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The Hippo signalling pathway, an evolutionarily conserved kinase cascade, has been shown to be crucial for cell fate determination, homeostasis and tissue regeneration. Recent experimental and clinical studies have demonstrated that the Hippo signalling pathway is involved in the pathophysiology of ocular diseases. This article provides the first systematic review of studies on the regulatory and functional roles of mammalian Hippo signalling systems in eye diseases. More comprehensive studies on this pathway are required for a better understanding of the pathophysiology of eye diseases and the development of effective therapies.
Collapse
Affiliation(s)
- Yuxiang Du
- Precision Medicine Laboratory for Chronic Non‐communicable Diseases of Shandong Province, Institute of Precision MedicineJining Medical UniversityJiningShandongPeople's Republic of China
| |
Collapse
|
36
|
Liu Z, Li S, Chen S, Sheng J, Li Z, Lv T, Yu W, Fan Y, Wang J, Liu W, Hu S, Jin J. YAP-mediated GPER signaling impedes proliferation and survival of prostate epithelium in benign prostatic hyperplasia. iScience 2024; 27:109125. [PMID: 38420594 PMCID: PMC10901089 DOI: 10.1016/j.isci.2024.109125] [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: 09/13/2023] [Revised: 12/21/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) occurs when there is an imbalance between the proliferation and death of prostate cells, which is regulated tightly by estrogen signaling. However, the role of G protein-coupled estrogen receptor (GPER) in prostate cell survival remains ambiguous. In this study, we observed that prostates with epithelial hyperplasia showed increased yes-associated protein 1 (YAP) expression and decreased levels of estrogen and GPER. Blocking YAP through genetic or drug interventions led to reduced proliferation and increased apoptosis in the prostate epithelial cells. Interestingly, GPER agonists produced similar effects. GPER activation enhanced the phosphorylation and degradation of YAP, which was crucial for suppressing cell proliferation and survival. The Gαs/cAMP/PKA/LATS pathway, downstream of GPER, transmitted signals that facilitated YAP inhibition. This study investigated the interaction between GPER and YAP in the prostate epithelial cells and its contribution to BPH development. It lays the groundwork for future research on developing BPH treatments.
Collapse
Affiliation(s)
- Zhifu Liu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Senmao Li
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Shengbin Chen
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jindong Sheng
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Gynaecological Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zheng Li
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Tianjing Lv
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Wei Yu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jinlong Wang
- Department of Urology, Tibet Autonomous Region People's Hospital, Lhasa 850000, China
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen 518036, China
- Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Shuai Hu
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
| | - Jie Jin
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, Beijing 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male), Molecular Diagnosis and Treatment Center, National Research Center for Genitourinary Oncology, Beijing 100034, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| |
Collapse
|
37
|
Rodriguez I, Rossi NM, Keskus AG, Xie Y, Ahmad T, Bryant A, Lou H, Paredes JG, Milano R, Rao N, Tulsyan S, Boland JF, Luo W, Liu J, O'Hanlon T, Bess J, Mukhina V, Gaykalova D, Yuki Y, Malik L, Billingsley KJ, Blauwendraat C, Carrington M, Yeager M, Mirabello L, Kolmogorov M, Dean M. Insights into the mechanisms and structure of breakage-fusion-bridge cycles in cervical cancer using long-read sequencing. Am J Hum Genet 2024; 111:544-561. [PMID: 38307027 PMCID: PMC10940022 DOI: 10.1016/j.ajhg.2024.01.002] [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: 08/18/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024] Open
Abstract
Cervical cancer is caused by human papillomavirus (HPV) infection, has few approved targeted therapeutics, and is the most common cause of cancer death in low-resource countries. We characterized 19 cervical and four head and neck cancer cell lines using long-read DNA and RNA sequencing and identified the HPV types, HPV integration sites, chromosomal alterations, and cancer driver mutations. Structural variation analysis revealed telomeric deletions associated with DNA inversions resulting from breakage-fusion-bridge (BFB) cycles. BFB is a common mechanism of chromosomal alterations in cancer, and our study applies long-read sequencing to this important chromosomal rearrangement type. Analysis of the inversion sites revealed staggered ends consistent with exonuclease digestion of the DNA after breakage. Some BFB events are complex, involving inter- or intra-chromosomal insertions or rearrangements. None of the BFB breakpoints had telomere sequences added to resolve the dicentric chromosomes, and only one BFB breakpoint showed chromothripsis. Five cell lines have a chromosomal region 11q BFB event, with YAP1-BIRC3-BIRC2 amplification. Indeed, YAP1 amplification is associated with a 10-year-earlier age of diagnosis of cervical cancer and is three times more common in African American women. This suggests that individuals with cervical cancer and YAP1-BIRC3-BIRC2 amplification, especially those of African ancestry, might benefit from targeted therapy. In summary, we uncovered valuable insights into the mechanisms and consequences of BFB cycles in cervical cancer using long-read sequencing.
Collapse
Affiliation(s)
- Isabel Rodriguez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Nicole M Rossi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Ayse G Keskus
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Yi Xie
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tanveer Ahmad
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Asher Bryant
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Hong Lou
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jesica Godinez Paredes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Rose Milano
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Nina Rao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Sonam Tulsyan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Joseph F Boland
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Wen Luo
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jia Liu
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Tim O'Hanlon
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jazmyn Bess
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Vera Mukhina
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medical Center, Baltimore, MD, USA
| | - Daria Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical System, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yuko Yuki
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias, National Institute on Aging, Bethesda, MD, USA
| | | | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Mary Carrington
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Meredith Yeager
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Mikhail Kolmogorov
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
38
|
Cruz SP, Zhang Q, Devarajan R, Paia C, Luo B, Zhang K, Koivusalo S, Qin L, Xia J, Ahtikoski A, Vaarala M, Wenta T, Wei G, Manninen A. Dampened Regulatory Circuitry of TEAD1/ITGA1/ITGA2 Promotes TGFβ1 Signaling to Orchestrate Prostate Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305547. [PMID: 38169150 PMCID: PMC10953553 DOI: 10.1002/advs.202305547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/18/2023] [Indexed: 01/05/2024]
Abstract
The extracellular matrix (ECM) undergoes substantial changes during prostate cancer (PCa) progression, thereby regulating PCa growth and invasion. Herein, a meta-analysis of multiple PCa cohorts is performed which revealed that downregulation or genomic loss of ITGA1 and ITGA2 integrin genes is associated with tumor progression and worse prognosis. Genomic deletion of both ITGA1 and ITGA2 activated epithelial-to-mesenchymal transition (EMT) in benign prostate epithelial cells, thereby enhancing their invasive potential in vitro and converting them into tumorigenic cells in vivo. Mechanistically, EMT is induced by enhanced secretion and autocrine activation of TGFβ1 and nuclear targeting of YAP1. An unbiased genome-wide co-expression analysis of large PCa cohort datasets identified the transcription factor TEAD1 as a key regulator of ITGA1 and ITGA2 expression in PCa cells while TEAD1 loss phenocopied the dual loss of α1- and α2-integrins in vitro and in vivo. Remarkably, clinical data analysis revealed that TEAD1 downregulation or genomic loss is associated with aggressive PCa and together with low ITGA1 and ITGA2 expression synergistically impacted PCa prognosis and progression. This study thus demonstrated that loss of α1- and α2-integrins, either via deletion/inactivation of the ITGA1/ITGA2 locus or via loss of TEAD1, contributes to PCa progression by inducing TGFβ1-driven EMT.
Collapse
Affiliation(s)
- Sara P. Cruz
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Qin Zhang
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Raman Devarajan
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Christos Paia
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Binjie Luo
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Kai Zhang
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Saara Koivusalo
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Longguang Qin
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Jihan Xia
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| | - Anne Ahtikoski
- Departments of Urology, Pathology and Radiology, and Medical Research Center OuluOulu University Hospital and University of OuluAapistie 5aOulu90220Finland
| | - Markku Vaarala
- Departments of Urology, Pathology and Radiology, and Medical Research Center OuluOulu University Hospital and University of OuluAapistie 5aOulu90220Finland
| | - Tomasz Wenta
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
- Department of General and Medical Biochemistry, Faculty of BiologyUniversity of GdanskJana Bażyńskiego 8Gdańsk80–309Poland
| | - Gong‐Hong Wei
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
- Fudan University Shanghai Cancer Center & MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical SciencesShanghai Medical College of Fudan University138 Yi Xue Yuan RoadShanghai200032China
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine, Biocenter OuluUniversity of OuluAapistie 5aOulu90220Finland
| |
Collapse
|
39
|
Li M, Zhang FJ, Bai RJ. The Hippo-YAP Signaling Pathway in Osteoarthritis and Rheumatoid Arthritis. J Inflamm Res 2024; 17:1105-1120. [PMID: 38406325 PMCID: PMC10891274 DOI: 10.2147/jir.s444758] [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: 10/14/2023] [Accepted: 02/08/2024] [Indexed: 02/27/2024] Open
Abstract
Arthritis is the most prevalent joint disease and is characterized by articular cartilage degradation, synovial inflammation, and changes in periarticular and subchondral bone. Recent studies have reported that Yes-associated protein (YAP) and the transcriptional coactivator with PDZ-binding motif (TAZ) have significant effects on the proliferation, migration, and survival of chondrocytes and fibroblast-like synovial cells (FLSs). YAP/TAZ signaling pathway, as well as the related Hippo-YAP signaling pathway, are responsible for the condition of cells and articular cartilage in joints. They are tightly regulated to maintain metabolism in chondrocytes and FLSs because abnormal expression may result in cartilage damage. However, the roles and mechanisms of the Hippo-YAP pathway in arthritis remain largely unknown. This review summarizes the roles and key functions of YAP/TAZ and the Hippo-YAP signaling pathway in FLSs and chondrocytes for the induction of proliferation, migration, survival, and differentiation in rheumatoid arthritis (RA) and osteoarthritis (OA) research. We also discuss the therapeutic strategies involving YAP/TAZ and the related Hippo-YAP signaling pathway involved in OA.
Collapse
Affiliation(s)
- Min Li
- Department of Orthopaedics, Wuxi Ninth People’s Hospital, Soochow University, Wuxi, Jiangsu, 214000, People’s Republic of China
| | - Fang-Jie Zhang
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, Hunan, 410008, People’s Republic of China
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People’s Republic of China
| | - Rui-Jun Bai
- Department of Orthopaedics, Wuxi Ninth People’s Hospital, Soochow University, Wuxi, Jiangsu, 214000, People’s Republic of China
| |
Collapse
|
40
|
Mandl A, Jasmine S, Krueger T, Kumar R, Coleman IM, Dalrymple SL, Antony L, Rosen DM, Jing Y, Hanratty B, Patel RA, Jin-Yih L, Dias J, Celatka CA, Tapper AE, Kleppe M, Kanayama M, Speranzini V, Wang YZ, Luo J, Corey E, Sena LA, Casero RA, Lotan T, Trock BJ, Kachhap SK, Denmeade SR, Carducci MA, Mattevi A, Haffner MC, Nelson PS, Rienhoff HY, Isaacs JT, Brennen WN. LSD1 inhibition suppresses ASCL1 and de-represses YAP1 to drive potent activity against neuroendocrine prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576106. [PMID: 38328141 PMCID: PMC10849473 DOI: 10.1101/2024.01.17.576106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Lysine-specific demethylase 1 (LSD1 or KDM1A ) has emerged as a critical mediator of tumor progression in metastatic castration-resistant prostate cancer (mCRPC). Among mCRPC subtypes, neuroendocrine prostate cancer (NEPC) is an exceptionally aggressive variant driven by lineage plasticity, an adaptive resistance mechanism to androgen receptor axis-targeted therapies. Our study shows that LSD1 expression is elevated in NEPC and associated with unfavorable clinical outcomes. Using genetic approaches, we validated the on-target effects of LSD1 inhibition across various models. We investigated the therapeutic potential of bomedemstat, an orally bioavailable, irreversible LSD1 inhibitor with low nanomolar potency. Our findings demonstrate potent antitumor activity against CRPC models, including tumor regressions in NEPC patient-derived xenografts. Mechanistically, our study uncovers that LSD1 inhibition suppresses the neuronal transcriptional program by downregulating ASCL1 through disrupting LSD1:INSM1 interactions and de-repressing YAP1 silencing. Our data support the clinical development of LSD1 inhibitors for treating CRPC - especially the aggressive NE phenotype. Statement of Significance Neuroendocrine prostate cancer presents a clinical challenge due to the lack of effective treatments. Our research demonstrates that bomedemstat, a potent and selective LSD1 inhibitor, effectively combats neuroendocrine prostate cancer by downregulating the ASCL1- dependent NE transcriptional program and re-expressing YAP1.
Collapse
|
41
|
Kervarrec T, Pissaloux D, Tirode F, de la Fouchardière A, Sohier P, Frouin E, Hamard A, Houben R, Schrama D, Barlier A, Cribier B, Battistella M, Macagno N. Gene fusions in poroma, porocarcinoma and related adnexal skin tumours: An update. Histopathology 2024; 84:266-278. [PMID: 37609771 DOI: 10.1111/his.15023] [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: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/24/2023]
Abstract
Poroma is a benign sweat gland tumour showing morphological features recapitulating the superficial portion of the eccrine sweat coil. A subset of poromas may transform into porocarcinoma, its malignant counterpart. Poroma and porocarcinoma are characterised by recurrent gene fusions involving YAP1, a transcriptional co-activator, which is controlled by the Hippo signalling pathway. The fusion genes frequently involve MAML2 and NUTM1, which are also rearranged in other cutaneous and extracutaneous neoplasms. We aimed to review the clinical, morphological and molecular features of this category of adnexal neoplasms with a special focus upon emerging differential diagnoses, and discuss how their systematic molecular characterisation may contribute to a standardisation of diagnosis, more accurate classification and, ultimately, refinement of their prognosis and therapeutic modalities.
Collapse
Affiliation(s)
- Thibault Kervarrec
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Department of Pathology, University Hospital of Tours, Tours, France
- 'Biologie des infections à polyomavirus' Team, UMR1282 INRAE, University of Tours, Tours, France
| | | | - Franck Tirode
- Department of Biopathology, Center Léon Bérard, Lyon, France
- Centre Léon Bérard, Cancer Research Center of Lyon, Equipe Labellisée, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Arnaud de la Fouchardière
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Department of Biopathology, Center Léon Bérard, Lyon, France
- Centre Léon Bérard, Cancer Research Center of Lyon, Equipe Labellisée, University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Pierre Sohier
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Department of Pathology, Hôpital Cochin, AP-HP, AP-HP Centre - Université Paris Cité, Paris, France
- Faculté de Médecine, University Paris Cité, Paris, France
| | - Eric Frouin
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Department of Pathology, University Hospital of Poitiers, University of Poitiers, LITEC, Poitiers, France
| | - Aymeric Hamard
- Department of Pathology, University Hospital of Tours, Tours, France
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Anne Barlier
- Aix-Marseille Univ, INSERM, MMG, U1251, Marmara Institute, Marseille, France
- Laboratory of Molecular Biology, La Conception Hospital, Marseille, France
| | - Bernard Cribier
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Clinique Dermatologique, Hôpital Civil, Hôpitaux Universitaires, Université de Strasbourg, Strasbourg, France
| | - Maxime Battistella
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Department of Pathology, Hospital Saint-Louis, AP-HP, Université Paris Cité, INSERM U976, Paris, France
| | - Nicolas Macagno
- CARADERM, French Network of Rare Cutaneous Cancer, Lille, France
- Aix-Marseille Univ, INSERM, MMG, U1251, Marmara Institute, Marseille, France
- Department of Pathology, APHM, Timone University Hospital, Marseille, France
| |
Collapse
|
42
|
Nomura M, Ohuchi M, Sakamoto Y, Kudo K, Yaku K, Soga T, Sugiura Y, Morita M, Hayashi K, Miyahara S, Sato T, Yamashita Y, Ito S, Kikuchi N, Sato I, Saito R, Yaegashi N, Fukuhara T, Yamada H, Shima H, Nakayama KI, Hirao A, Kawasaki K, Arai Y, Akamatsu S, Tanuma SI, Sato T, Nakagawa T, Tanuma N. Niacin restriction with NAMPT-inhibition is synthetic lethal to neuroendocrine carcinoma. Nat Commun 2023; 14:8095. [PMID: 38092728 PMCID: PMC10719245 DOI: 10.1038/s41467-023-43630-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) plays a major role in NAD biosynthesis in many cancers and is an attractive potential cancer target. However, factors dictating therapeutic efficacy of NAMPT inhibitors (NAMPTi) are unclear. We report that neuroendocrine phenotypes predict lung and prostate carcinoma vulnerability to NAMPTi, and that NAMPTi therapy against those cancers is enhanced by dietary modification. Neuroendocrine differentiation of tumor cells is associated with down-regulation of genes relevant to quinolinate phosphoribosyltransferase-dependent de novo NAD synthesis, promoting NAMPTi susceptibility in vitro. We also report that circulating nicotinic acid riboside (NAR), a non-canonical niacin absent in culture media, antagonizes NAMPTi efficacy as it fuels NAMPT-independent but nicotinamide riboside kinase 1-dependent NAD synthesis in tumors. In mouse transplantation models, depleting blood NAR by nutritional or genetic manipulations is synthetic lethal to tumors when combined with NAMPTi. Our findings provide a rationale for simultaneous targeting of NAR metabolism and NAMPT therapeutically in neuroendocrine carcinoma.
Collapse
Affiliation(s)
- Miyuki Nomura
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Mai Ohuchi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Yoshimi Sakamoto
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Kei Kudo
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mami Morita
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Kayoko Hayashi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Shuko Miyahara
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Sato
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Yoji Yamashita
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Shigemi Ito
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Naohiko Kikuchi
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Ikuro Sato
- Department of Pathology, Miyagi Cancer Center Hospital, Natori, Japan
| | - Rintaro Saito
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuro Fukuhara
- Department of Respiratory Medicine, Miyagi Cancer Center Hospital, Natori, Japan
| | - Hidekazu Yamada
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Hiroshi Shima
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyusyu University, Fukuoka, Japan
- TMDU Advanced Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer and Stem Cell Research Program, Cancer Research Institute and WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Kenta Kawasaki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoichi Arai
- Department of Urology, Miyagi Cancer Center Hospital, Natori, Japan
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sei-Ichi Tanuma
- Meikai University Research Institute of Odontology, Sakado, Japan
- University of Human Arts and Sciences, Saitama, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Nobuhiro Tanuma
- Division of Cancer Chemotherapy, Miyagi Cancer Center Research Institute, Natori, Japan.
- Department of Biochemical Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
43
|
Chow SE, Hsu CC, Yang CT, Meir YJJ. YAP co-localizes with the mitotic spindle and midbody to safeguard mitotic division in lung-cancer cells. FEBS J 2023; 290:5704-5719. [PMID: 37549045 DOI: 10.1111/febs.16926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/22/2023] [Accepted: 08/07/2023] [Indexed: 08/09/2023]
Abstract
YES-associated protein (YAP) is a part of the Hippo pathway, with pivotal roles in several developmental processes and dual functionality as both a tumor suppressor and an oncogene. In the present study, we identified YAP activity as a microtubular scaffold protein that maintains the stability of the mitotic spindle and midbody by physically interacting with α-tubulin during mitotic progression. The interaction of YAP and α-tubulin was evident in co-immunoprecipitation assays, as well as observing their co-localization in the microtubular structure of the mitotic spindle and midbody in immunostainings. With YAP depletion, levels of ECT2, MKLP-1, and Aurora B are reduced, which is consistent with YAP functioning in midbody formation during cytokinesis. The concomitant decrease in α-tubulin and increase in acetyl-α-tubulin during YAP depletion occurred at the post-transcriptional level. This suggests that YAP maintains the stability of the mitotic spindle and midbody, which ensures appropriate chromosome segregation during mitotic division. The increase in acetyl-α-tubulin during YAP depletion may provide a lesion-halting mechanism in maintaining the microtubule structure. The depletion of YAP also results in multinuclearity and aneuploidy, which supports its role in stabilizing the mitotic spindle and midbody.
Collapse
Affiliation(s)
- Shu-Er Chow
- Department of Otolaryngology-Head and Neck Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Nature Science, Center for General Studies, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chi Hsu
- Department of Nature Science, Center for General Studies, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Taoyuan Chang Gung Memorial Hospital, Taiwan
| | - Yaa-Jyuhn J Meir
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
44
|
Zhao Y, Sheldon M, Sun Y, Ma L. New Insights into YAP/TAZ-TEAD-Mediated Gene Regulation and Biological Processes in Cancer. Cancers (Basel) 2023; 15:5497. [PMID: 38067201 PMCID: PMC10705714 DOI: 10.3390/cancers15235497] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 02/12/2024] Open
Abstract
The Hippo pathway is conserved across species. Key mammalian Hippo pathway kinases, including MST1/2 and LATS1/2, inhibit cellular growth by inactivating the TEAD coactivators, YAP, and TAZ. Extensive research has illuminated the roles of Hippo signaling in cancer, development, and regeneration. Notably, dysregulation of Hippo pathway components not only contributes to tumor growth and metastasis, but also renders tumors resistant to therapies. This review delves into recent research on YAP/TAZ-TEAD-mediated gene regulation and biological processes in cancer. We focus on several key areas: newly identified molecular patterns of YAP/TAZ activation, emerging mechanisms that contribute to metastasis and cancer therapy resistance, unexpected roles in tumor suppression, and advances in therapeutic strategies targeting this pathway. Moreover, we provide an updated view of YAP/TAZ's biological functions, discuss ongoing controversies, and offer perspectives on specific debated topics in this rapidly evolving field.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Marisela Sheldon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
45
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
46
|
Kim MK, Han SH, Park TG, Song SH, Lee JY, Lee YS, Yoo SY, Chi XZ, Kim EG, Jang JW, Lim DS, van Wijnen AJ, Lee JW, Bae SC. The TGFβ→TAK1→LATS→YAP1 Pathway Regulates the Spatiotemporal Dynamics of YAP1. Mol Cells 2023; 46:592-610. [PMID: 37706312 PMCID: PMC10590711 DOI: 10.14348/molcells.2023.0088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/10/2023] [Accepted: 07/25/2023] [Indexed: 09/15/2023] Open
Abstract
The Hippo kinase cascade functions as a central hub that relays input from the "outside world" of the cell and translates it into specific cellular responses by regulating the activity of Yes-associated protein 1 (YAP1). How Hippo translates input from the extracellular signals into specific intracellular responses remains unclear. Here, we show that transforming growth factor β (TGFβ)-activated TAK1 activates LATS1/2, which then phosphorylates YAP1. Phosphorylated YAP1 (p-YAP1) associates with RUNX3, but not with TEAD4, to form a TGFβ-stimulated restriction (R)-point-associated complex which activates target chromatin loci in the nucleus. Soon after, p-YAP1 is exported to the cytoplasm. Attenuation of TGFβ signaling results in re-localization of unphosphorylated YAP1 to the nucleus, where it forms a YAP1/TEAD4/SMAD3/AP1/p300 complex. The TGFβ-stimulated spatiotemporal dynamics of YAP1 are abrogated in many cancer cells. These results identify a new pathway that integrates TGFβ signals and the Hippo pathway (TGFβ→TAK1→LATS1/2→YAP1 cascade) with a novel dynamic nuclear role for p-YAP1.
Collapse
Affiliation(s)
- Min-Kyu Kim
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Sang-Hyun Han
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Tae-Geun Park
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Soo-Hyun Song
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Ja-Youl Lee
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - You-Soub Lee
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Seo-Yeong Yoo
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Xin-Zi Chi
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Eung-Gook Kim
- Department of Biochemistry, College of Medicine and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Ju-Won Jang
- Department of Biomedical Science, Cheongju University, Cheongju 28503, Korea
| | - Dae Sik Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Andre J. van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Jung-Won Lee
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| | - Suk-Chul Bae
- Department of Biochemistry, College of Medicine and Institute for Tumour Research, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
47
|
Safari M, Scotto L, Litman T, Petrukhin LA, Zhu H, Shen M, Robey RW, Hall MD, Fojo T, Bates SE. Novel Therapeutic Strategies Exploiting the Unique Properties of Neuroendocrine Neoplasms. Cancers (Basel) 2023; 15:4960. [PMID: 37894327 PMCID: PMC10605125 DOI: 10.3390/cancers15204960] [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: 08/21/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Over the last few decades of treatment, the outcomes for at least some subsets of neuroendocrine neoplasms (NENs) have improved. However, the identification of new vulnerabilities for this heterogeneous group of cancers remains a priority. METHODS Using two libraries of compounds selected for potential repurposing, we identified the inhibitors of nicotinamide phosphoribosyltransferase (NAMPT) and histone deacetylases (HDAC) as the agents with the highest activity. We validated the hits in an expanded set of neuroendocrine cell lines and examined the mechanisms of action. RESULTS In Kelly, NH-6, and NCI-H82, which are two neuroblastoma and one small cell lung cancer cell lines, respectively, metabolic studies suggested that cell death following NAMPT inhibition is the result of a reduction in basal oxidative phosphorylation and energy production. NAMPT is the rate-limiting enzyme in the production of NAD+, and in the three cell lines, NAMPT inhibition led to a marked reduction in the ATP and NAD+ levels and the catalytic activity of the citric acid cycle. Moreover, comparative analysis of the mRNA expression in drug-sensitive and -insensitive cell lines found less dependency of the latter on oxidative phosphorylation for their energy requirement. Further, the analysis of HDAC and NAMPT inhibitors administered in combination found marked activity using low sub-lethal concentrations of both agents, suggesting a synergistic effect. CONCLUSION These data suggest NAMPT inhibitors alone or in combination with HDAC inhibitors could be particularly effective in the treatment of neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Maryam Safari
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Luigi Scotto
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Thomas Litman
- Department of Immunology and Microbiology, University of Copenhagen, 1172 Copenhagen, Denmark
| | - Lubov A. Petrukhin
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Hu Zhu
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Min Shen
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Robert W. Robey
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20892, USA
| | - Tito Fojo
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Susan E. Bates
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
- James J. Peters Bronx Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
48
|
Lv L, Zhou X. Targeting Hippo signaling in cancer: novel perspectives and therapeutic potential. MedComm (Beijing) 2023; 4:e375. [PMID: 37799806 PMCID: PMC10547939 DOI: 10.1002/mco2.375] [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: 04/19/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
As highly conserved among diverse species, Hippo signaling pathway regulates various biological processes, including development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size. Studies in the last two decades have provided a good framework for how these fundamental functions of Hippo signaling are tightly regulated by a network with numerous intracellular and extracellular factors. The Hippo signaling pathway, when dysregulated, may lead to a wide variety of diseases, especially cancer. There is growing evidence demonstrating that dysregulated Hippo signaling is closely associated with tumorigenesis, cancer cell invasion, and migration, as well as drug resistance. Therefore, the Hippo pathway is considered an appealing therapeutic target for the treatment of cancer. Promising novel agents targeting the Hippo signaling pathway for cancers have recently emerged. These novel agents have shown antitumor activity in multiple cancer models and demonstrated therapeutic potential for cancer treatment. However, the detailed molecular basis of the Hippo signaling-driven tumor biology remains undefined. Our review summarizes current advances in understanding the mechanisms by which Hippo signaling drives tumorigenesis and confers drug resistance. We also propose strategies for future preclinical and clinical development to target this pathway.
Collapse
Affiliation(s)
- Liemei Lv
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
49
|
Wu Z, Su J, Li FL, Chen T, Mayner J, Engler A, Ma S, Li Q, Guan KL. YAP silencing by RB1 mutation is essential for small-cell lung cancer metastasis. Nat Commun 2023; 14:5916. [PMID: 37739954 PMCID: PMC10516997 DOI: 10.1038/s41467-023-41585-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023] Open
Abstract
Small cell lung cancer (SCLC) is highly lethal due to its prevalent metastasis. Most SCLCs have inactivating mutations in TP53 and RB1. We find that loss of YAP expression is key for SCLC cells to acquire rapid ameboid migration and high metastatic potential. YAP functions through its target genes CCN1/CCN2 to inhibit SCLC ameboid migration. RB1 mutation contributes to YAP transcriptional silencing via E2F7, which recruits the RCOR co-repressor complex to YAP promoter. We discover that benzamide family HDAC inhibitors stimulate YAP expression by inhibiting the RCOR-HDAC complex, thereby suppressing SCLC metastasis and improving survival in a mouse model. Our study unveils the molecular and cellular basis underlying SCLC's high metastatic potential, the previously unrecognized role of YAP in suppressing ameboid migration and tumor metastasis, and the mechanism of YAP transcription regulation involving E2F7, RCOR, and Sin3 HDAC. This study reveals a therapeutic potential of benzamides for SCLC treatment.
Collapse
Affiliation(s)
- Zhengming Wu
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Junhui Su
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Fu-Long Li
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jaimie Mayner
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Adam Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Shenghong Ma
- Wellcome Sanger Institute, Cambridge, CB10 1RQ, UK
| | - Qingquan Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA.
- School of Life Sciences, Westlake University, Hangzhou, 310030, China.
| |
Collapse
|
50
|
Aubry A, Pearson JD, Charish J, Yu T, Sivak JM, Xirodimas DP, Avet-Loiseau H, Corre J, Monnier PP, Bremner R. Deneddylation of ribosomal proteins promotes synergy between MLN4924 and chemotherapy to elicit complete therapeutic responses. Cell Rep 2023; 42:112925. [PMID: 37552601 DOI: 10.1016/j.celrep.2023.112925] [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: 11/14/2022] [Revised: 05/29/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
The neddylation inhibitor MLN4924/Pevonedistat is in clinical trials for multiple cancers. Efficacy is generally attributed to cullin RING ligase (CRL) inhibition, but the contribution of non-CRL targets is unknown. Here, CRISPR screens map MLN4924-monotherapy sensitivity in retinoblastoma to a classic DNA damage-induced p53/E2F3/BAX-dependent death effector network, which synergizes with Nutlin3a or Navitoclax. In monotherapy-resistant cells, MLN4924 plus standard-of-care topotecan overcomes resistance, but reduces DNA damage, instead harnessing ribosomal protein nucleolar-expulsion to engage an RPL11/p21/MYCN/E2F3/p53/BAX synergy network that exhibits extensive cross-regulation. Strikingly, unneddylatable RPL11 substitutes for MLN4924 to perturb nucleolar function and enhance topotecan efficacy. Orthotopic tumors exhibit complete responses while preserving visual function. Moreover, MLN4924 plus melphalan deploy this DNA damage-independent strategy to synergistically kill multiple myeloma cells. Thus, MLN4924 synergizes with standard-of-care drugs to unlock a nucleolar death effector network across cancer types implying broad therapeutic relevance.
Collapse
Affiliation(s)
- Arthur Aubry
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France
| | - Joel D Pearson
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Jason Charish
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Tao Yu
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Jeremy M Sivak
- Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Hervé Avet-Loiseau
- Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM, Toulouse, France
| | - Jill Corre
- Centre Hospitalo-universitaire (CHU) de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Université de Toulouse, UPS, Unité de Génomique du Myélome, Toulouse, France; Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM, Toulouse, France
| | - Philippe P Monnier
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada; Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|