1
|
Lu TY, Ji Y, Lyu C, Shen EN, Sun Y, Xiang Y, Meng-Saccoccio T, Feng GS, Chen S. Bioprinted high cell density liver model with improved hepatic metabolic functions. Biomaterials 2025; 320:123256. [PMID: 40101310 DOI: 10.1016/j.biomaterials.2025.123256] [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: 11/16/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
In vitro liver tissue models are valuable for studying liver function, understanding liver diseases, and screening candidate drugs for toxicity and efficacy. While three-dimensional (3D) bioprinting shows promise in creating various types of functional tissues, current efforts to engineer a functional liver tissue face challenges in replicating native high cell density (HCD) and maintaining long-term cell viability. HCD is crucial for establishing the cell-cell interactions necessary to mimic the liver's metabolic and detoxification functions. However, HCD bioinks exacerbate light scattering in light-based 3D bioprinting. In this study, we incorporated iodixanol into our bioink formulation to minimize light scattering, enabling the fabrication of hepatic tissue constructs with an HCD of 8 × 107 cells/mL while maintaining high cell viability (∼80 %). The printed dense hepatic tissue constructs showed enhanced cell-cell interactions, as evidenced by increased expression of E-cadherin and ZO-1. Furthermore, these constructs promoted albumin secretion, urea production, and P450 metabolic activity. Additionally, HCD hepatic tissue inactivated the YAP/TAZ pathway via cell-cell interactions, preserving primary hepatocyte functions. Further screening revealed that hepatocytes in the dense model were more sensitive to drug treatments than those in a lower-density hepatic model, highlighting the importance of HCD in recapitulating the physiological drug responses. Overall, our approach represents a significant advancement in liver tissue engineering, providing a promising platform for the development of physiologically relevant in vitro liver models for drug screening and toxicity testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yichun Ji
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cheng Lyu
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Erin Nicole Shen
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yazhi Sun
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yi Xiang
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tobias Meng-Saccoccio
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Gen-Sheng Feng
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Yuan Q, Xia X, Yuan Y, Chen Q, Feng X. A YAP-derived peptide blocks YAP-TEAD signaling and suppresses cell proliferation. Cell Signal 2025; 131:111738. [PMID: 40081550 DOI: 10.1016/j.cellsig.2025.111738] [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: 12/16/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Yes-associated protein (YAP), a pivotal transcriptional co-activator in cell growth regulation, exerts its function through interactions with transcriptional factors like TEAD. Ectopic activation of YAP causes excessive cell proliferation, leading to multiple human diseases, including cancers. However, current pharmacological YAP inhibition lacks specificity and may have unintended effects, necessitating the development of direct YAP-derived inhibitors. In this study, we designed a novel YAP-derived peptide, TBDi, that specifically disrupted YAP-TEAD interaction and exhibited robust inhibition of TEAD activity. Mechanistically, TBDi directly binds to TEAD, blocking the physical interaction between YAP and TEAD. Transcriptomic analysis revealed that TBDi significantly altered gene expression profiles associated with TEAD activity, including downregulation of signature genes like CYR61 and CTGF. Functionally, TBDi emerged as a potent suppressor of cell proliferation, inhibiting cell proliferation to a degree comparable to YAP/TAZ knockdown. Altogether, our study not only identifies TBDi as a promising tool to block YAP-TEAD axis, but also offers insights for potential therapeutic interventions in diseases.
Collapse
Affiliation(s)
- Qiuyun Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, Zhejiang 310006, China
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Luo C, Liang H, Ji M, Ye C, Lin Y, Guo Y, Zhang Z, Shu Y, Jin X, Lu S, Lu W, Dang Y, Zhang H, Li B, Zhou G, Zhang Z, Chang L. Autophagy induced by mechanical stress sensitizes cells to ferroptosis by NCOA4-FTH1 axis. Autophagy 2025; 21:1263-1282. [PMID: 39988734 DOI: 10.1080/15548627.2025.2469129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025] Open
Abstract
Ferroptosis is an iron-dependent regulated form of cell death implicated in various diseases, including cancers, with its progression influenced by iron-dependent peroxidation of phospholipids and dysregulation of the redox system. Whereas the extracellular matrix of tumors provides mechanical cues influencing tumor initiation and progression, its impact on ferroptosis and its mechanisms remains largely unexplored. In this study, we reveal that heightened mechanical tension sensitizes cells to ferroptosis, whereas decreased mechanics confers resistance. Mechanistically, reduced mechanical tension reduces intracellular free iron levels by enhancing FTH1 protein expression. Additionally, low mechanics significantly diminishes NCOA4, pivotal in mediating FTH1 phase separation-induced ferritinophagy. Targeting NCOA4 effectively rescues ferroptosis susceptibility under low mechanical tension through modulation of FTH1 phase separation-driven autophagy. In conclusion, our findings demonstrate that mechanics regulates iron metabolism via NCOA4-FTH1 phase separation-mediated autophagy, thereby influencing ferroptosis sensitivity and offering promising therapeutic avenues for future exploration.Abbreviations: ACO1: aconitase 1; ATG5: autophagy related 5; DMSO: dimethyl sulfoxide; EGFP: enhanced green fluorescent protein; FACS: fluorescence-activated cell sorting; FER-1: ferrostatin-1; FTH1: ferritin heavy chain 1; FTL: ferritin light chain; GPX4: glutathione peroxidase 4; IR: ionizing radiation; IREB2: iron responsive element binding protein 2; NCOA4: nuclear receptor coactivator 4; NFE2L2: NFE2 like bZIP transcription factor 2; NOPP: norepinephrine; PBS: phosphate-buffered saline; PI: propidium iodide; RSL3: (1S,3 R)-RSL3; TCGA: The Cancer Genome Atlas; WWTR1: WW domain containing transcription regulator 1; YAP1: Yes1 associated transcriptional regulator.
Collapse
Affiliation(s)
- Chenyu Luo
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
- Department of Hematology and Oncology, 986 Hospital of People's Liberation Army Air Force, Xian, China
| | - Haisheng Liang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Mintao Ji
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Caiyong Ye
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Yiping Lin
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Yuhan Guo
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Zhisen Zhang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Yinyin Shu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Xiaoni Jin
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Shuangshuang Lu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Wanling Lu
- Department of Hematology and Oncology, 986 Hospital of People's Liberation Army Air Force, Xian, China
| | - Yazheng Dang
- Department of Hematology and Oncology, 986 Hospital of People's Liberation Army Air Force, Xian, China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bingyan Li
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
| | - Zengli Zhang
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, China
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou, China
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Mukherjee S, Warden EA, Zhang J. YAP/TAZ: An epitome of tumorigenesis. Cancer Lett 2025; 625:217806. [PMID: 40381686 DOI: 10.1016/j.canlet.2025.217806] [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: 03/29/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Mounting evidence has demonstrated that the transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are the main effectors of the Hippo signal transduction pathway that is involved in multiple layered events in tumorigenesis. The role of YAP/TAZ in cancer development is critical in a context dependent manner. Overexpression of YAP/TAZ induces cell proliferation and is elevated in various cancers and many other malignancies. On the other hand, studies have shown YAP binds p73 to activate PML transcription in response to DNA damage and generate a DNA-damage-induced feedback loop. Intriguingly, at the genomic level, YAP/TAZ genes are rarely mutated in cancer, except in specific tumors. The central role of YAP/TAZ in driving tumorigenesis is attributed through diverse mechanisms, such as regulatory kinases, cellular mechano-transduction, epigenetic modification/alterations, post-translational modifications, protein -protein interaction and nucleo-cytoplasmic export import. The complex interplay among feedback loops and crosstalk between various signaling pathways portrays the dynamic nature of YAP/TAZ. Thus, a comprehensive understanding of how posttranslational modifications and nucleo-cytoplasmic traffic of YAP/TAZ dynamically regulate and control each other holds great promise for selectively targeting YAP/TAZ import and export for drug therapy.
Collapse
Affiliation(s)
- Soumya Mukherjee
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Emily A Warden
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Jianmin Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA.
| |
Collapse
|
5
|
Yu M, Wang J, Zhang X, Zhang H, Li C, Li J, Lin J, Zheng J, Huang L, Li Y, Sun S. The mechanism of YAP/TAZ transactivation and dual targeting for cancer therapy. Nat Commun 2025; 16:3855. [PMID: 40274828 PMCID: PMC12022045 DOI: 10.1038/s41467-025-59309-w] [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: 06/26/2024] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) play key roles in cancers through transcriptional outputs. However, their transactivation mechanisms remain unclear, and effective targeting strategies are lacking. Here, we show that YAP/TAZ possess a hydrophobic transactivation domain (TAD). TAD knockout prevents tumor establishment due to growth defects and enhances immune attack. Mechanistically, TADs facilitate preinitiation complex (PIC) assembly by recruiting the TATA-binding protein-associated factor 4 (TAF4)-dependent TFIID complex and enhance RNA polymerase II (Pol II) elongation through mediator complex subunit 15 (MED15)-dependent mediator recruitment for the expressions of oncogenic/immune-suppressive programs. The synthesized peptide TJ-M11 selectively disrupts TAD interactions with MED15 and TAF4, suppressing tumor growth and sensitizing tumors to immunotherapy. Our findings demonstrate that YAP/TAZ TADs exhibit dual functions in PIC assembly and Pol II elongation via hydrophobic interactions, which represent actionable targets for cancer therapy and combination immunotherapy.
Collapse
Affiliation(s)
- Man Yu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jingning Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoqiang Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Juebei Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaming Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, China
| | - Liu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, China.
| | - Shuguo Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Hao F, Zhang Y, Hou J, Zhao B. Chromatin remodeling and cancer: the critical influence of the SWI/SNF complex. Epigenetics Chromatin 2025; 18:22. [PMID: 40269969 PMCID: PMC12016160 DOI: 10.1186/s13072-025-00590-w] [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: 12/12/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
The SWI/SNF complex was first identified in yeast and named after studies of mutants critical for the mating-type switch (SWI) and sucrose non-fermenting (SNF) pathways.The SWI/SNF complex plays a pivotal role in regulating gene expression by altering chromatin structure to promote or suppress the expression of specific genes, maintain stem cell pluripotency, and participate in various biological processes. Mutations in the SWI/SNF complex are highly prevalent in various human cancers, significantly impacting tumor suppressive or oncogenic functions and influencing tumor initiation and progression. This review focuses on the mechanisms by which ARID1A/ARID1B, PBRM1, SMARCB1, and SMARCA2/SMARCA4 contribute to cancer, the immunoregulatory roles of the SWI/SNF complex, its involvement in DNA repair pathways, synthetic lethality, and applications in precision oncology.
Collapse
Affiliation(s)
- Fengxiang Hao
- Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China
| | - Ying Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China
| | - Jiayi Hou
- Department of Clinical Laboratory, Shanxi Provincial Academy of Traditional Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Bin Zhao
- Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi Province, 030001, China.
| |
Collapse
|
7
|
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
|
8
|
Guo Y, Zhang J, Gong L, Liu N, Liu Q, Liu Z, Guo B, Yang J. Mechanical Loading Induces NRF2 Nuclear Translocation to Epigenetically Remodel Oxidative Stress Defense in Osteocytes. Antioxidants (Basel) 2025; 14:346. [PMID: 40227420 PMCID: PMC11939503 DOI: 10.3390/antiox14030346] [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: 02/01/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
The mechano-responsiveness of osteocytes is critical for maintaining bone health and associated with a reduced oxidative stress defense, yet the precise molecular mechanisms remain incompletely understood. Here, we address the gap by investigating the epigenetic reprogramming that drives osteocyte responses to mechanical loading. We found overall remodeling of antioxidant response under mechanical loading and identified NRF2, a key transcription factor in oxidative stress response, which plays a vital role in the epigenetic remodeling of osteocytes. The results showed that mechanical loading enhanced NRF2 protein stability, promoted its nuclear translocation, and activated osteocyte-specific transcriptional programs. In contrast, pharmacological stabilization of NRF2 failed to fully replicate these effects, underscoring the unique role of mechanical stimuli in modulating NRF2 activity and antioxidant function. Our findings highlight the potential therapeutic limitations of NRF2-stabilizing drugs and suggest that combining pharmacological approaches with mechanical interventions could offer more effective treatments to maintain oxidative homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Baosheng Guo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China; (Y.G.); (J.Z.); (L.G.); (N.L.); (Q.L.); (Z.L.)
| | - Jingping Yang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China; (Y.G.); (J.Z.); (L.G.); (N.L.); (Q.L.); (Z.L.)
| |
Collapse
|
9
|
Tibebe H, Marquez D, McGraw A, Gagliardi S, Sullivan C, Hillmer G, Narayan K, Izumi C, Keating A, Izumi T. Targeting Latent HIV Reservoirs: Effectiveness of Combination Therapy with HDAC and PARP Inhibitors. Viruses 2025; 17:400. [PMID: 40143326 PMCID: PMC11945899 DOI: 10.3390/v17030400] [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: 02/14/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
The "Kick and Kill" strategy, which aims to reactivate latent HIV reservoirs and facilitate the clearance of reactivated HIV-infected cells, has yet to achieve a functional cure due to the limited efficacy of current latency reversal agents. This study evaluates the combination efficacy of histone deacetylase (HDAC) inhibitor with poly(ADP-ribose) polymerase (PARP) inhibitor in latency reversal and immune-mediated clearance. Latently infected J-Lat cells and dual-fluorescent HIV-infected primary CD4 T cells were treated with the HDAC inhibitor (vorinostat) and one of four PARP inhibitors (olaparib, rucaparib, niraparib, or talazoparib). PARP inhibitors, when administered alone, showed no latency reversal activity. However, when combined with vorinostat, their efficacy increased threefold compared to vorinostat alone. This effect was mediated by the inhibition of tankyrase, a PARP superfamily member, which modulates the Hippo signaling pathway. In HIVGR670-infected primary cells, the combination reduced the reservoir size by 67%. In addition, talazoparib alone significantly reduced actively infected cells by 50%. Talazoparib-treated peripheral blood mononuclear cells co-cultured with K562 cells demonstrated enhanced NK-cell-mediated cytotoxicity, with a 10% reduction in K562 cell viability. These findings demonstrate that combining HDAC and PARP inhibitors augments latency reversal and reservoir reduction. With both the HDAC inhibitors and PARP inhibitors used in this study approved by the FDA for cancer treatment, this combination therapy holds strong potential for rapid clinical integration, contingent upon the confirmation of efficacy and safety in ongoing in vivo studies.
Collapse
Affiliation(s)
- Hasset Tibebe
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Dacia Marquez
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Aidan McGraw
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Sophia Gagliardi
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Cailyn Sullivan
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Grace Hillmer
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Kedhar Narayan
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Coco Izumi
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Adleigh Keating
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
| | - Taisuke Izumi
- Department of Biology, College of Arts & Sciences, American University, Washington, DC 20016, USA; (H.T.); (D.M.); (A.M.); (S.G.); (C.S.); (G.H.); (K.N.); (C.I.); (A.K.)
- District of Columbia Center for AIDS Research, Washington, DC 20052, USA
| |
Collapse
|
10
|
Liu X, Zhang M, Wang P, Zheng K, Wang X, Xie W, Pan X, Shen R, Liu R, Ding J, Wei Q. Nanoscale distribution of bioactive ligands on biomaterials regulates cell mechanosensing through translocation of actin into the nucleus. Proc Natl Acad Sci U S A 2025; 122:e2501264122. [PMID: 40042901 PMCID: PMC11912452 DOI: 10.1073/pnas.2501264122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cells respond to adhesive ligands such as arginine-glycine-aspartate (RGD) through integrins, which regulates cellular activities via influencing cytoskeleton assembly. Herein, we report that the nanoscale distribution of active ligands on biomaterials regulates cells through not only cytoplasmic tension but also nuclear tension. This is particularly related to translocation of actin into nucleus and highlighted in our interpretation of an "abnormal" phenomenon that large RGD nanospacing (>70 nm) disassembles integrin clusters, inhibits cell adhesion, but promotes osteogenic differentiation of mesenchymal stem cells. Our studies reveal that the unstable adhesion at the 150 nm RGD distance increases actin dynamics, resulting in the nuclear translocation of globular (G) actin. The compartment polymerization of more G-actins to filamentous actins in nucleus increases nuclear tension, facilitating transcription activity and releasing calcium ions from the endoplasmic reticulum. This noncanonical mechanotransduction process sheds insight into mechanotransduction pertinent to cell-material interactions.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan250012, China
| | - Man Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Biomedical Engineering, Sichuan University, Chengdu610065, China
| | - Peng Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology, Shenzhen518055, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Xinlei Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Wenyan Xie
- National Key Laboratory of Biotherapy, Sichuan University, Chengdu610041, China
| | - Xiaokai Pan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Runjia Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Qiang Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| |
Collapse
|
11
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
12
|
Guo P, Wan S, Guan KL. The Hippo pathway: Organ size control and beyond. Pharmacol Rev 2025; 77:100031. [PMID: 40148032 DOI: 10.1016/j.pharmr.2024.100031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
The Hippo signaling pathway is a highly conserved signaling network for controlling organ size, tissue homeostasis, and regeneration. It integrates a wide range of intracellular and extracellular signals, such as cellular energy status, cell density, hormonal signals, and mechanical cues, to modulate the activity of YAP/TAZ transcriptional coactivators. A key aspect of Hippo pathway regulation involves its spatial organization at the plasma membrane, where upstream regulators localize to specific membrane subdomains to regulate the assembly and activation of the pathway components. This spatial organization is critical for the precise control of Hippo signaling, as it dictates the dynamic interactions between pathway components and their regulators. Recent studies have also uncovered the role of biomolecular condensation in regulating Hippo signaling, adding complexity to its control mechanisms. Dysregulation of the Hippo pathway is implicated in various pathological conditions, particularly cancer, where alterations in YAP/TAZ activity contribute to tumorigenesis and drug resistance. Therapeutic strategies targeting the Hippo pathway have shown promise in both cancer treatment, by inhibiting YAP/TAZ signaling, and regenerative medicine, by enhancing YAP/TAZ activity to promote tissue repair. The development of small molecule inhibitors targeting the YAP-TEAD interaction and other upstream regulators offers new avenues for therapeutic intervention. SIGNIFICANCE STATEMENT: The Hippo signaling pathway is a key regulator of organ size, tissue homeostasis, and regeneration, with its dysregulation linked to diseases such as cancer. Understanding this pathway opens new possibilities for therapeutic approaches in regenerative medicine and oncology, with the potential to translate basic research into improved clinical outcomes.
Collapse
Affiliation(s)
- Pengfei Guo
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| | - Sicheng Wan
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
13
|
Katayama Y, Iwasaki T, Yamamoto T, Shimada N, Nakashima M, Toya M, Narutomi F, Tomonaga T, Kato K, Oda Y. Loss of SMARCA4 induces sarcomatogenesis through epithelial-mesenchymal transition in ovarian carcinosarcoma. Cancer Sci 2025; 116:835-845. [PMID: 39716847 PMCID: PMC11875775 DOI: 10.1111/cas.16423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Ovarian carcinosarcoma (OCS) is a rare and aggressive tumor, and the development of its sarcomatous component is believed to be due to epithelial-mesenchymal transition (EMT). The SWIch/sucrose nonfermentable chromatin remodeling factor (CRF) is closely related to EMT; however, the relationship between CRF and EMT in OCS remains unclear. In this study, we analyzed the protein expression of CRFs, including ARID1A and SMARCA4, and their downstream mRNA expression in 28 OCS cases, two fallopian tube CS cases, and one peritoneal CS case. ARID1A and SMARCA4 exhibited a histological type-specific loss of protein expression in 5 of 11 (45%) endometrioid cases and all 5 serous/homologous OCS cases, respectively. The mRNA analysis suggested that sarcomatogenesis is induced by the transforming growth factor-β and Hippo signaling pathways, both of which regulate YAP1. Immunostaining for YAP1 suggested YAP1-associated sarcomatogenesis in the CRF-retained group, whereas YAP1-unassociated sarcomatogenesis was suggested in the CRF-reduced group. High-grade serous carcinoma cell line experiments showed that the transcriptome of the SMARCA4-knockdown group showed lower expression of the epithelial gene CDH1 and higher expression of mesenchymal genes such as VIM, ZEB1, and SNAI1 than the control group. Moreover, cell adhesion disappeared and cell morphology changed to a spindle shape, indicating sarcomatogenesis. In conclusion, this study reveals a mechanism for sarcoma development in OCS and provides novel therapeutic possibilities.
Collapse
Affiliation(s)
- Yoshihiro Katayama
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Departments of Gynecology and Obstetrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takeo Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Naomi Shimada
- Department of GynecologyKyushu University Beppu HospitalBeppuJapan
| | - Miya Nakashima
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Departments of Gynecology and Obstetrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masato Toya
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Departments of Gynecology and Obstetrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Fumiya Narutomi
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Takumi Tomonaga
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kiyoko Kato
- Departments of Gynecology and Obstetrics, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
14
|
Zhu Y, Zhu J, Wang X, Wang P, Liu R. Molecular roles in membrane receptor signaling pathways and cascade reactions in chondrocytes: a review. J Mol Histol 2025; 56:94. [PMID: 39988650 DOI: 10.1007/s10735-025-10368-9] [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: 10/11/2023] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
Articular cartilage (AC) is a specialized connective tissue with unique biological and mechanical properties, which depends on the biological effects of each resident chondrocyte and its surrounding extracellular matrix (ECM) to form a unit that operates in a constant and balanced feedback loop. The surface membrane receptors of chondrocytes play a crucial role in the feedback balance of this biological unit. Various biological signals outside chondrocytes, such as water-soluble chemical signal molecules and mechanical signals, are unable to directly enter the cell and must first bind to the plasma membrane receptors to induce changes in the level and activity of intracellular signal transduction molecules. These changes then transmit through signaling cascade pathways into the nucleus, changing the cell phenotype, and producing physiological or pathological changes. Specific chemical and mechanical signals break the feedback balance of cartilage tissue units through membrane receptors. In the ECM environment, the molecular actions of chondrocyte membrane receptors in response to these specific signals, along with associated ion channel receptors, collectively regulate the biological effects of chondrocytes. This leads to decreased chondrocyte survival and an imbalance in ECM regulation, ultimately disrupting the tissue's molecular framework and physiological feedback mechanisms, and resulting in pathological changes in cartilage tissue. To provide insights into addressing the complexities associated with cartilage tissue injury and repair engineering, this review provides a comprehensive overview of the molecular mechanisms and biological implications of chondrocyte membrane receptor-mediated signal transduction, including G protein-coupled receptors (GPCRs), enzyme-linked receptors (tyrosine kinase receptors (TKRs)), and integrin receptors.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jingjing Zhu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Pengbo Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
15
|
Kelebeev J, MacKeracher A, Miyake T, McDermott JC. TAZ interactome analysis using nanotrap-based affinity purification-mass spectrometry. J Cell Sci 2025; 138:jcs263527. [PMID: 39898439 PMCID: PMC11928053 DOI: 10.1242/jcs.263527] [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: 09/05/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025] Open
Abstract
Characterization of protein-protein interactions (PPIs) is a fundamental goal in the post-genomic era. Here, we document a generally applicable approach to identify cellular protein interactomes using a combination of nanobody-based affinity purification (AP) coupled with liquid chromatography and tandem mass spectrometry (LC-MS/MS). The Hippo signaling regulator TAZ (also known as WWTR1) functions as a transcriptional co-repressor or activator depending on its PPI network; we therefore undertook an unbiased proteomic screen to identify TAZ PPIs in striated muscle cells. A GFP nanotrap-based AP approach coupled with protein identification through LC-MS/MS was used to document a comprehensive list of known and novel TAZ interactome components. Informatic analysis of the interactome documented known components of the Hippo signaling pathway and multiple epigenetic regulators such as the NuRD, FACT and SWI/SNF complexes and the pro-myogenic CARM1 methyltransferase. Hippo pathway reporter gene (HOP/HIP) analysis indicated that CARM1 represses TAZ transcriptional co-activator function, promoting TAZ Ser89 phosphorylation and TAZ cytoplasmic sequestration. MS analysis revealed that CARM1 dimethylates TAZ at Arg77 in a PGPR*LAGG consensus peptide, resulting in enhanced TAZ Ser89 phosphorylation. These studies underline the utility of a nanobody-based AP approach for interactome analysis.
Collapse
Affiliation(s)
- Jonathan Kelebeev
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Anastasia MacKeracher
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - John C. McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| |
Collapse
|
16
|
Chatzifrangkeskou M, Stanly T, Koennig D, Campos-Soares L, Eyres M, Hasson A, Perdiou A, Vendrell I, Fischer R, Das S, Gardner S, Go S, Futcher B, Newton A, Skourides P, Szele F, O’Neill E. ATR-hippo drives force signaling to nuclear F-actin and links mechanotransduction to neurological disorders. SCIENCE ADVANCES 2025; 11:eadr5683. [PMID: 39951537 PMCID: PMC11827640 DOI: 10.1126/sciadv.adr5683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
The mechanical environment is sensed through cell-matrix contacts with the cytoskeleton, but how signals transit the nuclear envelope to affect cell fate decisions remains unknown. Nuclear actin coordinates chromatin motility during differentiation and genome maintenance, yet it remains unclear how nuclear actin responds to mechanical force. The DNA-damage kinase ataxia telangiectasia and Rad3-related protein (ATR) translocates to the nuclear envelope to protect the nucleus during cell motility or compression. Here, we show that ATR drives nuclear actin assembly via recruitment of Filamin-A to the inner nuclear membrane through binding of the hippo pathway scaffold and ATR substrate, RASSF1A. Moreover, we demonstrate how germline RASSF1 mutation disables nuclear mechanotransduction resulting in cerebral cortex thinning and associates with common psychological traits. Thus, defective mechanical-regulated pathways may contribute to complex neurological disorders.
Collapse
Affiliation(s)
- Maria Chatzifrangkeskou
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Tess Stanly
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Delia Koennig
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Luana Campos-Soares
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Department Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Michael Eyres
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alexander Hasson
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Alexandra Perdiou
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Iolanda Vendrell
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Roman Fischer
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sayoni Das
- PrecisionLife, Bankside, Long Hanborough, Oxford OX29 8LJ, UK
| | - Steve Gardner
- PrecisionLife, Bankside, Long Hanborough, Oxford OX29 8LJ, UK
| | - Simei Go
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Ben Futcher
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Ashley Newton
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Paris Skourides
- Department of Biological Sciences, University of Cyprus, P.O. Box 20537, 2109 Nicosia, Cyprus
| | - Francis Szele
- Department Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Eric O’Neill
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
17
|
Byfield FJ, Eftekhari B, Kaymak-Loveless K, Mandal K, Li D, Wells RG, Chen W, Brujic J, Bergamaschi G, Wuite GJL, Patteson AE, Janmey PA. Metabolically intact nuclei are fluidized by the activity of the chromatin remodeling motor BRG1. Biophys J 2025; 124:494-507. [PMID: 39616442 PMCID: PMC11866952 DOI: 10.1016/j.bpj.2024.11.3322] [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: 06/04/2024] [Revised: 10/29/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024] Open
Abstract
The structure and dynamics of the nucleus regulate cellular functions, with shape changes impacting cell motility. Although the nucleus is generally seen as the stiffest organelle in the cell, cells can nevertheless deform the nucleus to large strains by small mechanical stresses. Here, we show that the mechanical response of the cell nucleus exhibits active fluidization that is driven by the BRG1 motor of the SWI/SNF/BAF chromatin remodeling complex. Atomic force microscopy measurements show that the nucleus alters stiffness in response to the cell substrate stiffness, which is retained after the nucleus is isolated, and that the work of nuclear compression is mostly dissipated rather than elastically stored. Inhibiting BRG1 stiffens the nucleus and eliminates dissipation and nuclear remodeling both in isolated nuclei and in intact cells. These findings uncover a novel role of the BRG1 motor in nuclear mechanics, advancing our understanding of cell motility mechanisms.
Collapse
Affiliation(s)
- Fitzroy J Byfield
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Behnaz Eftekhari
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kaeli Kaymak-Loveless
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kalpana Mandal
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Li
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rebecca G Wells
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; NSF Science and Technology Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wenjun Chen
- Center for Soft Matter Research, New York University, New York, New York
| | - Jasna Brujic
- Center for Soft Matter Research, New York University, New York, New York
| | - Giulia Bergamaschi
- Faculty of Sciences/Division of Physics and Astronomy, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, the Netherlands
| | - Gijs J L Wuite
- Faculty of Sciences/Division of Physics and Astronomy, Vrije Universiteit Amsterdam, De Boelelaan, Amsterdam, the Netherlands
| | - Alison E Patteson
- Department of Physics and BioInspired Institute, Syracuse University, Syracuse, New York
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania; Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; NSF Science and Technology Center for Engineering MechanoBiology, University of Pennsylvania, Philadelphia, Pennsylvania; Laboratory of Magnetic Soft Materials, Department of Physics, University of Latvia, Riga, Latvia.
| |
Collapse
|
18
|
Guo D, Yao B, Shao W, Zuo J, Chang Z, Shi J, Hu N, Bao S, Chen M, Fan X, Li X. The Critical Role of YAP/BMP/ID1 Axis on Simulated Microgravity-Induced Neural Tube Defects in Human Brain Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410188. [PMID: 39656892 PMCID: PMC11792043 DOI: 10.1002/advs.202410188] [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: 08/24/2024] [Revised: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Integrated biochemical and biophysical signals regulate embryonic development. Correct neural tube formation is critical for the development of central nervous system. However, the role of microgravity in neurodevelopment and its underlying molecular mechanisms remain unclear. In this study, the effects of stimulated microgravity (SMG) on the development of human brain organoids are investigated. SMG impairs N-cadherin-based adherens junction formation, leading to neural tube defects associated with dysregulated self-renewal capacity and neuroepithelial disorganization in human brain organoids. Bulk gene expression analyses reveal that SMG alters Hippo and BMP signaling in brain organoids. The neuropathological deficits in SMG-treated organoids can be rescued by regulating YAP/BMP/ID1 axis. Furthermore, sing-cell RNA sequencing data show that SMG results in perturbations in the number and function of neural stem and progenitor cell subpopulations. One of these subpopulations senses SMG cues and transmits BMP signals to the subpopulation responsible for tube morphogenesis, ultimately affecting the proliferating cell population. Finally, SMG intervention leads to persistent neurologic damage even after returning to normal gravity conditions. Collectively, this study reveals molecular and cellular abnormalities associated with SMG during human brain development, providing opportunities for countermeasures to maintain normal neurodevelopment in space.
Collapse
Affiliation(s)
- Di Guo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Wen‐Wei Shao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jia‐Chen Zuo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Zhe‐Han Chang
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jian‐Xin Shi
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Nan Hu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Shuang‐Qing Bao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Meng‐Meng Chen
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiu Fan
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiao‐Hong Li
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| |
Collapse
|
19
|
Ye Y, Xie W, Wang X, Tan S, Yang L, Ma Z, Zhu Z, Chen X, Liu X, O'Neill E, Chang L, Zhang W. DNA-damage orchestrates self-renewal and differentiation via reciprocal p53 family and Hippo/Wnt/TGF-β pathway activation in embryonic stem cells. Cell Mol Life Sci 2025; 82:38. [PMID: 39762370 PMCID: PMC11704118 DOI: 10.1007/s00018-024-05561-0] [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: 04/17/2024] [Revised: 11/18/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025]
Abstract
The mechanism by which DNA-damage affects self-renewal and pluripotency remains unclear. DNA damage and repair mechanisms have been largely elucidated in mutated cancer cells or simple eukaryotes, making valid interpretations on early development difficult. Here we show the impact of ionizing irradiation on the maintenance and early differentiation of mouse embryonic stem cells (ESCs). Our findings demonstrate that irradiation induces the upregulation of the p53 family genes, including p53, p63, and p73, resulting in elevated expression of the E3 ubiquitin ligases Itch and Trim32. Consequently, this impairs ESC maintenance by reducing the protein levels of key pluripotency transcription factors in both mouse ESCs and early embryos. Notably, our study reveals that irradiation-induced DNA damage leads to the recruitment of the BAF complex, causing it to dissociate from its binding sites on the target genes associated with the Yap, Wnt, and TGF-β pathways, thereby increasing signaling and promoting differentiation of ESCs into all three lineages. Importantly, pathway inhibition demonstrates that DNA damage accelerated ESC differentiation relies on Wnt and TGF-β, and is selectively dependent on p53 or p63/ p73 for mesoderm and endoderm respectively. Finally, our study reveals that p53 family proteins form complexes with effector proteins of key signaling pathways which actively contribute to ESC differentiation. In summary, this study uncovered a mechanism by which multiple differentiation signaling pathways converge on the p53 family genes to promote ESC differentiation and are impacted by exposure to ionizing radiation.
Collapse
Affiliation(s)
- Ying Ye
- Department of Clinical Pathobiology and Immunological Testing, School of Medical Laboratory, Qilu Medical University, Zibo, 255300, China
| | - Wenyan Xie
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Xuepeng Wang
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Shuping Tan
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Lingyue Yang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, Guangdong, China
| | - Zhaoru Ma
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Zhexin Zhu
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, 4090 Guanhai Road, Heifei, China
| | - Xi Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xiaoyu Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK.
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Province International Joint Laboratory for Regeneration Medicine, Medical College of Soochow University, Suzhou, China.
| | - Wensheng Zhang
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
20
|
Ng CS, Qin J. Switch/Sucrose Nonfermentable-Deficient Tumors-Morphology, Immunophenotype, Genetics, Epigenetics, Nosology, and Therapy. J Transl Med 2025; 105:102185. [PMID: 39542101 DOI: 10.1016/j.labinv.2024.102185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
About 20% of human cancers harbor mutations of genes encoding switch/sucrose nonfermentable (SWI/SNF) complex subunits. Deficiency of subunits of the complex is present in 10% of non-small-cell lung cancers (NSCLC; SMARCA4/SMARCA2 deficient), 100% thoracic SMARCA4/A2-deficient undifferentiated tumors (TSADUDT; SMARCA4/A2 deficient), malignant rhabdoid tumor, and atypical/teratoid tumor (SMARCB1-deficient), >90% of small cell carcinoma of the ovary, hypercalcemic type (SMARCA4/SMARCA2 deficient), frequently in undifferentiated/dedifferentiated endometrial carcinoma (SMARCA4, SMARCA2, SMARCB1, and ARID1A/B deficient), 100% SMARCA4 deficient undifferentiated uterine sarcoma (SMARCA4 deficient); and in various other tumors from multifarious anatomical sites. Silencing of SWI/SNF gene expression may be genomically or epigenetically driven, causing loss of tumor suppression function or facilitating other oncogenic events. The SWI/SNF-deficient tumors share the phenotype of poor or no differentiation, often with a variable component of rhabdoid tumor cells. They present at advanced stages with poor prognosis. Rhabdoid tumor cell phenotype is a useful feature to prompt investigation for this group of tumors. In the thoracic space, the overlap in morphology, immunophenotype, genetics, and epigenetics of SMARCA4/A2-deficient NSCLC and TSADUDT appears more significant. This raises a possible nosologic relationship between TSADUDT and SMARCA4/A2-deficient NSCLC. Increased understanding of the genetics, epigenetics, and mechanisms of oncogenesis in these poor prognostic tumors, which are often resistant to conventional treatment, opens a new horizon of therapy for the tumors.
Collapse
Affiliation(s)
- Chi Sing Ng
- Department of Pathology, Caritas Medical Center, Kowloon, Hong Kong.
| | - Jilong Qin
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
21
|
Maeda H, Sasaki H. Blastocoel expansion and AMOT degradation cooperatively promote YAP nuclear localization during epiblast formation. Dev Biol 2025; 517:234-247. [PMID: 39486633 DOI: 10.1016/j.ydbio.2024.10.007] [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: 07/04/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The epiblast is a pluripotent cell population formed in the late blastocyst stage of preimplantation embryos. During the process of epiblast formation from the inner cell mass (ICM) of the early blastocyst, activation of the Hippo pathway transcription factor TEAD by the nuclear translocation of the coactivator protein YAP is required for the robust expression of pluripotency factors. However, the mechanisms that alter YAP localization during epiblast formation remain unknown. Here, we reveal two such mechanisms. Expansion of the blastocoel promotes nuclear YAP localization by increasing cytoplasmic F-actin and reducing YAP phosphorylation. Additionally, cell differentiation regulates YAP. Expression of the junctional Hippo component, AMOT, gradually decreases during epiblast formation through a tankyrase-mediated degradation. SOX2 expression in the ICM is necessary for the reduction of AMOT and YAP phosphorylation. These two mechanisms function in parallel. Thus, the blastocoel-F-actin and SOX2-AMOT axes cooperatively suppress YAP phosphorylation and promote YAP nuclear localization during epiblast formation. The cooperation of these two distinct mechanisms likely contributes to the robustness of epiblast cell differentiation.
Collapse
Affiliation(s)
- Hinako Maeda
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
22
|
Jiang M, Fang H, Tian H. Metabolism of cancer cells and immune cells in the initiation, progression, and metastasis of cancer. Theranostics 2025; 15:155-188. [PMID: 39744225 PMCID: PMC11667227 DOI: 10.7150/thno.103376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
The metabolism of cancer and immune cells plays a crucial role in the initiation, progression, and metastasis of cancer. Cancer cells often undergo metabolic reprogramming to sustain their rapid growth and proliferation, along with meeting their energy demands and biosynthetic needs. Nevertheless, immune cells execute their immune response functions through the specific metabolic pathways, either to recognize, attack, and eliminate cancer cells or to promote the growth or metastasis of cancer cells. The alteration of cancer niches will impact the metabolism of both cancer and immune cells, modulating the survival and proliferation of cancer cells, and the activation and efficacy of immune cells. This review systematically describes the key characteristics of cancer cell metabolism and elucidates how such metabolic traits influence the metabolic behavior of immune cells. Moreover, this article also highlights the crucial role of immune cell metabolism in anti-tumor immune responses, particularly in priming T cell activation and function. By comprehensively exploring the metabolic crosstalk between cancer and immune cells in cancer niche, the aim is to discover novel strategies of cancer immunotherapy and provide effective guidance for clinical research in cancer treatment. In addition, the review also discusses current challenges such as the inadequacy of relevant diagnostic technologies and the issue of multidrug resistance, and proposes potential solutions including bolstering foundational cancer research, fostering technological innovation, and implementing precision medicine approaches. In-depth research into the metabolic effects of cancer niches can improve cancer treatment outcomes, prolong patients' survival period and enhance their quality of life.
Collapse
Affiliation(s)
- Mingxia Jiang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| | - Huayu Tian
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen University, Xiamen 361005, China
| |
Collapse
|
23
|
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
|
24
|
Fernandez MK, Sinha M, Zidan M, Renz M. Nuclear actin filaments - a historical perspective. Nucleus 2024; 15:2320656. [PMID: 38384139 PMCID: PMC10885181 DOI: 10.1080/19491034.2024.2320656] [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: 11/22/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024] Open
Abstract
The view on nuclear filaments formed by non-skeletal β-actin has significantly changed over the decades. Initially, filamentous actin was observed in amphibian oocyte nuclei and only under specific cell stress conditions in mammalian cell nuclei. Improved labeling and imaging technologies have permitted insights into a transient but microscopically apparent filament network that is relevant for chromatin organization, biomechanics of the mammalian cell nucleus, gene expression, and DNA damage repair. Here, we will provide a historical perspective on the developing insight into nuclear actin filaments.
Collapse
Affiliation(s)
| | - Molika Sinha
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| | - Mia Zidan
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| | - Malte Renz
- Gynecologic Oncology Division, School of Medicine Stanford University, Palo Alto, CA, USA
| |
Collapse
|
25
|
del Rosario-Gilabert D, Valenzuela-Miralles A, Esquiva G. Advances in mechanotransduction and sonobiology: effects of audible acoustic waves and low-vibration stimulations on mammalian cells. Biophys Rev 2024; 16:783-812. [PMID: 39830129 PMCID: PMC11735818 DOI: 10.1007/s12551-024-01242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/25/2024] [Indexed: 01/22/2025] Open
Abstract
In recent decades, research on mechanotransduction has advanced considerably, focusing on the effects of audible acoustic waves (AAWs) and low-vibration stimulation (LVS), which has propelled the field of sonobiology forward. Taken together, the current evidence demonstrates the influence of these biosignals on key cellular processes, such as growth, differentiation and migration in mammalian cells, emphasizing the determining role of specific physical parameters during stimulation, such as frequency, sound pressure level/amplitude and exposure time. These mechanical waves interact with various cellular elements, including ion channels, primary cilia, cell-cell adhesion receptors, cell-matrix and extracellular matrix proteins, and focal adhesion complexes. These components connect with the cytoskeletal fibre network, enabling the transmission of mechanical stimuli towards the nucleus. The nucleus, in turn, linked to the cytoskeleton via the linkers of the nucleoskeleton and cytoskeleton complex, acts as a mechanosensitive centre, not only responding to changes in cytoskeletal stiffness and nuclear tension but also regulating gene expression through the transcriptional co-activator YAP/TAZ and interactions between chromatin and the nuclear envelope. This intricate chain of mechanisms highlights the potential of sonobiology in various fields, including dentistry, regenerative medicine, tissue engineering and cancer research. However, progress in these fields requires the establishment of standardized measurement methodologies and biocompatible experimental setups to ensure the reproducibility of results.
Collapse
Affiliation(s)
- D. del Rosario-Gilabert
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Department of Physics, Systems Engineering and Signal Theory, University of Alicante, San Vicente del Raspeig, Spain
- Department of Computer Technology, University of Alicante, San Vicente del Raspeig, Spain
- Institute for Advanced Neuroscience of Barcelona (INAB), Barcelona, Spain
| | - A. Valenzuela-Miralles
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
| | - G. Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| |
Collapse
|
26
|
Pankratova MD, Riabinin AA, Butova EA, Selivanovskiy AV, Morgun EI, Ulianov SV, Vorotelyak EA, Kalabusheva EP. YAP/TAZ Signalling Controls Epidermal Keratinocyte Fate. Int J Mol Sci 2024; 25:12903. [PMID: 39684613 DOI: 10.3390/ijms252312903] [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: 11/05/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
The paralogues Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) control cell proliferation and cell fate determination from embryogenesis to ageing. In the skin epidermis, these proteins are involved in both homeostatic cell renewal and injury-induced regeneration and also drive carcinogenesis and other pathologies. YAP and TAZ are usually considered downstream of the Hippo pathway. However, they are the central integrating link for the signalling microenvironment since they are involved in the interplay with signalling cascades induced by growth factors, cytokines, and physical parameters of the extracellular matrix. In this review, we summarise the evidence on how YAP and TAZ are activated in epidermal keratinocytes; how YAP/TAZ-mediated signalling cooperates with other signalling molecules at the plasma membrane, cytoplasmic, and nuclear levels; and how YAP/TAZ ultimately controls transcription programmes, defining epidermal cell fate.
Collapse
Affiliation(s)
- Maria D Pankratova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Andrei A Riabinin
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elizaveta A Butova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Arseniy V Selivanovskiy
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Elena I Morgun
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Sergey V Ulianov
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ekaterina A Vorotelyak
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Ekaterina P Kalabusheva
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
27
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
28
|
Li XH, Guo D, Chen LQ, Chang ZH, Shi JX, Hu N, Chen C, Zhang XW, Bao SQ, Chen MM, Ming D. Low-intensity ultrasound ameliorates brain organoid integration and rescues microcephaly deficits. Brain 2024; 147:3817-3833. [PMID: 38739753 DOI: 10.1093/brain/awae150] [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: 11/08/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Human brain organoids represent a remarkable platform for modelling neurological disorders and a promising brain repair approach. However, the effects of physical stimulation on their development and integration remain unclear. Here, we report that low-intensity ultrasound significantly increases neural progenitor cell proliferation and neuronal maturation in cortical organoids. Histological assays and single-cell gene expression analyses revealed that low-intensity ultrasound improves the neural development in cortical organoids. Following organoid grafts transplantation into the injured somatosensory cortices of adult mice, longitudinal electrophysiological recordings and histological assays revealed that ultrasound-treated organoid grafts undergo advanced maturation. They also exhibit enhanced pain-related gamma-band activity and more disseminated projections into the host brain than the untreated groups. Finally, low-intensity ultrasound ameliorates neuropathological deficits in a microcephaly brain organoid model. Hence, low-intensity ultrasound stimulation advances the development and integration of brain organoids, providing a strategy for treating neurodevelopmental disorders and repairing cortical damage.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
29
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [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: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
30
|
Han Z, Shen Z, Pei J, You Q, Zhang Q, Wang L. Transformation of peptides to small molecules in medicinal chemistry: Challenges and opportunities. Acta Pharm Sin B 2024; 14:4243-4265. [PMID: 39525591 PMCID: PMC11544290 DOI: 10.1016/j.apsb.2024.06.019] [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: 03/07/2024] [Revised: 05/14/2024] [Accepted: 06/11/2024] [Indexed: 11/16/2024] Open
Abstract
Peptides are native binders involved in numerous physiological life procedures, such as cellular signaling, and serve as ready-made regulators of biochemical processes. Meanwhile, small molecules compose many drugs owing to their outstanding advantages of physiochemical properties and synthetic convenience. A novel field of research is converting peptides into small molecules, providing a convenient portable solution for drug design or peptidomic research. Endowing properties of peptides onto small molecules can evolutionarily combine the advantages of both moieties and improve the biological druggability of molecules. Herein, we present eight representative recent cases in this conversion and elaborate on the transformation process of each case. We discuss the innovative technological methods and research approaches involved, and analyze the applicability conditions of the approaches and methods in each case, guiding further modifications of peptides to small molecules. Finally, based on the aforementioned cases, we summarize a general procedure for peptide-to-small molecule modifications, listing the technological methods available for each transformation step and providing our insights on the applicable scenarios for these methods. This review aims to present the progress of peptide-to-small molecule modifications and propose our thoughts and perspectives for future research in this field.
Collapse
Affiliation(s)
- Zeyu Han
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zekai Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiayue Pei
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
31
|
Abu Sailik F, Emerald BS, Ansari SA. Opening and changing: mammalian SWI/SNF complexes in organ development and carcinogenesis. Open Biol 2024; 14:240039. [PMID: 39471843 PMCID: PMC11521604 DOI: 10.1098/rsob.240039] [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: 07/04/2024] [Accepted: 09/18/2024] [Indexed: 11/01/2024] Open
Abstract
The switch/sucrose non-fermentable (SWI/SNF) subfamily are evolutionarily conserved, ATP-dependent chromatin-remodelling complexes that alter nucleosome position and regulate a spectrum of nuclear processes, including gene expression, DNA replication, DNA damage repair, genome stability and tumour suppression. These complexes, through their ATP-dependent chromatin remodelling, contribute to the dynamic regulation of genetic information and the maintenance of cellular processes essential for normal cellular function and overall genomic integrity. Mutations in SWI/SNF subunits are detected in 25% of human malignancies, indicating that efficient functioning of this complex is required to prevent tumourigenesis in diverse tissues. During development, SWI/SNF subunits help establish and maintain gene expression patterns essential for proper cellular identity and function, including maintenance of lineage-specific enhancers. Moreover, specific molecular signatures associated with SWI/SNF mutations, including disruption of SWI/SNF activity at enhancers, evasion of G0 cell cycle arrest, induction of cellular plasticity through pro-oncogene activation and Polycomb group (PcG) complex antagonism, are linked to the initiation and progression of carcinogenesis. Here, we review the molecular insights into the aetiology of human malignancies driven by disruption of the SWI/SNF complex and correlate these mechanisms to their developmental functions. Finally, we discuss the therapeutic potential of targeting SWI/SNF subunits in cancer.
Collapse
Affiliation(s)
- Fadia Abu Sailik
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- ASPIRE Precision Medicine Research Institute Abu Dhabi (PMRI-AD), United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- ASPIRE Precision Medicine Research Institute Abu Dhabi (PMRI-AD), United Arab Emirates University, Al Ain, Abu Dhabi, UAE
| |
Collapse
|
32
|
Villares E, Gerecht S. Engineered Biomaterials and Model Systems to Study YAP/TAZ in Cancer. ACS Biomater Sci Eng 2024; 10:5550-5561. [PMID: 39190867 DOI: 10.1021/acsbiomaterials.4c01170] [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] [Indexed: 08/29/2024]
Abstract
The transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are master regulators involved in a multitude of cancer types and a wide range of tumorigenic events, including cancer stem cell renewal, invasion, metastasis, tumor precursor emergence, and drug resistance. YAP/TAZ are known to be regulated by several external cues and stimuli, such as extracellular matrix stiffness, cell spreading, cell geometry, and shear stress. Therefore, there is a need in the field of cancer research to develop and design relevant in vitro models that can accurately reflect the complex biochemical and biophysical cues of the tumor microenvironment central to the YAP/TAZ signaling nexus. While much progress has been made, this remains a major roadblock to advancing research in this field. In this review, we highlight the current engineered biomaterials and in vitro model systems that can be used to advance our understanding of how YAP/TAZ shapes several aspects of cancer. We begin by discussing current 2D and 3D hydrogel systems that model the YAP/TAZ response to ECM stiffness. We then examine the current trends in organoid culture systems and the use of microfluidics to model the effects of cellular density and shear stress on YAP/TAZ. Finally, we analyze the ongoing pitfalls of the present models used and important future directions in engineering systems that will advance our current knowledge of YAP/TAZ in cancer.
Collapse
Affiliation(s)
- Emma Villares
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| |
Collapse
|
33
|
Shu Y, Jin X, Ji M, Zhang Z, Wang X, Liang H, Lu S, Dong S, Lin Y, Guo Y, Zhuang Q, Wang Y, Lei Z, Guo L, Meng X, Zhou G, Zhang W, Chang L. Ku70 Binding to YAP Alters PARP1 Ubiquitination to Regulate Genome Stability and Tumorigenesis. Cancer Res 2024; 84:2836-2855. [PMID: 38862269 DOI: 10.1158/0008-5472.can-23-4034] [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: 12/19/2023] [Revised: 04/16/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Yes-associated protein (YAP) is a central player in cancer development, with functions extending beyond its recognized role in cell growth regulation. Recent work has identified a link between YAP/transcriptional coactivator with PDZ-binding motif (TAZ) and the DNA damage response. Here, we investigated the mechanistic underpinnings of the cross-talk between DNA damage repair and YAP activity. Ku70, a key component of the nonhomologous end joining pathway to repair DNA damage, engaged in a dynamic competition with TEAD4 for binding to YAP, limiting the transcriptional activity of YAP. Depletion of Ku70 enhanced interaction between YAP and TEAD4 and boosted YAP transcriptional capacity. Consequently, Ku70 loss enhanced tumorigenesis in colon cancer and hepatocellular carcinoma (HCC) in vivo. YAP impeded DNA damage repair and elevated genome instability by inducing PARP1 degradation through the SMURF2-mediated ubiquitin-proteasome pathway. Analysis of samples from patients with HCC substantiated the link between Ku70 expression, YAP activity, PARP1 levels, and genome instability. In conclusion, this research provides insight into the mechanistic interactions between YAP and key regulators of DNA damage repair, highlighting the role of a Ku70-YAP-PARP1 axis in preserving genome stability. Significance: Increased yes-associated protein transcriptional activity stimulated by loss of Ku70 induces PARP1 degradation by upregulating SMURF2 to inhibit DNA damage, driving genome instability and tumorigenesis.
Collapse
Affiliation(s)
- Yinyin Shu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xiaoni Jin
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Mintao Ji
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhisen Zhang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xiuxiu Wang
- Department of Anatomy, Wannan Medical College, Wuhu, China
| | - Haisheng Liang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shuangshuang Lu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shuai Dong
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yiping Lin
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yuhan Guo
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, P. R. China
| | - Yuhong Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhe Lei
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuanyu Meng
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wensheng Zhang
- Suzhou Medical College of Soochow University, Suzhou, China
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Jeong W, Kwon H, Park SK, Lee IS, Jho EH. Retinoic acid-induced protein 14 links mechanical forces to Hippo signaling. EMBO Rep 2024; 25:4033-4061. [PMID: 39160347 PMCID: PMC11387738 DOI: 10.1038/s44319-024-00228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024] Open
Abstract
Cells sense and respond to various mechanical forces from the extracellular matrix primarily by modulating the actin cytoskeleton. Mechanical forces can be translated into biochemical signals in a process called mechanotransduction. Yes-associated protein (YAP) is an effector of Hippo signaling and a mediator of mechanotransduction, but how mechanical forces regulate Hippo signaling is still an open question. We propose that retinoic acid-induced protein 14 (RAI14) responds to mechanical forces and regulates Hippo signaling. RAI14 positively regulates the activity of YAP. RAI14 interacts with NF2, a key component of the Hippo pathway, and the interaction occurs on filamentous actin. When mechanical forces are kept low in cells, NF2 dissociates from RAI14 and filamentous actin, resulting in increased interactions with LATS1 and activation of the Hippo pathway. Clinical data show that tissue stiffness and expression of RAI14 and YAP are upregulated in tumor tissues and that RAI14 is strongly associated with adverse outcome in patients with gastric cancer. Our data suggest that RAI14 links mechanotransduction with Hippo signaling and mediates Hippo-related biological functions such as cancer progression.
Collapse
Affiliation(s)
- Wonyoung Jeong
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Hyeryun Kwon
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - In-Seob Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea.
| |
Collapse
|
35
|
Mo Y, Sun L, Li S, Luo L, Liu H, Huang S, Chen Z, Li G. The mechanism of INO80D involved in chromatin remodeling regulating spermatogenesis in Chinese mitten crab (Eriocheir sinensis). Mol Genet Genomics 2024; 299:83. [PMID: 39212752 DOI: 10.1007/s00438-024-02177-8] [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: 04/12/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
The INO80D protein, a component of the INO80 chromatin remodeling complex, plays a pivotal role in chromatin remodeling, gene expression, and DNA repair within mammalian sperm. In contrast to the condensed nuclear structure of mammalian sperm, Chinese mitten crab, Eriocheir sinensis, exhibits a distinctively decondensed sperm nucleus. The distribution and function of INO80D during the E. sinensis spermatogenesis were previously enigmatic. Our research endeavored to elucidate the distribution and function of INO80D, thereby enhancing our comprehension of sperm decondensation and the process of spermatogenesis in this species. Employing transcriptome sequencing, RT-qPCR, western blot analysis, and immunofluorescence techniques, we observed a pronounced upregulation of INO80D in the adult E. sinensis in comparison to the juvenile. The protein predominantly resides in the cellular nucleus, with high levels in spermatogonia and spermatocytes, less in stage I and III spermatids, and lowest in mature sperm. The results indicated that INO80D is initially instrumental in chromatin decondensation to facilitate gene accessibility and DNA repair during the early phases of spermatogenesis. Its role subsequently shifts to maintaining decondensed chromatin stability and genetic integrity during spermiogenesis. The sustained presence of INO80D during spermiogenesis is essential for the ultimate maturation of the decondensed sperm nucleus, imperative for preserving the unique decondensed state and the protection of genetic material in E. sinensis. Our study concludes that INO80D exerts a multifaceted influence on the spermatogenesis of E. sinensis, impacting chromatin decondensation, genetic integrity, and the regulation of early gene expression. This understanding could potentially improve crab breeding in aquaculture.
Collapse
Affiliation(s)
- Yinyin Mo
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Lishuang Sun
- Hainan General Hospital, Haikou, 570311, Hainan, China
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Shu Li
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Lvjing Luo
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Huiting Liu
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Shi Huang
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Zhengyu Chen
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Genliang Li
- Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
36
|
Jiang L, Yan C, Yi Y, Zhu L, Liu Z, Zhang D, Jiang W. Cell size regulates human endoderm specification through actomyosin-dependent AMOT-YAP signaling. Stem Cell Reports 2024; 19:1137-1155. [PMID: 39094563 PMCID: PMC11368700 DOI: 10.1016/j.stemcr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Cell size is a crucial physical property that significantly impacts cellular physiology and function. However, the influence of cell size on stem cell specification remains largely unknown. Here, we investigated the dynamic changes in cell size during the differentiation of human pluripotent stem cells into definitive endoderm (DE). Interestingly, cell size exhibited a gradual decrease as DE differentiation progressed with higher stiffness. Furthermore, the application of hypertonic pressure or chemical to accelerate the reduction in cell size significantly and specifically enhanced DE differentiation. By functionally intervening in mechanosensitive elements, we have identified actomyosin activity as a crucial mediator of both DE differentiation and cell size reduction. Mechanistically, the reduction in cell size induces actomyosin-dependent angiomotin (AMOT) nuclear translocation, which suppresses Yes-associated protein (YAP) activity and thus facilitates DE differentiation. Together, our study has established a novel connection between cell size diminution and DE differentiation, which is mediated by AMOT nuclear translocation. Additionally, our findings suggest that the application of osmotic pressure can effectively promote human endodermal lineage differentiation.
Collapse
Affiliation(s)
- Lai Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China; Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Ying Yi
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Lihang Zhu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
37
|
Morgan JE, Jaferi N, Shonibare Z, Huang GS. ARID1A in Gynecologic Precancers and Cancers. Reprod Sci 2024; 31:2150-2162. [PMID: 38740655 DOI: 10.1007/s43032-024-01585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
The highest frequency of genetic alterations in the tumor suppressor ARID1A occurs in malignancies of the female reproductive tract. The prevalence of ARID1A alterations in gynecologic precancers and cancers is summarized from the literature, and the putative mechanisms of tumor suppressive action examined both in benign/precursor lesions including endometriosis and atypical hyperplasia and in malignancies of the ovary, uterus, cervix and vagina. ARID1A alterations in gynecologic cancers are usually loss-of-function mutations, resulting in diminished or absent protein expression. ARID1A deficiency results in pleiotropic downstream effects related not only to its role in transcriptional regulation as a SWI/SNF complex subunit, but also related to the functions of ARID1A in DNA replication and repair, immune modulation, cell cycle progression, endoplasmic reticulum (ER) stress and oxidative stress. The most promising actionable signaling pathway interactions and therapeutic vulnerabilities of ARID1A mutated cancers are presented with a critical review of the currently available experimental and clinical evidence. The role of ARID1A in response to chemotherapeutic agents, radiation therapy and immunotherapy is also addressed. In summary, the multi-faceted role of ARID1A mutation in precancer and cancer is examined through a clinical lens focused on development of novel preventive and therapeutic interventions for gynecological cancers.
Collapse
Affiliation(s)
- Jaida E Morgan
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Nishah Jaferi
- Yale College, Yale University, New Haven, Connecticut, USA
| | - Zainab Shonibare
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gloria S Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, USA.
- Department of Obstetrics, Gynecology & Reproductive Sciences, Division of Gynecologic Oncology, Yale School of Medicine, Yale Cancer Center, Yale University, PO Box 208063, New Haven, CT, 06520-8063, USA.
| |
Collapse
|
38
|
Xu S, Zhu C, Xu Q, An Z, Xu S, Xuan G, Lin C, Tang C. ARID1A restrains EMT and stemness of ovarian cancer cells through the Hippo pathway. Int J Oncol 2024; 65:76. [PMID: 38873993 PMCID: PMC11251745 DOI: 10.3892/ijo.2024.5664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
Genes encoding subunits of SWI/SNF (BAF) chromatin‑remodeling complexes are recurrently mutated in a broad array of tumor types, and among the subunits, ARID1A is the most frequent target with mutations. In the present study, it was reported that ARID1A inhibits the epithelial‑mesenchymal transition (EMT) and stemness of ovarian cancer cells, accompanied by reduced cell viability, migration and colony formation, suggesting that ARID1A acts as a tumor suppressor in ovarian cancer. Mechanistically, ARID1A exerts its inhibitory effects on ovarian cancer cells by activating the Hippo signaling pathway. Conversely, the overexpression of a gain‑of‑function transcriptional co‑activator with PDZ‑binding motif (TAZ) mutant (TAZ‑Ser89) effectively reverses the effects induced by ARID1A. In addition, activation of Hippo signaling apparently upregulates ARID1A protein expression, whereas ectopic expression of TAZ‑Ser89 results in the markedly decreased ARID1A levels, indicating a feedback of ARID1A‑TAZ in regulating ovarian cancer cell EMT and stemness. Thus, the present study uncovered the role of ARID1A through the Hippo/TAZ pathway in modulating EMT and stemness of ovarian cancer cells, and providing with evidence that TAZ inhibitors could effectively prevent initiation and metastasis of ovarian cancer cases where ARID1A is lost or mutated.
Collapse
Affiliation(s)
- Shouying Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Chongying Zhu
- The Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Zihao An
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Shu Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Ge Xuan
- Department of Gynecology, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315012, P.R. China
| | - Chao Lin
- Department of Neurosurgery, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| |
Collapse
|
39
|
Liang H, Zheng X, Zhang X, Zhang Y, Zheng J. The role of SWI/SNF complexes in digestive system neoplasms. Med Oncol 2024; 41:119. [PMID: 38630164 DOI: 10.1007/s12032-024-02343-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/22/2024] [Indexed: 04/19/2024]
Abstract
Chromatin remodeling is a critical step in the DNA damage response, and the ATP-dependent chromatin remodelers are a group of epigenetic regulators that alter nucleosome assembly and regulate transcription factor accessibility to DNA, preventing genomic instability and tumorigenesis caused by DNA damage. The SWI/SNF chromatin remodeling complex is one of them, and mutations in the gene encoding the SWI/SNF subunit are frequently found in digestive tumors. We review the most recent literature on the role of SWI/SNF complexes in digestive tumorigenesis, with different SWI/SNF subunits playing different roles. They regulate the biological behavior of tumor cells, participate in multiple signaling pathways, interact with multiple genes, and have some correlation with the prognosis of patients. Their carcinogenic properties may help discover new therapeutic targets. Understanding the mutations and defects of SWI/SNF complexes, as well as the underlying functional mechanisms, may lead to new strategies for treating the digestive system by targeting relevant genes or modulating the tumor microenvironment.
Collapse
Affiliation(s)
- Hanyun Liang
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang, 261053, China
| | - Xin Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang, 261053, China
| | - Xiao Zhang
- Department of Ultrasound, Weifang People's Hospital, Weifang, 261041, China
| | - Yan Zhang
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, 261053, China.
| | - Jie Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang, 261053, China.
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang, 261053, China.
| |
Collapse
|
40
|
Byfield FJ, Eftekhari B, Kaymak-Loveless K, Mandal K, Li D, Wells RG, Chen W, Brujic J, Bergamaschi G, Wuite GJL, Patteson AE, Janmey PA. Metabolically intact nuclei are fluidized by the activity of the chromatin remodeling motor BRG1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589275. [PMID: 38659735 PMCID: PMC11042217 DOI: 10.1101/2024.04.12.589275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The structure and dynamics of the cell nucleus regulate nearly every facet of the cell. Changes in nuclear shape limit cell motility and gene expression. Although the nucleus is generally seen as the stiffest organelle in the cell, cells can nevertheless deform the nucleus to large strains by small mechanical stresses. Here, we show that the mechanical response of the cell nucleus exhibits active fluidization that is driven by the BRG 1 motor of the SWI/SNF/BAF chromatin-remodeling complex. Atomic force microscopy measurements show that the nucleus alters stiffness in response to the cell substrate stiffness, which is retained after the nucleus is isolated and that the work of nuclear compression is mostly dissipated rather than elastically stored. Inhibiting BRG 1 stiffens the nucleus and eliminates dissipation and nuclear remodeling both in isolated nuclei and in intact cells. These findings demonstrate a novel link between nuclear motor activity and global nuclear mechanics.
Collapse
|
41
|
Lin S, He X, Wang Y, Chen Y, Lin A. Emerging role of lncRNAs as mechanical signaling molecules in mechanotransduction and their association with Hippo-YAP signaling: a review. J Zhejiang Univ Sci B 2024; 25:280-292. [PMID: 38584091 PMCID: PMC11009445 DOI: 10.1631/jzus.b2300497] [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: 07/10/2023] [Accepted: 12/11/2023] [Indexed: 04/09/2024]
Abstract
Cells within tissues are subject to various mechanical forces, including hydrostatic pressure, shear stress, compression, and tension. These mechanical stimuli can be converted into biochemical signals through mechanoreceptors or cytoskeleton-dependent response processes, shaping the microenvironment and maintaining cellular physiological balance. Several studies have demonstrated the roles of Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ) as mechanotransducers, exerting dynamic influence on cellular phenotypes including differentiation and disease pathogenesis. This regulatory function entails the involvement of the cytoskeleton, nucleoskeleton, integrin, focal adhesions (FAs), and the integration of multiple signaling pathways, including extracellular signal-regulated kinase (ERK), wingless/integrated (WNT), and Hippo signaling. Furthermore, emerging evidence substantiates the implication of long non-coding RNAs (lncRNAs) as mechanosensitive molecules in cellular mechanotransduction. In this review, we discuss the mechanisms through which YAP/TAZ and lncRNAs serve as effectors in responding to mechanical stimuli. Additionally, we summarize and elaborate on the crucial signal molecules involved in mechanotransduction.
Collapse
Affiliation(s)
- Siyi Lin
- College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyu He
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Ying Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Yu Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Cancer Center, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China.
- International School of Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310058, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
42
|
Zhang H, Li S, Zhou R, Dong T, Zhang X, Yu M, Lin J, Shi M, Geng E, Li J, Wang M, Huang L, Yang XP, Sun S. SRCAP complex promotes lung cancer progression by reprograming the oncogenic transcription of Hippo-YAP/TAZ signaling pathway. Cancer Lett 2024; 585:216667. [PMID: 38280479 DOI: 10.1016/j.canlet.2024.216667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
The activation of YAP/TAZ, a pair of paralogs of transcriptional coactivators, initiates a dysregulated transcription program, which is a key feature of human cancer cells. However, it is not fully understood how YAP/TAZ promote dysregulated transcription for tumor progression. In this study, we employed the BioID method to identify the interactome of YAP/TAZ and discovered that YAP/TAZ interact with multiple components of SRCAP complex, a finding that was further validated through endogenous and exogenous co-immunoprecipitation, as well as immunofluorescence experiments. CUT&Tag analysis revealed that SRCAP complex facilitates the deposition of histone variant H2A.Z at target promoters. The depletion of SRCAP complex resulted in a decrease in H2A.Z occupancy and the oncogenic transcription of YAP/TAZ target genes. Additionally, the blockade of SRCAP complex suppressed YAP-driven tumor growth. In a genetically engineered lung adenocarcinoma mouse model and non-small cell lung cancer patients, SRCAP complex and H2A.Z deposition were found to be upregulated. This upregulation was statistically correlated with YAP expression, pathological stages, and poor survival in lung cancer patients. Together, our study uncovers that SRCAP complex plays a critical role in YAP/TAZ oncogenic transcription by coordinating H2A.Z deposition during cancer progression, providing potential targets for cancer diagnosis and prevention.
Collapse
Affiliation(s)
- Huixia Zhang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Shasha Li
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Runxin Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Tianqi Dong
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Xiao Zhang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Man Yu
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Jiaming Lin
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Mingjun Shi
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Ershuo Geng
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Juebei Li
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Mingwei Wang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Liu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jie Fang Avenue, Wuhan, 430030, China
| | - Xiang-Ping Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
| | - Shuguo Sun
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China; Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
43
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
44
|
Mannion AJ, Zhao H, Zhang Y, von Wright Y, Bergman O, Roy J, Saharinen P, Holmgren L. Regulation of YAP Promotor Accessibility in Endothelial Mechanotransduction. Arterioscler Thromb Vasc Biol 2024; 44:666-689. [PMID: 38299356 PMCID: PMC10880945 DOI: 10.1161/atvbaha.123.320300] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Endothelial cells are constantly exposed to mechanical forces in the form of fluid shear stress, extracellular stiffness, and cyclic strain. The mechanoresponsive activity of YAP (Yes-associated protein) and its role in vascular development are well described; however, whether changes to transcription or epigenetic regulation of YAP are involved in these processes remains unanswered. Furthermore, how mechanical forces are transduced to the nucleus to drive transcriptional reprogramming in endothelial cells is poorly understood. The YAP target gene, AmotL2 (angiomotin-like 2), is a junctional mechanotransducer that connects cell-cell junctions to the nuclear membrane via the actin cytoskeleton. METHODS We applied mechanical manipulations including shear flow, stretching, and substrate stiffness to endothelial cells to investigate the role of mechanical forces in modulating YAP transcription. Using in vitro and in vivo endothelial depletion of AmotL2, we assess nuclear morphology, chromatin organization (using transposase-accessible chromatin sequencing), and whole-mount immunofluorescent staining of the aorta to determine the regulation and functionality of YAP. Finally, we use genetic and chemical inhibition to uncouple the nuclear-cytoskeletal connection to investigate the role of this pathway on YAP transcription. RESULTS Our results reveal that mechanical forces sensed at cell-cell junctions by the YAP target gene AmotL2 are directly involved in changes in global chromatin accessibility and activity of the histone methyltransferase EZH2, leading to modulation of YAP promotor activity. Functionally, shear stress-induced proliferation of endothelial cells in vivo was reliant on AmotL2 and YAP/TAZ (transcriptional coactivator with PDZ-binding motif) expression. Mechanistically, uncoupling of the nuclear-cytoskeletal connection from junctions and focal adhesions led to altered nuclear morphology, chromatin accessibility, and YAP promotor activity. CONCLUSIONS Our findings reveal a role for AmotL2 and nuclear-cytoskeletal force transmission in modulating the epigenetic and transcriptional regulation of YAP to maintain a mechano-enforced positive feedback loop of vascular homeostasis. These findings may offer an explanation as to the proinflammatory phenotype that leads to aneurysm formation observed in AmotL2 endothelial deletion models.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
- Department of Cell and Tissue Dynamics, Max Planck Institute of Molecular Biomedicine, Münster, Germany (A.J.M.)
| | - Honglei Zhao
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Yuanyuan Zhang
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Ylva von Wright
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
| | - Otto Bergman
- Medicine (O.B.), Karolinska Institute, Stockholm, Sweden
| | - Joy Roy
- Molecular Medicine and Surgery (J.R.), Karolinska Institute, Stockholm, Sweden
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden (J.R.)
| | - Pipsa Saharinen
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Finland (P.S.)
| | - Lars Holmgren
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
45
|
Hein KZ, Stephen B, Fu S. Therapeutic Role of Synthetic Lethality in ARID1A-Deficient Malignancies. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:41-52. [PMID: 38327752 PMCID: PMC10846636 DOI: 10.36401/jipo-22-37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/28/2023] [Accepted: 09/21/2023] [Indexed: 02/09/2024]
Abstract
AT-rich interaction domain 1A (ARID1A), a mammalian switch/sucrose nonfermenting complex subunit, modulates several cellular processes by regulating chromatin accessibility. It is encoded by ARID1A, an immunosuppressive gene frequently disrupted in a many tumors, affecting the proliferation, migration, and invasion of cancer cells. Targeting molecular pathways and epigenetic regulation associated with ARID1A loss, such as inhibiting the PI3K/AKT pathway or modulating Wnt/β-catenin signaling, may help suppress tumor growth and progression. Developing epigenetic drugs like histone deacetylase or DNA methyltransferase inhibitors could restore normal chromatin structure and function in cells with ARID1A loss. As ARID1A deficiency correlates with enhanced tumor mutability, microsatellite instability, high tumor mutation burden, increased programmed death-ligand 1 expression, and T-lymphocyte infiltration, ARID1A-deficient cells can be a potential therapeutic target for immune checkpoint inhibitors that warrants further exploration. In this review, we discuss the role of ARID1A in carcinogenesis, its crosstalk with other signaling pathways, and strategies to make ARID1A-deficient cells a potential therapeutic target for patients with cancer.
Collapse
Affiliation(s)
- Kyaw Z. Hein
- Department of Internal Medicine, HCA Florida Westside Hospital, Plantation, FL, USA
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Pelosi G, Melocchi V, Dama E, Hofman P, De Luca M, Albini A, Gemelli M, Ricotta R, Papotti M, La Rosa S, Uccella S, Harari S, Sonzogni A, Asiedu MK, Wigle DA, Bianchi F. An in-silico analysis reveals further evidence of an aggressive subset of lung carcinoids sharing molecular features of high-grade neuroendocrine neoplasms. Exp Mol Pathol 2024; 135:104882. [PMID: 38237798 DOI: 10.1016/j.yexmp.2024.104882] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/23/2023] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Little is known as to whether there may be any pathogenetic link between pulmonary carcinoids and neuroendocrine carcinomas (NECs). A gene signature we previously found to cluster pulmonary carcinoids, large cell neuroendocrine carcinoma (LCNEC) and small cell lung carcinoma (SCLC), and which encompassed MEN1, MYC, MYCL1, RICTOR, RB1, SDHA, SRC and TP53 mutations or copy number variations (CNVs), was used to reclassify an independent cohort of 54 neuroendocrine neoplasms (NENs) [31 typical carcinoids (TC), 11 atypical carcinoids (AC) and 12 SCLC], by means of transcriptome and mutation data. Unsupervised clustering analysis identified two histology-independent clusters, namely CL1 and CL2, where 17/42 (40.5%) carcinoids and all the SCLC samples fell into the latter. CL2 carcinoids affected survival adversely, were enriched in T to G transversions or T > C/C > T transitions in the context of specific mutational signatures, presented with at least 1.5-fold change (FC) increase of gene mutations including TSC2, SMARCA2, SMARCA4, ERBB4 and PTPRZ1, differed for gene expression and showed epigenetic changes in charge of MYC and MTORC1 pathways, cellular senescence, inflammation, high-plasticity cell state and immune system exhaustion. Similar results were also found in two other independent validation sets comprising 101 lung NENs (24 carcinoids, 21 SCLC and 56 LCNEC) and 30 carcinoids, respectively. We herein confirmed an unexpected sharing of molecular traits along the spectrum of lung NENs, with a subset of genomically distinct aggressive carcinoids sharing molecular features of high-grade neuroendocrine neoplasms.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy.
| | - Valentina Melocchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Elisa Dama
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025 and Centre Hospitalier Universitaire de Nice, FHU OncoAge, Université Côte d'Azur, 06100 Nice, France
| | - Marco De Luca
- Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | | | - Maria Gemelli
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Riccardo Ricotta
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| | - Stefano La Rosa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Silvia Uccella
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Sergio Harari
- Department of Medical Sciences and Community Health, University of Milan, Milan, Italy; Division of Pneumology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Angelica Sonzogni
- Department of Pathology and Laboratory Medicine, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michael K Asiedu
- Division of General Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dennis A Wigle
- Division of General Thoracic Surgery, Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
47
|
Cosgrove BD, Bounds LR, Taylor CK, Su AL, Rizzo AJ, Barrera A, Crawford GE, Hoffman BD, Gersbach CA. Mechanosensitive genomic enhancers potentiate the cellular response to matrix stiffness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574997. [PMID: 38260455 PMCID: PMC10802421 DOI: 10.1101/2024.01.10.574997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Epigenetic control of cellular transcription and phenotype is influenced by changes in the cellular microenvironment, yet how mechanical cues from these microenvironments precisely influence epigenetic state to regulate transcription remains largely unmapped. Here, we combine genome-wide epigenome profiling, epigenome editing, and phenotypic and single-cell RNA-seq CRISPR screening to identify a new class of genomic enhancers that responds to the mechanical microenvironment. These 'mechanoenhancers' could be active on either soft or stiff extracellular matrix contexts, and regulated transcription to influence critical cell functions including apoptosis, mechanotransduction, proliferation, and migration. Epigenetic editing of mechanoenhancers on rigid materials tuned gene expression to levels observed on softer materials, thereby reprogramming the cellular response to the mechanical microenvironment. These editing approaches may enable the precise alteration of mechanically-driven disease states.
Collapse
Affiliation(s)
- Brian D. Cosgrove
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Lexi R. Bounds
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Carson Key Taylor
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alan L. Su
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Anthony J. Rizzo
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University; Durham, NC 27708, USA
| | - Gregory E. Crawford
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Pediatrics, Duke University Medical Center; Durham, NC 27708, USA
| | - Brenton D. Hoffman
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
| | - Charles A. Gersbach
- Department of Biomedical Engineering, Duke University; Durham, NC 27708, USA
- Center for Advanced Genomic Technologies, Duke University; Durham, NC 27708, USA
- Department of Cell Biology, Duke University; Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center; Durham, NC 27708, USA
| |
Collapse
|
48
|
Li S, Wang L, Shi J, Chen Y, Xiao A, Huo B, Tian W, Zhang S, Yang G, Gong W, Zhang H. Chromatin accessibility complex subunit 1 enhances tumor growth by regulating the oncogenic transcription of YAP in breast and cervical cancer. PeerJ 2024; 12:e16752. [PMID: 38223760 PMCID: PMC10787542 DOI: 10.7717/peerj.16752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024] Open
Abstract
Background As a component of chromatin remodeling complex, chromatin accessibility complex subunit 1 (CHRAC1) is critical in transcription and DNA replication. However, the significance of CHRAC1 in cancer progression has not been investigated extensively. This research aimed to determine the function of CHRAC1 in breast and cervical cancer and elucidate the molecular mechanism. Methods The Bio-ID method was used to identify the interactome of transcriptional activator Yes-associated protein (YAP) and the binding between YAP and CHRAC1 was verified by immunofluorescence. CCK8, colony formation and subcutaneous xenograft assays were conducted to explore the function of CHRAC1 in cancer cell proliferation. RNA-seq analysis and RT-PCR were used to analyze the transcription program change after CHRAC1 ablation. The diagnostic value of CHRAC1 was analyzed by TCGA database and further validated by immunohistochemistry staining. Results In the current study, we found that the chromatin remodeler CHRAC1 was a potential YAP interactor. CHRAC1 depletion suppressed breast and cervical cancer cell proliferation and tumor growth. The potential mechanism may be that CHRAC1 interacts with YAP to facilitate oncogenic transcription of YAP target genes in Hippo pathway, thereby promoting tumorigenesis. CHRAC1 was elevated in cervical and breast cancer biopsies and the upregulation correlated with shorter survival, poor pathological stages and metastasis of cancer patients. Moreover, CHRAC1 expression was statistically associated with YAP in breast and cervical cancer biopsies. Conclusions These findings highlight that CHRAC1 contributes to cancer progression through regulating the oncogenic transcription of YAP, which makes it a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Shasha Li
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lulu Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Shi
- Xiangyang Center for Disease Control and Prevention, Xiangyang, China
| | - Yi Chen
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ang Xiao
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingyue Huo
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjing Tian
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shilu Zhang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Yang
- Xiangyang Center for Disease Control and Prevention, Xiangyang, China
| | - Wensheng Gong
- Xiangyang Center for Disease Control and Prevention, Xiangyang, China
| | - Huixia Zhang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Kazmi I, Altamimi ASA, Afzal M, Majami AA, Abbasi FA, Almalki WH, Alzera SI, Kukreti N, Fuloria NK, Fuloria S, Sekar M, Abida. Non-coding RNAs: Emerging biomarkers and therapeutic targets in ulcerative colitis. Pathol Res Pract 2024; 253:155037. [PMID: 38160482 DOI: 10.1016/j.prp.2023.155037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Ulcerative colitis (UC) is a persistent inflammatory condition affecting the colon's mucosal lining, leading to chronic bowel inflammation. Despite extensive research, the precise molecular mechanisms underlying UC pathogenesis remain elusive. NcRNAs form a category of functional RNA molecules devoid of protein-coding capacity. They have recently surfaced as pivotal modulators of gene expression and integral participants in various pathological processes, particularly those related to inflammatory disorders. The diverse classes of ncRNAs, encompassing miRNAs, circRNAs, and lncRNAs, have been implicated in UC. It highlights their involvement in key UC-related processes, such as immune cell activation, epithelial barrier integrity, and the production of pro-inflammatory mediators. ncRNAs have been identified as potential biomarkers for UC diagnosis and monitoring disease progression, offering promising avenues for personalized medicine. This approach may pave the way for novel, more specific treatments with reduced side effects, addressing the current limitations of conventional therapies. A comprehensive understanding of the interplay between ncRNAs and UC will advance our knowledge of the disease, potentially leading to more effective and personalized treatments for patients suffering from this debilitating condition. This review explores the pivotal role of ncRNAs in the context of UC, shedding light on their possible targets for diagnosis, prognosis, and therapeutic interventions.
Collapse
Affiliation(s)
- Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | | | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Abdullah A Majami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad Al Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzera
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Al-Jouf, Saudi Arabia
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | | | - Shivkanya Fuloria
- Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia
| | - Mahendran Sekar
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Abida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
50
|
Zhang Y, Yan HJ, Wu J. The Tumor Immune Microenvironment plays a Key Role in Driving the Progression of Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:681-700. [PMID: 38213139 DOI: 10.2174/0115680096267791231115101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 01/13/2024]
Abstract
Cholangiocarcinoma (CCA) is an epithelial cancer distinguished by bile duct cell differentiation and is also a fibroproliferative tumor. It is characterized by a dense mesenchyme and a complex tumor immune microenvironment (TME). The TME comprises both cellular and non-cellular components. The celluar component includes CCA cells, immune cells and mesenchymal cells represented by the cancer-associated fibroblasts (CAFs), while the non-cellular component is represented by mesenchymal elements such as the extracellular matrix (ECM). Recent studies have demonstrated the important role of the TME in the development, progression, and treatment resistance of CCA. These cell-associated prognostic markers as well as intercellular connections, may serve as potential therapeutic targets and could inspire new treatment approaches for CCA in the future. This paper aims to summarize the current understanding of CCA's immune microenvironment, focusing on immune cells, mesenchymal cells, ECM, intercellular interactions, and metabolism within the microenvironment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Hai-Jiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| |
Collapse
|