1
|
Haripriya E, Hemalatha K, Matada GSP, Pal R, Das PK, Ashadul Sk MD, Mounika S, Viji MP, Aayishamma I, Jayashree KR. Advancements of anticancer agents by targeting the Hippo signalling pathway: biological activity, selectivity, docking analysis, and structure-activity relationship. Mol Divers 2025; 29:2829-2862. [PMID: 39436581 DOI: 10.1007/s11030-024-11009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
The Hippo signalling pathway is prominent and governs cell proliferation and stem cell activity, acting as a growth regulator and tumour suppressor. Defects in Hippo signalling and hyperactivation of its downstream effector's Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play roles in cancer development, implying that pharmacological inhibition of YAP and TAZ activity could be an effective cancer treatment strategy. Conversely, YAP and TAZ can also have beneficial effects in promoting tissue repair and regeneration following damage, therefore their activation may be therapeutically effective in certain instances. Recently, a complex network of intracellular and extracellular signalling mechanisms that affect YAP and TAZ activity has been uncovered. The YAP/TAZ-TEAD interaction leads to tumour development and the protein structure of YAP/TAZ-TEAD includes three interfaces and one hydrophobic pocket. There are clinical and preclinical trial drugs available to inhibit the hippo signalling pathway, but these drugs have moderate to severe side effects, so researchers are in search of novel, potent, and selective hippo signalling pathway inhibitors. In this review, we have discussed the hippo pathway in detail, including its structure, activation, and role in cancer. We have also provided the various inhibitors under clinical and preclinical trials, and advancement of small molecules their detailed docking analysis, structure-activity relationship, and biological activity. We anticipate that the current study will be a helpful resource for researchers.
Collapse
Affiliation(s)
- E Haripriya
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K Hemalatha
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Pronoy Kanti Das
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M D Ashadul Sk
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - S Mounika
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M P Viji
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - I Aayishamma
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K R Jayashree
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| |
Collapse
|
2
|
Li H, Li S, Kanamori Y, Liu S, Moroishi T. Auranofin resensitizes ferroptosis-resistant lung cancer cells to ferroptosis inducers. Biochem Biophys Res Commun 2025; 770:151992. [PMID: 40373379 DOI: 10.1016/j.bbrc.2025.151992] [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: 02/01/2025] [Revised: 03/23/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
Lung cancer, a major cause of cancer-related mortality, has limited therapeutic options, especially for advanced cases. Ferroptosis, an iron-dependent form of cell death, is a potential therapeutic strategy for this disease; however, resistance mechanisms in the tumor microenvironment impede its effectiveness. Therefore, in this study, we aimed to investigate the efficacy of sulfasalazine (SAS), a ferroptosis inducer, and auranofin (AUR), a Food and Drug Administration-approved anti-inflammatory agent, combination to counteract ferroptosis resistance in lung cancer. SAS induced ferroptosis in vitro; however, its efficacy in vivo was limited, possibly because of factors, such as nutrient deprivation and high cell density, in the microenvironment that suppressed the activities of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of ferroptosis resistance. Screening of 2483 drugs revealed AUR as a compound resensitizing the YAP/TAZ-deficient lung cancer cells to ferroptosis. Moreover, SAS and AUR combination significantly enhanced lipid peroxidation and reactive oxygen species accumulation, further driving ferroptosis in cells. This combination effectively inhibited tumor growth and enhanced survival in a murine lung cancer model. Overall, our findings suggest that AUR potentiates ferroptosis-based therapies, serving as an effective candidate to overcome ferroptosis resistance in lung cancer.
Collapse
Affiliation(s)
- Hao Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Shuran Li
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yohei Kanamori
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Saisai Liu
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan; Division of Cellular Dynamics, Medical Research Laboratory, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Tokyo, 113-8510, Japan.
| |
Collapse
|
3
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. Dev Biol 2025; 524:105-115. [PMID: 40349907 DOI: 10.1016/j.ydbio.2025.05.006] [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: 02/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
During the first cell fate decision in mammalian embryos, the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell and nuclear shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
4
|
Harding-Fox SL, Cellek S. The role of cyclic adenosine monophosphate (cAMP) in pathophysiology of fibrosis. Drug Discov Today 2025; 30:104368. [PMID: 40318753 DOI: 10.1016/j.drudis.2025.104368] [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: 02/05/2025] [Revised: 04/11/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Fibrosis, the excessive production and disorganised deposition of extracellular matrix proteins, can occur in any organ system, disrupting functionality and causing fatality. The number, efficacy and safety of antifibrotic drugs are incredibly limited. Therapeutics which elevate intracellular cyclic adenosine monophosphate (cAMP) offer a potential solution. In this review, we present the signalling mechanisms involved in fibrosis pathophysiology, how cAMP and its effectors might interact with these pathways, and the current preclinical and clinical efforts in this field. cAMP elevating agents have the potential to be future antifibrotic drug candidates, but further studies are required, particularly to develop tissue specific therapeutics.
Collapse
Affiliation(s)
- Sophie L Harding-Fox
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health and Social Care, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, Essex CM1 1SQ, UK.
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health and Social Care, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, Essex CM1 1SQ, UK
| |
Collapse
|
5
|
Tseng PL, Sun W, Salem A, Alaklobie M, Macfarlane SC, Gad AK, Collins MO, Erdmann KS. Mechanical control of the alternative splicing factor PTBP1 regulates extracellular matrix stiffness induced proliferation and cell spreading. iScience 2025; 28:112273. [PMID: 40241749 PMCID: PMC12002664 DOI: 10.1016/j.isci.2025.112273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/23/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Cells sense mechanical cues and convert them into biochemical responses to regulate biological processes such as embryonic development, aging, cellular homeostasis, and disease progression. In this study, we introduce a large-scale, systematic approach to identify proteins with mechanosensitive nuclear localization, highlighting their potential roles in mechanotransduction. Among the proteins identified, we focus here on the splicing factor PTBP1. We demonstrate that its nuclear abundance is regulated by mechanical cues such as cell density, size, and extracellular matrix (ECM) stiffness and that PTBP1 medicates the mechanosensitive alternative splicing of the endocytic adapter protein Numb. Furthermore, we show that PTBP1 and Numb alternative splicing is critical for ECM stiffness-induced epithelial cell spreading and proliferation as well as for mesenchymal stem cell differentiation into osteoblasts on a stiff matrix. Our results underscore the emerging role of alternative splicing in mechanotransduction and provide novel mechanistic insights into how matrix stiffness modulates cellular mechanoresponses.
Collapse
Affiliation(s)
- Pei-Li Tseng
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Weiwei Sun
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Ahmed Salem
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- Department of Biomedical Laboratory Science, Sheba University, Sheba, Libya
| | - Mubarak Alaklobie
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 67714, Saudi Arabia
| | - Sarah C. Macfarlane
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield S10 2TN, UK
| | - Annica K.B. Gad
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield S10 2TN, UK
- Department of Oncology-Pathology, Karolinska Institutet, Anna Steckséns gata 30A, 171 64 Solna, Sweden
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Mark O. Collins
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- biOMICS Mass Spectrometry Facility, University of Sheffield, Sheffield S10 2TN, UK
| | - Kai S. Erdmann
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
6
|
Frömmichen J, Bungert E, Ströble J, Gläser M, Gottwald C, Zeqiri K, Reinhard T, Lübke J, Schlunck G, Wiedenmann CJ. Effects of Verteporfin on Interstitial Fluid Flow-Induced Fibrotic Transdifferentiation of Human Tenon Fibroblasts. Invest Ophthalmol Vis Sci 2025; 66:17. [PMID: 40197780 PMCID: PMC11993124 DOI: 10.1167/iovs.66.4.17] [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: 10/17/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Postoperative scarring remains the major challenge in achieving long-term success after glaucoma filtration surgery. In a previous study, we showed that slow continuous fluid flow is sufficient to induce fibrotic responses in human tenon fibroblasts (HTFs) in two-dimensional (2D) and three-dimensional (3D) in vitro models. In the present study, we investigated the role of the mechanosensitive Yes-associated protein (YAP) and transcriptional coactivator (TAZ) signaling pathway in flow-induced fibrosis. Methods HTFs were exposed to continuous fluid flow for 48 or 72 hours in the presence or absence of the YAP/TAZ-transcriptional enhanced associated domain inhibitor verteporfin (VP). In a 2D model, the F-actin cytoskeleton, fibronectin 1 (FN1), YAP, and TAZ were visualized by confocal immunofluorescence microscopy. In a 3D model, mRNA was extracted, and the expression of fibrosis-associated genes was detected by quantitative PCR. Results HTFs exposed to slow fluid flow showed increased staining intensities for YAP/TAZ. Inhibition of YAP/TAZ by VP slightly reduced flow-induced fibrotic changes in the 2D model. The flow-induced increase in the expression of the extracellular matrix (ECM) genes COL1A1, CTGF, and FN1 was significantly inhibited by VP in the 3D model. Conclusions Slow interstitial fluid flow activates the YAP/TAZ pathway. VP exerts antifibrotic potential by reducing morphologic changes and suppressing the expression of ECM genes induced by flow. Therefore, YAP/TAZ inhibition may exhibit therapeutic potential after glaucoma filtration surgery by inhibiting fibrotic changes induced by mechanical stimuli.
Collapse
Affiliation(s)
- Janne Frömmichen
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Emma Bungert
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jeanne Ströble
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Moritz Gläser
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Gottwald
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kosovare Zeqiri
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Lübke
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
7
|
Lao Z, Chen X, Pan B, Fang B, Yang W, Qian Y. Pharmacological regulators of Hippo pathway: Advances and challenges of drug development. FASEB J 2025; 39:e70438. [PMID: 40100056 DOI: 10.1096/fj.202401895rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
The Hippo signaling pathway is crucial in regulating organ size, tumor progression, tissue regeneration, and bone homeostasis. Inactivation of the Hippo pathway results in the nuclear translocation and activation of YAP/TAZ. This activation not only promotes tumor progression but also enhances tissue regeneration, wound healing, and maintenance of bone stability Although its discovery occurred over two decades ago, developing effective inhibitors or activators for the Hippo pathway remains challenging. Recently, however, the pace of advancements in developing Hippo signaling-related agonists and antagonists has accelerated, with some drugs that target TEAD advancing to clinical trials and showing promise for treating related diseases. This review summarizes the progress in research on Hippo signaling-related agonists and inhibitors, offering an in-depth analysis of their regulatory mechanisms, pharmacological properties, and potential in vivo applications.
Collapse
Affiliation(s)
- Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Fang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Guo Z, Yao J, Zheng X, Cao J, Lv X, Gao Z, Guo S, Li H, Guan D, Li L, Qin D, Li D, Wang X, Tan M, Zhang J, Zhang Y, Wang B, Bu W, Li J, Zhao X, Meng F, Feng Y, Li L, Du J, Fan Y. Cavity oscillation drives pattern formation in early mammalian embryos. Cell Rep 2025; 44:115342. [PMID: 39985766 DOI: 10.1016/j.celrep.2025.115342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/02/2024] [Accepted: 01/31/2025] [Indexed: 02/24/2025] Open
Abstract
During the second cell fate in mouse embryos, the inner cell mass (ICM) segregates into the spatially distinct epiblast (EPI) and primitive endoderm (PrE) layers. The mechanism driving this pattern formation, however, remains unresolved. Here, we report that, concomitant with the segregation process of EPI/PrE precursors starting from mid-blastocyst, the blastocyst cavity begins to oscillate cyclically with rapid contraction yet slow expansion, triggering a phase transition in the ICM to a fluid-like state. This asymmetric oscillation of the blastocyst cavity facilitates EPI/PrE segregation by enhancing cell-cell contact fluctuations within the ICM and initiating convergent cell flows, which induce movement of these two cell types in opposite directions, wherein PrE precursors move toward the ICM-lumen interface, whereas EPI precursors move toward the trophectoderm. Last, we found that both PDGFRα expression and YAP nuclear accumulation in PrE precursors increase in response to blastocyst cavity oscillation. This study reveals the foundational role of physical oscillation in driving embryonic pattern formation during early mammalian embryonic development.
Collapse
Affiliation(s)
- Zheng Guo
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jie Yao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xu Zheng
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Jialing Cao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xinxin Lv
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zheng Gao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Shuyu Guo
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hangyu Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Dongshi Guan
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Dandan Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dong Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoxiao Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Min Tan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jing Zhang
- Laboratory Animal Research Center, Tsinghua University, Beijing 100084, China
| | - Yanli Zhang
- Imaging Core Facility, Technology Center for Protein Science, Tsinghua University, Beijing 100084, China
| | - Bo Wang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xi'ning 810008, China
| | - Wanjuan Bu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Jianwen Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xinbin Zhao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Fanzhe Meng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yue Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Du
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
9
|
Melrose J. Glycosaminoglycans, Instructive Biomolecules That Regulate Cellular Activity and Synaptic Neuronal Control of Specific Tissue Functional Properties. Int J Mol Sci 2025; 26:2554. [PMID: 40141196 PMCID: PMC11942259 DOI: 10.3390/ijms26062554] [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: 01/08/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Glycosaminoglycans (GAGs) are a diverse family of ancient biomolecules that evolved over millennia as key components in the glycocalyx that surrounds all cells. GAGs have molecular recognition and cell instructive properties when attached to cell surface and extracellular matrix (ECM) proteoglycans (PGs), which act as effector molecules that regulate cellular behavior. The perception of mechanical cues which arise from perturbations in the ECM microenvironment allow the cell to undertake appropriate biosynthetic responses to maintain ECM composition and tissue function. ECM PGs substituted with GAGs provide structural support to weight-bearing tissues and an ability to withstand shear forces in some tissue contexts. This review outlines the structural complexity of GAGs and the diverse functional properties they convey to cellular and ECM PGs. PGs have important roles in cartilaginous weight-bearing tissues and fibrocartilages subject to tension and high shear forces and also have important roles in vascular and neural tissues. Specific PGs have roles in synaptic stabilization and convey specificity and plasticity in the regulation of neurophysiological responses in the CNS/PNS that control tissue function. A better understanding of GAG instructional roles over cellular behavior may be insightful for the development of GAG-based biotherapeutics designed to treat tissue dysfunction in disease processes and in novel tissue repair strategies following trauma. GAGs have a significant level of sophistication over the control of cellular behavior in many tissue contexts, which needs to be fully deciphered in order to achieve a useful therapeutic product. GAG biotherapeutics offers exciting opportunities in the modern glycomics arena.
Collapse
Affiliation(s)
- James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia;
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School, Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
10
|
Fang X, Jia H, Pan S, Liu Q, Wang Q, Feng Y, Ding W, Luo T. Matrix Stiffness Regulates Interleukin-10 Secretion in Human Microglia (HMC3) via YAP-Mediated Mechanotransduction. Cell Biochem Funct 2025; 43:e70061. [PMID: 40011226 DOI: 10.1002/cbf.70061] [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: 09/10/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Microglia, as resident immune cells in the brain, adhere to the extracellular matrix and typically exhibit anti-inflammatory polarization under normal physiological conditions. Despite their pivotal roles, the regulatory effects of extracellular matrix properties on microglial function and the associated molecular mechanisms remain inadequately understood. Here, we elucidate how matrix stiffness modulates interleukin-10 (IL-10) secretion in human microglia (HMC3) via yes-associated protein (YAP)-mediated mechanotransduction. Using soft collagen Ⅰ-coated hydrogels, we observed a substantial reduction in IL-10 secretion, accompanied by a decrease in the expression and nuclear localization of YAP compared to cells adhered to glass substrates. With increasing hydrogel substrate stiffness, the expression and nuclear localization of YAP were enhanced, leading to an elevated secretion of IL-10. Subsequently, to further investigate the relationship between YAP and IL-10, we performed YAP depletion experiments, which revealed that nuclear exclusion of YAP suppressed IL-10 secretion. Interestingly, overexpression of YAP in microglia did not markedly affect IL-10 levels. We seeded YAP-knockdown microglia onto hydrogels of varying stiffness, and no significant differences were observed in IL-10 secretion. Our findings suggested that cytoskeletal polymerization was crucial for the regulation of IL-10 secretion mediated by YAP. Given the crucial role of IL-10 in the tumor microenvironment, we further found shYAP-microglia attenuated the pro-proliferative effect of microglia on gliomas. Besides, when YAP was silenced, actin of human microglia decreased, and their contractility was weakened. In summary, this study identifies YAP as a pivotal molecule in controlling cytokine secretion and sensing matrix stiffness in microglia. These insights offer potential therapeutic avenues for glioma treatment by targeting YAP-mediated pathways in microglial cells.
Collapse
Affiliation(s)
- Xue Fang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Haiying Jia
- Chinese People's Liberation Army Strategic Support Force Special Medical Center, Beijing, China
| | - Shaoshan Pan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Liu
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Qian Wang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Ye Feng
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Weiping Ding
- School of Information Science and Technology, University of Science and Technology of China, Hefei, China
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
- The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| |
Collapse
|
11
|
Chalifoux M, Avdeeva M, Posfai E. Geometric, cell cycle and maternal-to-zygotic transition-associated YAP dynamics during preimplantation embryo development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640568. [PMID: 40060487 PMCID: PMC11888467 DOI: 10.1101/2025.02.27.640568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
During the first cell fate decision in mammalian embryos the inner cell mass cells, which will give rise to the embryo proper and other extraembryonic tissues, segregate from the trophectoderm cells, the precursors of the placenta. Cell fate segregation proceeds in a gradual manner encompassing two rounds of cell division, as well as cell positional and morphological changes. While it is known that the activity of the Hippo signaling pathway and the subcellular localization of its downstream effector YAP dictate lineage specific gene expression, the response of YAP to these dynamic cellular changes remains incompletely understood. Here we address these questions by quantitative live imaging of endogenously tagged YAP while simultaneously monitoring geometric cellular features and cell cycle progression throughout cell fate segregation. We apply a probabilistic model to our dynamic data, providing a quantitative characterization of the mutual effects of YAP and cellular relative exposed area, which has previously been shown to correlate with subcellular YAP localization in fixed samples. Additionally, we study how nuclear YAP levels are influenced by other factors, such as the decreasing pool of maternally provided YAP that is partitioned to daughter cells through cleavage divisions, cell cycle-associated nuclear volume changes, and a delay after divisions in adjusting YAP levels to new cell positions. Interestingly, we find that establishing low nuclear YAP levels required for the inner cell mass fate is largely achieved by passive cell cycle-associated mechanisms. Moreover, contrary to expectations, we find that mechanical perturbations that result in cell shape changes do not influence YAP localization in the embryo. Together our work identifies how various inputs are integrated over a dynamic developmental time course to shape the levels of a key molecular determinant of the first cell fate choice.
Collapse
Affiliation(s)
- Madeleine Chalifoux
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
| | - Maria Avdeeva
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
12
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
13
|
Nakagawa R, Beardsley A, Durney S, Hayward MK, Subramanyam V, Meyer NP, Wismer H, Goodarzi H, Weaver VM, Van de Mark D, Goga A. Tumor Cell Spatial Organization Directs EGFR/RAS/RAF Pathway Primary Therapy Resistance through YAP Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.26.615226. [PMID: 39386679 PMCID: PMC11463411 DOI: 10.1101/2024.09.26.615226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Non-small cell lung cancers (NSCLC) harboring common mutations in EGFR and KRAS characteristically respond transiently to targeted therapies against those mutations, but invariably, tumors recur and progress. Resistance often emerges through mutations in the therapeutic target or activation of alternative signaling pathways. Mechanisms of acute tumor cell resistance to initial EGFR (EGFRi) or KRASG12C (G12Ci) pathway inhibition remain poorly understood. Our study reveals that acute response to EGFR/RAS/RAF-pathway inhibition is spatial and culture context specific. In vivo, EGFR mutant tumor xenografts shrink by > 90% following acute EGFRi therapy, and residual tumor cells are associated with dense stroma and have increased nuclear YAP. Interestingly, in vitro EGFRi induced cell cycle arrest in NSCLC cells grown in monolayer, while 3D spheroids preferentially die upon inhibitor treatment. We find differential YAP nuclear localization and activity, driven by the distinct culture conditions, as a common resistance mechanism for selective EGFR/KRAS/BRAF pathway therapies. Forced expression of the YAPS127A mutant partially protects cells from EGFR-mediated cell death in spheroid culture. These studies identify YAP activation in monolayer culture as a non-genetic mechanism of acute EGFR/KRAS/BRAF therapy resistance, highlighting that monolayer vs spheroid cell culture systems can model distinct stages of patient cancer progression.
Collapse
Affiliation(s)
- Rachel Nakagawa
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrew Beardsley
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Sophia Durney
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Mary-Kate Hayward
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
| | - Vishvak Subramanyam
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Nathaniel P. Meyer
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Harrison Wismer
- Biological Imaging Development CoLab, UCSF, San Francisco, CA, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Daniel Van de Mark
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrei Goga
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
14
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
15
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [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: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
16
|
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
|
17
|
Yu Lin MO, Sampath D, Bosykh DA, Wang C, Wang X, Subramaniam T, Han W, Hong W, Chakraborty S. YAP/TAZ Drive Agrin-Matrix Metalloproteinase 12-Mediated Diabetic Skin Wound Healing. J Invest Dermatol 2025; 145:155-170.e2. [PMID: 38810954 DOI: 10.1016/j.jid.2024.05.005] [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: 09/05/2023] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024]
Abstract
Macroscopic loss of extracellular matrix can lead to chronic defects in skin wound healing, but supplementation of extracellular matrix holds promise for facilitating wound closure, particularly in diabetic wound healing. We recently showed that the extracellular matrix proteoglycan agrin accelerates cutaneous wound healing by improving mechanoperception of migrating keratinocytes and allowing them to respond to mechanical stresses through matrix metalloproteinase 12 (MMP12). RNA-sequencing analysis revealed that in addition to a disorganized extracellular matrix, agrin-depleted skin cells have impaired YAP/TAZ transcriptional outcomes, leading us to hypothesize that YAP/TAZ, as central mechanosensors, drive the functionality of agrin-MMP12 signaling during cutaneous wound repair. In this study, we demonstrate that agrin activates YAP/TAZ during migration of keratinocytes after wounding in vitro and in vivo. Mechanistically, YAP/TAZ sustain agrin and MMP12 protein expression during migration after wounding through positive feedback. YAP/TAZ silencing abolishes agrin-MMP12-mediated force recognition and geometrical constraints. Importantly, soluble agrin therapy accelerates wound closure in diabetic mouse models by engaging MMP12-YAP. Because patients with diabetic foot ulcers and impaired wound healing have reduced expression of agrin-MMP12 that correlates with YAP/TAZ inactivation, we propose that timely activation of YAP/TAZ by soluble agrin therapy can accentuate mechanobiological microenvironments for efficient wound healing, under normal and diabetic conditions.
Collapse
Affiliation(s)
| | | | - Dmitriy A Bosykh
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Chengchun Wang
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Xiaomeng Wang
- Institute of Molecular and Cell Biology, Singapore, Singapore; Centre for Vision Research, Duke-NUS Medical School, Singapore, Singapore
| | - Tavintharan Subramaniam
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore; Division of Endocrinology, Department of Medicine, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Weiping Han
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Singapore, Singapore.
| | - Sayan Chakraborty
- Institute of Molecular and Cell Biology, Singapore, Singapore; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Program of Developmental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| |
Collapse
|
18
|
Lipari S, Sacco P, Cok M, Scognamiglio F, Romano M, Brun F, Giulianini PG, Marsich E, Aachmann FL, Donati I. Hydrogel Elastic Energy: A Stressor Triggering an Adaptive Stress-Mediated Cell Response. Adv Healthc Mater 2025; 14:e2402400. [PMID: 39535422 PMCID: PMC11730662 DOI: 10.1002/adhm.202402400] [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: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The crosstalk between the cells and the extracellular matrix (ECM) is bidirectional and consists of a pushing/pulling stretch exerted by the cells and a mechanical resistance counteracted by the surrounding microenvironment. It is widely recognized that the stiffness of the ECM, its viscoelasticity, and its overall deformation are the most important traits influencing the response of the cells. Here these three parameters are combined into a concept of elastic energy, which in biological terms represents the mechanical feedback that cells perceive when the ECM is deformed. It is shown that elastic energy is a stress factor that influences the response of cells in three-dimensional (3D) cultures. Strikingly, the higher the elastic energy of the matrix and thus the mechanical feedback, the higher the stress state of the cells, which correlates with the formation of G3BP-mediated stress granules. This condition is associated with an increase in alkaline phosphatase (ALP) activity but a decrease in gene expression and is mediated by the nuclear translocation of Yes-associated protein (YAP). This work supports the importance of considering the elastic energy as mechano-controller in regulating cellular stress state in 3D cultures.
Collapse
Affiliation(s)
- Sara Lipari
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Pasquale Sacco
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Michela Cok
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | | | - Maurizio Romano
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| | - Francesco Brun
- Department of Engineering and ArchitectureUniversity of TriesteVia A. Valerio 6/1Trieste34127Italy
| | | | - Eleonora Marsich
- Department of MedicineSurgery and Health SciencesUniversity of TriestePiazza dell'Ospitale 1Trieste34129Italy
| | - Finn L. Aachmann
- Department of Biotechnology and Food ScienceNorwegian Biopolymer Laboratory (NOBIPOL)NTNU Norwegian University of Science and TechnologySem Sælands vei 6/8Trondheim7491Norway
| | - Ivan Donati
- Department of Life SciencesUniversity of TriesteVia L. Giorgieri 5Trieste34127Italy
| |
Collapse
|
19
|
Vaitinadapoulé H, Ben Moussa O, Maurin C, Aouimeur I, Perrache C, Thomas J, Forestier P, Crouzet E, He Z, Gain P, Thuret G, Mascarelli F. Expression of Yes-associated protein in endothelial cells of human corneas before and after storage in organ culture. Sci Rep 2024; 14:31073. [PMID: 39730686 DOI: 10.1038/s41598-024-82269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/04/2024] [Indexed: 12/29/2024] Open
Abstract
The cornea, the anterior meniscus-shaped transparent and refractive structure of the eyeball, is the first mechanical barrier of the eye. Its functionality heavily relies on the health of its endothelium, its most posterior layer. The treatment of corneal endothelial cells (CECs) deficiency is allogeneic corneal graft using stored donor corneas. One of the main goals of eye banks is to maintain endothelial cell density (ECD) and endothelial barrier function, critical parameters influencing transplantation outcomes. Unlike in vivo, the stored cornea is not subjected to physiological mechanical stimuli, such as the hydrokinetic pressure of the aqueous humor and intraocular pressure (IOP). YAP (Yes-Associated Protein), a pivotal transcriptional coactivator, is recognized for its ability to sense diverse biomechanical cues and transduce them into specific biological signals, varying for each cell type and mechanical forces. The biomechanical cues that might regulate YAP in human corneal endothelium remain unidentified. Therefore, we investigated the expression and subcellular localization of YAP in the endothelium of corneas stored in organ culture (OC). Our findings demonstrated that CEC morphology, ECD and cell-cell interactions are distinctly and differentially associated with modifications in the expression, subcellular localization and phosphorylation of YAP. Notably, this phosphorylation occurs in the basal region of the primary cilium, which may play central cellular roles in sensing mechanical stimuli. The sustained recruitment of YAP in cellular junctions, nucleus, and cilium under long-term OC conditions strongly indicates its specific role in maintaining CEC homeostasis. Understanding these biophysical influences could aid in identifying molecules that promote homeostasis and enhance the functionality of CECs.
Collapse
Affiliation(s)
- Hanielle Vaitinadapoulé
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Olfa Ben Moussa
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Corantin Maurin
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Inès Aouimeur
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Chantal Perrache
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Justin Thomas
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Pierre Forestier
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Emmanuel Crouzet
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Zhiguo He
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
| | - Philippe Gain
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
- Ophthalmology Department, University Hospital, Avenue Albert Raimond, 42055, Saint-Etienne Cedex 02, France
| | - Gilles Thuret
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France.
- Ophthalmology Department, University Hospital, Avenue Albert Raimond, 42055, Saint-Etienne Cedex 02, France.
| | - Frédéric Mascarelli
- Laboratory of Biology, Engineering, and Imaging for Ophthalmology, BiiO, Faculty of Medicine, University of Jean Monnet, 10 rue de la Marandière, 42270, Saint-Priest en Jarez, France
- Centre de Recherche des Cordeliers, UMR S1138, Université de Paris Descartes, Paris, France
| |
Collapse
|
20
|
Hu X, Liu Y, Tang B, Hu J, He H, Liu H, Li L, Hu S, Wang J. Comparative transcriptomic analysis revealed potential mechanisms regulating the hypertrophy of goose pectoral muscles. Poult Sci 2024; 103:104498. [PMID: 39504833 PMCID: PMC11577216 DOI: 10.1016/j.psj.2024.104498] [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: 08/09/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Pectoral muscle development is an important economic trait. According to the different essence, muscle development can be divided into 2 processes: embryonic muscle fiber generation and postnatal muscle fiber hypertrophy, and postnatal muscle fiber hypertrophy has a greater impact on muscle development than the number of muscle fibers formed during the embryonic phase in poultry. However, the underlying mechanisms regulating the hypertrophy of goose pectoral muscles have not been elucidated. Therefore, the purpose of the present study was to conduct transcriptome sequencing in pectoral muscles of both Landes (LD) and Sichuan White (SW) geese at 6, 10, and 30 weeks of age to reveal the molecular mechanisms regulating pectoral muscle hypertrophy through intra-breed and inter-breed bioinformatics analyses. Phenotypically, the pectoral muscle weight/index of LD and SW geese increased from 6 to 30 weeks of age, and except for the pectoral muscle index at 10 weeks of age (P = 0.962), at the same age, the pectoral muscle weight/index of LD geese were significantly higher than that of SW geese (P < 0.05). In transcriptional regulation, intra-breed bioinformatics analysis identified 3331 genes whose expression levels were opposite to the trend of pectoral muscle hypertrophy both in LD and SW geese, and the 3331 genes were mainly enriched into abundant KEGG pathways related to lipid metabolism, proliferation/apoptosis, and immune response. Moreover, 23 genes (including SLC2A10, TNFRSF1A, PRKAA1, SLC27A4, ITGB2, THY1, RHOA, MYL10, ACTB, PRKCB, PIK3R2, RAC2, DMD, LATS2, YAP1, WWTR1, SMAD7, CTGF, FGF1, AXIN2, GLI2, ID2, and CCND2) who were enriched in 6 crosstalk pathways named viral myocarditis, insulin resistance, sphingolipid signaling pathway, hippo signaling pathway, chemokine signaling pathway, and leukocyte transendothelial migration were identified as the key candidate genes regulating the hypertrophy of goose pectoral muscles. In inter-breed bioinformatics analysis, abundant different expression genes (DEGs) related to lipid metabolism, immune response, and proliferation/apoptosis were identified between LD and SW geese too, and compared with SW geese, the expression level of MYL10 in LD geese was lower, while the expression levels of GLI2/CTGF/SMAD7 in LD geese were higher. These results suggested that the hypertrophy of goose pectoral muscles might be achieved through more lipid deposition and less leukocyte infiltration to promote the proliferation of cells within the muscles, and the low expression of MYL10 and high expressions of GLI2/CTGF/SMAD7 might the keys to induce the pectoral muscle hypertrophy of LD geese from 6 to 30 weeks of age over that of SW geese. All data the present study obtained will provide new insights into the molecular mechanisms regulating the hypertrophy of goose pectoral muscles.
Collapse
Affiliation(s)
- Xinyue Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Yali Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Bincheng Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiwei Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hua He
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hehe Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Liang Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Shenqiang Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiwen Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.
| |
Collapse
|
21
|
Sharaf A, Frimat JP, Accardo A. Mechanical confinement matters: Unveiling the effect of two-photon polymerized 2.5D and 3D microarchitectures on neuronal YAP expression and neurite outgrowth. Mater Today Bio 2024; 29:101325. [PMID: 39569166 PMCID: PMC11576396 DOI: 10.1016/j.mtbio.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
The effect of mechanical cues on cellular behaviour has been reported in multiple studies so far, and a specific aspect of interest is the role of mechanotransductive proteins in neuronal development. Among these, yes-associated protein (YAP) is responsible for multiple functions in neuronal development such as neuronal progenitor cells migration and differentiation while myocardin-related transcription factor A (MRTFA) facilitates neurite outgrowth and axonal pathfinding. Both proteins have indirectly intertwined fates via their signalling pathways. There is little literature investigating the roles of YAP and MRTFA in vitro concerning neurite outgrowth in mechanically confined microenvironments. Moreover, our understanding of their relationship in immature neurons cultured within engineered confined microenvironments is still lacking. In this study, we fabricated, via two-photon polymerization (2PP), 2.5D microgrooves and 3D polymeric microchannels, with a diameter range from 5 to 30 μm. We cultured SH-SY5Y cells and differentiated them into immature neuron-like cells on both 2.5D and 3D microstructures to investigate the effect of mechanical confinement on cell morphology and protein expression. In 2.5D microgrooves, both YAP and MRTFA nuclear/cytoplasmic (N/C) ratios exhibited maxima in the 10 μm grooves indicating a strong relation with mechanical-stress-inducing confinement. In 3D microchannels, both proteins' N/C ratio exhibited minima in presence of 5 or 10 μm channels, a behaviour that was opposite to the ones observed in the 2.5D microgrooves and that indicates how the geometry and mechanical confinement of 3D microenvironments are unique compared to 2.5D ones due to focal adhesion, actin, and nuclear polarization. Further, especially in presence of 2.5D microgrooves, cells featured an inversely proportional relationship between YAP N/C ratio and the average neurite length. Finally, we also cultured human induced pluripotent stem cells (hiPSCs) and differentiated them into cortical neurons on the microstructures for up to 2 weeks. Interestingly, YAP and MRTFA N/C ratios also showed a maximum around the 10 μm 2.5D microgrooves, indicating the physiological relevance of our study. Our results elucidate the possible differences induced by 2.5D and 3D confining microenvironments in neuronal development and paves the way for understanding the intricate interplay between mechanotransductive proteins and their effect on neural cell fate within engineered cell microenvironments.
Collapse
Affiliation(s)
- Ahmed Sharaf
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, the Netherlands
| | - Jean-Philippe Frimat
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Faculty of Mechanical Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, the Netherlands
| |
Collapse
|
22
|
Wang K, Lu J, Song C, Qiao M, Li Y, Chang M, Bao H, Qiu Y, Qian B. Extracellular Vesicles Derived from Ligament Tissue Transport Interleukin-17A to Mediate Ligament-To-Bone Crosstalk in Ankylosing Spondylitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406876. [PMID: 39308181 PMCID: PMC11633500 DOI: 10.1002/advs.202406876] [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: 06/20/2024] [Revised: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Pathological new bone formation is a critical feature of the progression of ankylosing spondylitis (AS), and spine ankylosis is a distinctive feature of this condition. Ligaments are the primary regions of pathological new bone formation in AS. Here, it is demonstrated that ligament tissue-derived extracellular vesicles (EVs) and their interleukin-17A (IL-17A) cargo mediate the communication between the tissue and other cells. The investigation revealed that IL-17A in EVs can activate the JAK-STAT3 pathway, thereby stimulating the expression of MMP14 in AS ligament. Overexpression of MMP14 can lead to changes in the cytoskeleton and mechanical signaling of mesenchymal stem cells and other cells. These alterations in cellular cytoskeleton and mechanical signaling at ligament sites in patients with AS or in stem cells treated with EVs can result in pathological new bone formation. Finally, inhibiting IL-17A activity and EV endocytosis effectively controlled inflammation and pathological new bone formation. Overall, these data suggest that ligament-derived EVs and the enclosed IL-17A have a potential role in driving pathological new bone formation in AS, and targeting EVs may therefore emerge as a novel approach to delaying ectopic ossification in AS.
Collapse
Affiliation(s)
- Kaiyang Wang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Jingshun Lu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Chenyu Song
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Mu Qiao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yao Li
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Menghan Chang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Hongda Bao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yong Qiu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Bang‐Ping Qian
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| |
Collapse
|
23
|
Wang TC, Abolghasemzade S, McKee BP, Singh I, Pendyala K, Mohajeri M, Patel H, Shaji A, Kersey AL, Harsh K, Kaur S, Dollahon CR, Chukkapalli S, Lele PP, Conway DE, Gaharwar AK, Dickinson RB, Lele TP. Matrix stiffness drives drop like nuclear deformation and lamin A/C tension-dependent YAP nuclear localization. Nat Commun 2024; 15:10151. [PMID: 39578439 PMCID: PMC11584751 DOI: 10.1038/s41467-024-54577-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Extracellular matrix (ECM) stiffness influences cancer cell fate by altering gene expression. Previous studies suggest that stiffness-induced nuclear deformation may regulate gene expression through YAP nuclear localization. We investigated the role of the nuclear lamina in this process. We show that the nuclear lamina exhibits mechanical threshold behavior: once unwrinkled, the nuclear lamina is inextensible. A computational model predicts that the unwrinkled lamina is under tension, which is confirmed using a lamin tension sensor. Laminar unwrinkling is caused by nuclear flattening during cell spreading on stiff ECM. Knockdown of lamin A/C eliminates nuclear surface tension and decreases nuclear YAP localization. These findings show that nuclear deformation in cells conforms to the nuclear drop model and reveal a role for lamin A/C tension in controlling YAP localization in cancer cells.
Collapse
Affiliation(s)
- Ting-Ching Wang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Samere Abolghasemzade
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Brendan P McKee
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ishita Singh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kavya Pendyala
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Mohammad Mohajeri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Aakansha Shaji
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Anna L Kersey
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Kajol Harsh
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Simran Kaur
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Christina R Dollahon
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Pushkar P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Tanmay P Lele
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Translational Medical Sciences, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
24
|
Rolfe RA, Bastürkmen ET, Sliney L, Hayden G, Dunne N, Buckley N, McCarthy H, Szczesny SE, Murphy P. Embryo movement is required for limb tendon maturation. Front Cell Dev Biol 2024; 12:1466872. [PMID: 39574785 PMCID: PMC11579356 DOI: 10.3389/fcell.2024.1466872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Following early cell specification and tenocyte differentiation at the sites of future tendons, very little is known about how tendon maturation into robust load-bearing tissue is regulated. Between embryonic day (E)16 and E18 in the chick, there is a rapid change in mechanical properties which is dependent on normal embryo movement. However, the tissue, cellular and molecular changes that contribute to this transition are not well defined. Methods Here we profiled aspects of late tendon development (collagen fibre alignment, cell organisation and Yap pathway activity), describing changes that coincide with tissue maturation. We compared effects of rigid (constant static loading) and flaccid (no loading) immobilisation to gain insight into developmental steps influenced by mechanical cues. Results We show that YAP signalling is active and responsive to movement in late tendon. Collagen fibre alignment increased over time and under static loading. Cells organise into end-to-end stacked columns with increased distance between adjacent columns, where collagen fibres are deposited; this organisation was lost following both types of immobilisation. Discussion We conclude that specific aspects of tendon maturation require controlled levels of dynamic muscle-generated stimulation. Such a developmental approach to understanding how tendons are constructed will inform future work to engineer improved tensile load-bearing tissues.
Collapse
Affiliation(s)
- Rebecca A. Rolfe
- Zoology, School of Natural Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Ebru Talak Bastürkmen
- Zoology, School of Natural Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Lauren Sliney
- Zoology, School of Natural Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Grace Hayden
- Zoology, School of Natural Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Nicholas Dunne
- School of Mechanical and Manufacturing Engineering, Dublin College University, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, University of Dublin, Dublin, Ireland
- Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin, Ireland
| | - Niamh Buckley
- School of Pharmacy, Queens University Belfast, Belfast, United Kingdom
| | - Helen McCarthy
- School of Pharmacy, Queens University Belfast, Belfast, United Kingdom
| | - Spencer E. Szczesny
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, United States
- Department of Orthopaedics and Rehabilitation, Pennsylvania State University, Hershey, PA, United States
| | - Paula Murphy
- Zoology, School of Natural Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Ferrick KR, Fan Y, Ratnayeke N, Teruel MN, Meyer T. Transient proliferation by reversible YAP and mitogen-control of the cyclin D1/p27 ratio. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617852. [PMID: 39416132 PMCID: PMC11482934 DOI: 10.1101/2024.10.11.617852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Hippo-YAP signaling orchestrates epithelial tissue repair and is therefore an attractive target in regenerative medicine. Yet it is unresolved how YAP integrates with mitogen signaling and contact inhibition to control the underlying transient proliferative response. Here we show that reduced contact inhibition, increased mitogen signaling, and YAP-TEAD activation converge on increasing the nuclear cyclin D1/p27 protein ratio during G1 phase, towards a threshold ratio that dictates whether individual cells enter or exit the cell cycle. YAP increases this ratio indirectly, in concert with mitogen signaling, by increasing EGFR and other receptors that signal primarily through ERK. After a delay, contact inhibition suppresses YAP activity which gradually downregulates mitogen signaling and the cyclin D1/p27 ratio. Increasing YAP activity by ablating the suppressor Merlin/NF2 reveals a balancing mechanism in which YAP suppression and contact inhibition of proliferation can be recovered but only at higher local cell density. Thus, critical for tissue repair, robust proliferation responses result from the YAP-induced and receptor-mediated prolonged increase in the cyclin D1/p27 ratio, which is only reversed by delayed suppression of receptor signaling after contact inhibition of YAP.
Collapse
Affiliation(s)
- Katherine R. Ferrick
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
- Department of Chemical and Systems Biology, Stanford Medicine, Stanford, CA, USA
| | - Yilin Fan
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
- Department of Chemical and Systems Biology, Stanford Medicine, Stanford, CA, USA
- Current: Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Nalin Ratnayeke
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
- Department of Chemical and Systems Biology, Stanford Medicine, Stanford, CA, USA
- Current: Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary N. Teruel
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Tobias Meyer
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
- Department of Chemical and Systems Biology, Stanford Medicine, Stanford, CA, USA
- Lead contact
| |
Collapse
|
26
|
Liao J, Li X, Yang H, He W, Wang B, Liu S, Fan Y. Construction of a Curcumin‐Loaded PLLA/PCL Micro‐Nano Conjugated Fibrous Membrane to Synergistically Prevent Postoperative Adhesion From Multiple Perspectives. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202407983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Indexed: 02/02/2025]
Abstract
AbstractPostoperative adhesion (POA) has emerged as a prevalent clinical challenge in soft tissue repair, emphasizing the critical need for preventive measures. However, the complex POA development process makes POA prevention from a single aspect insufficient. Hence, a curcumin‐loaded poly‐L‐lactic acid‐poly (caprolactone) micro‐nano conjugated fibrous membrane (PAPC MCFM (cur)) is engineered to synergistically prevent POA from multiple perspectives, in which poly (caprolactone) (PCL) nanofibers (118 ± 12 nm) with low orientation traverse the oriented poly‐L‐lactic acid (PLLA) microfibers (2.0 ± 0.3 µm). The PAPC MCFM not only significantly improves the mechanical properties of the anisotropic fibrous membrane (AIFM) that the modulus of elasticity and the tensile strength in the direction vertical to microfiber orientation increase by 4.5 and 13.0 times, respectively, but also can further enhance the “contact guidance effect” of AIFM, i.e., hindering fibroblast adhesion, proliferation, and differentiation to myofibroblast through inhibiting integrin β1 activation, vinculin expression and focal adhesion (FA) formation, and the nuclear localization activation of yes‐associated protein (YAP). Except for these effects, PAPC MCFM loading with 2.5 mg mL−1 curcumin can further prevent POA by delivering anti‐inflammatory, antioxidant, and antibacterial properties, and by suppressing fibrosis through decreased transforming growth factor‐β1(TGF‐β1) expression, showing effective POA prevention in rat abdominal cavity and rabbit dura mater models.
Collapse
Affiliation(s)
- Jie Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
- Department of Biomedical Materials Science College of Biomedical Engineering Third Military Medical University Chongqing 400038 China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
| | - Huiqi Yang
- Department of Hernia and Abdominal Wall Surgery Beijing Chao‐Yang Hospital Beijing 100043 China
| | - Wei He
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
| | - Bingbing Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
| | - Shuyu Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Center for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100083 China
| |
Collapse
|
27
|
Chen H, Cui H, Liu W, Li BW, Tian Z, Zhao YY, Yu GT. Manganese drives ferroptosis of cancer cells via YAP/TAZ phase separation activated ACSL4 in OSCC. Oral Dis 2024; 30:4898-4908. [PMID: 38462885 DOI: 10.1111/odi.14925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE Ferroptosis has been defined as a novel form of regulated cell death characterized by iron-dependent lipid peroxidation. Manganese has been used to induce ferroptosis in cancer cells recently. This study aims to investigate whether manganese can induce ferroptosis in oral squamous cell carcinoma (OSCC) and the underlying biological mechanisms. MATERIALS AND METHODS Cancer cells with or without manganese treatment were analyzed by RNA-sequencing to identify ferroptosis-related genes. Next, the activation of YAP/TAZ/ACSL4-ferroptosis signaling pathway was detected. Bioinformatic analysis and immunofluorescence assay were used to explore the phase separation of YAP/TAZ. Finally, specimens of OSCC patients were applied to analyze the clinical significance of YAP/TAZ/ACSL4. RESULTS RNA-sequencing analysis showed the ferroptosis-related genes and YAP/TAZ were upregulated after manganese treatment. The results of immunofluorescence, ELISA, western blotting, etc. further confirmed that manganese-induced ferroptosis depends on YAP/TAZ/ACSL4 signaling pathway. Moreover, the activation of ACSL4 was achieved by YAP/TAZ phase separation. The survival analysis in OSCC specimen suggested that the higher level of YAP/TAZ-ACSL4 axis expression indicates longer survival. CONCLUSIONS Manganese induces YAP/TAZ phase separation and subsequent ACSL4 activation via YAP/TAZ nuclear translocation, which facilitates ferroptosis of OSCC. Then YAP/TAZ-ACSL4 axis can be used as a potential prognostic predictor of OSCC patients.
Collapse
Affiliation(s)
- Hao Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo-Wen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yu-Yue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Guang-Tao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Wu LW, Jang SJ, Shapiro C, Fazlollahi L, Wang TC, Ryeom SW, Moy RH. Diffuse Gastric Cancer: A Comprehensive Review of Molecular Features and Emerging Therapeutics. Target Oncol 2024; 19:845-865. [PMID: 39271577 PMCID: PMC11557641 DOI: 10.1007/s11523-024-01097-2] [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] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
Diffuse-type gastric cancer (DGC) accounts for approximately one-third of gastric cancer diagnoses but is a more clinically aggressive disease with peritoneal metastases and inferior survival compared with intestinal-type gastric cancer (IGC). The understanding of the pathogenesis of DGC has been relatively limited until recently. Multiomic studies, particularly by The Cancer Genome Atlas, have better characterized gastric adenocarcinoma into molecular subtypes. DGC has unique molecular features, including alterations in CDH1, RHOA, and CLDN18-ARHGAP26 fusions. Preclinical models of DGC characterized by these molecular alterations have generated insight into mechanisms of pathogenesis and signaling pathway abnormalities. The currently approved therapies for treatment of gastric cancer generally provide less clinical benefit in patients with DGC. Based on recent phase II/III clinical trials, there is excitement surrounding Claudin 18.2-based and FGFR2b-directed therapies, which capitalize on unique biomarkers that are enriched in the DGC populations. There are numerous therapies targeting Claudin 18.2 and FGFR2b in various stages of preclinical and clinical development. Additionally, there have been preclinical advancements in exploiting unique therapeutic vulnerabilities in several models of DGC through targeting of the focal adhesion kinase (FAK) and Hippo pathways. These preclinical and clinical advancements represent a promising future for the treatment of DGC.
Collapse
Affiliation(s)
- Lawrence W Wu
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, 161 Fort Washington Avenue, Room 956, New York, NY, 10032, USA
| | - Sung Joo Jang
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Cameron Shapiro
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Ladan Fazlollahi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sandra W Ryeom
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Ryan H Moy
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, 161 Fort Washington Avenue, Room 956, New York, NY, 10032, USA.
| |
Collapse
|
29
|
Ni K, Che B, Gu R, Wang C, Pan Y, Li J, Liu L, Luo M, Deng L. Single-Cell Hypertrophy Promotes Contractile Function of Cultured Human Airway Smooth Muscle Cells via Piezo1 and YAP Auto-Regulation. Cells 2024; 13:1697. [PMID: 39451215 PMCID: PMC11505810 DOI: 10.3390/cells13201697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/30/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
Severe asthma is characterized by increased cell volume (hypertrophy) and enhanced contractile function (hyperresponsiveness) of the airway smooth muscle cells (ASMCs). The causative relationship and underlying regulatory mechanisms between them, however, have remained unclear. Here, we manipulated the single-cell volume of in vitro cultured human ASMCs to increase from 2.7 to 5.2 and 8.2 × 103 μm3 as a simulated ASMC hypertrophy by culturing the cells on micropatterned rectangular substrates with a width of 25 μm and length from 50 to 100 and 200 μm, respectively. We found that as the cell volume increased, ASMCs exhibited a pro-contractile function with increased mRNA expression of contractile proteins, increased cell stiffness and traction force, and enhanced response to contractile stimulation. We also uncovered a concomitant increase in membrane tension and Piezo1 mRNA expression with increasing cell volume. Perhaps more importantly, we found that the enhanced contractile function due to cell volume increase was largely attenuated when membrane tension and Piezo1 mRNA expression were downregulated, and an auto-regulatory loop between Piezo1 and YAP mRNA expression was also involved in perpetuating the contractile function. These findings, thus, provide convincing evidence of a direct link between hypertrophy and enhanced contractile function of ASMCs that was mediated via Piezo1 mRNA expression, which may be specifically targeted as a novel therapeutic strategy to treat pulmonary diseases associated with ASMC hypertrophy such as severe asthma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| |
Collapse
|
30
|
Lin CY, Sassi A, Wu Y, Seaman K, Tang W, Song X, Bienenstock R, Yokota H, Sun Y, Geng F, Wang L, You L. Mechanotransduction pathways regulating YAP nuclear translocation under Yoda1 and vibration in osteocytes. Bone 2024; 190:117283. [PMID: 39413946 DOI: 10.1016/j.bone.2024.117283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/22/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Yes-associated protein (YAP) is a mechanosensitive protein crucial for bone remodeling. Although research has identified pathways and components involved in YAP regulation, the precise mechanisms of its localization during Piezo1 activation or vibration remain unclear. Piezo1, a mechanosensitive ion channel, allows calcium ions to flow into cells upon activation. Recent studies suggest that combining Yoda1, a Piezo1 activator, with low-magnitude high-frequency (LMHF) vibration (>30 Hz, <1 g acceleration) enhances YAP nuclear translocation. This combination potentially improves the mechanoresponse and therapeutic efficacy of LMHF vibration in bone cells. This study aims to elucidate how Yoda1 and LMHF vibration regulate mechanosensitive structures and pathways, leading to YAP nuclear translocation in MLO-Y4 osteocyte like cells. We investigated the roles of the cytoskeleton and nuclear envelope (NE) in YAP activation under combined LMHF vibration and Yoda1 treatments. Additionally, we analyzed differentially expressed genes (DEGs) in MLO-Y4 cells subjected to these treatments and in Piezo1 knockdown MLO-Y4 cells exposed to vibration. Our findings indicated that increased YAP nuclear translocation with combined treatment may result from the distinct effects of Yoda1 and vibration. Specifically, Yoda1 influenced YAP through mechanisms involving actin and NE dynamics, while LMHF vibration may modulate YAP via the interleukin 6 (IL6)/signal transducer and activator of transcription 3 (STAT3) axis. This study provides new insights and potential therapeutic targets for osteocyte-related pathologies.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| | - Amel Sassi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| | - Yuning Wu
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON L8S 4L7, Canada.
| | - Kimberly Seaman
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| | - Wentian Tang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| | - Xin Song
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| | - Raphael Bienenstock
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| | - Fei Geng
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON L8S 4L7, Canada.
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Lidan You
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada; Department of Mechanical and Materials Engineering, Queen's University, Kingston, ON K7L3N6, Canada.
| |
Collapse
|
31
|
Nelson VL, Eadie AL, Perez L, Madhu M, Platt M, Mercer A, Pulinilkunnil T, Kienesberger P, Simpson JA, Brunt KR. Yap Is a Nutrient Sensor Sensitive to the Amino Acid L-Isoleucine and Regulates the Expression of Ctgf in Cardiomyocytes. Biomolecules 2024; 14:1299. [PMID: 39456232 PMCID: PMC11506509 DOI: 10.3390/biom14101299] [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: 09/10/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Myocardial infarction and reperfusion constitute a complex injury consisting of many distinct molecular stress patterns that influence cardiomyocyte survival and adaptation. Cell signalling, which is essential to cardiac development, also presents potential disease-modifying opportunities to recover and limit myocardial injury or maladaptive remodelling. Here, we hypothesized that Yap signalling could be sensitive to one or more molecular stress patterns associated with early acute ischemia. We found that Yap, and not Taz, expression patterns differed in a post-myocardial infarct compared to a peri-infarct region of rat hearts post-myocardial infarction, suggesting cell specificity that would be challenging to resolve for causation in vivo. Using H9c2 ventricular myotubes in vitro as a model, Yap levels were determined to be more sensitive to nutrient deprivation than other stress patterns typified by ischemia within the first hour of stress. Moreover, this is mediated by amino acid availability, predominantly L-isoleucine, and influences the expression of connective tissue growth factor (Ctgf)-a major determinant of myocardial adaptation after injury. These findings present novel opportunities for future therapeutic development and risk assessment for myocardial injury and adaptation.
Collapse
Affiliation(s)
- Victoria L. Nelson
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Ashley L. Eadie
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Lester Perez
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Malav Madhu
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| | - Mathew Platt
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Angella Mercer
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Thomas Pulinilkunnil
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Petra Kienesberger
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jeremy A. Simpson
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Keith R. Brunt
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Dalhousie Medicine New Brunswick, Faculty of Medicine, Dalhousie University, Saint John, NB E2L 4L5, Canada
- IMPART Investigator Team Canada, Saint John, NB E2L 4L5, Canada
| |
Collapse
|
32
|
Li P, Zhou H, Yan R, Yan W, Yang L, Li T, Qin X, Zhou Y, Li L, Bao J, Li J, Li S, Liu Y. Aligned fibrous scaffolds promote directional migration of breast cancer cells via caveolin-1/YAP-mediated mechanosensing. Mater Today Bio 2024; 28:101245. [PMID: 39318372 PMCID: PMC11421348 DOI: 10.1016/j.mtbio.2024.101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Tumorigenesis and metastasis are highly dependent on the interactions between the tumor and the surrounding microenvironment. In 3D matrix, the fibrous structure of the extracellular matrix (ECM) undergoes dynamic remodeling during tumor progression. In particular, during the late stage of tumor development, the fibers become more aggregated and oriented. However, it remains unclear how cancer cells respond to the organizational change of ECM fibers and exhibit distinct morphology and behavior. Here, we used electrospinning technology to fabricate biomimetic ECM with distinct fiber arrangements, which mimic the structural characteristics of normal or tumor tissues and found that aligned and oriented nanofibers induce cytoskeletal rearrangement to promote directed migration of cancer cells. Mechanistically, caveolin-1(Cav-1)-expressing cancer cells grown on aligned fibers exhibit increased integrin β1 internalization and actin polymerization, which promoted stress fiber formation, focal adhesion dynamics and YAP activity, thereby accelerating the directional cell migration. In general, the linear fibrous structure of the ECM provides convenient tracks on which tumor cells can invade and migrate. Moreover, histological data from both mice and patients with tumors indicates that tumor tissue exhibits a greater abundance of isotropic ECM fibers compared to normal tissue. And Cav-1 downregulation can suppress cancer cells muscle invasion through the inhibition of YAP-dependent mechanotransduction. Taken together, our findings revealed the Cav-1 is indispensable for the cellular response to topological change of ECM, and that the Cav-1/YAP axis is an attractive target for inhibiting cancer cell directional migration which induced by linearization of ECM fibers.
Collapse
Affiliation(s)
- Ping Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Hanying Zhou
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Ran Yan
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Wei Yan
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Lu Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Tingting Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiang Qin
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Yanyan Zhou
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Li Li
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Ji Bao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Junjie Li
- Breast Surgery Department, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Shun Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Yiyao Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, PR China
- Department of Urology, Deyang People's Hospital, Deyang, 618099, Sichuan, PR China
| |
Collapse
|
33
|
Choi YJ, Kim JH, Lee Y, Pyeon HJ, Yoo IK, Yoo JH. Anti-fibrogenic effect of umbilical cord-derived mesenchymal stem cell-conditioned media in human esophageal fibroblasts. Sci Rep 2024; 14:22233. [PMID: 39333200 PMCID: PMC11437107 DOI: 10.1038/s41598-024-73091-7] [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: 03/18/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Esophageal fibrosis can develop due to caustic or radiation injuries. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) are known to mitigate fibrosis in various organs. However, the potential effects of UC-MSCs on human esophageal fibrosis remain underexplored. This study investigated the anti-fibrogenic properties and mechanisms of UC-MSC-derived conditioned media (UC-MSC-CM) on human esophageal fibroblasts (HEFs). HEFs were treated with TGF-β1 and then cultured with UC-MSC-CM, and the expression levels of extracellular matrix (ECM) components, RhoA, myocardin related transcription factor A (MRTF-A), serum response factor (SRF), Yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ) were measured. UC-MSC-CM suppressed TGF-β1-induced fibrogenic activation in HEFs, as evidenced by the downregulation of ECM. UC-MSC-CM diminished the expression of RhoA, MRTF-A, and SRF triggered by TGF-β1. In TGF-β1-stimulated HEFs, UC-MSC-CM decreased the nuclear localization of MRTF-A and YAP. Additionally, UC-MSC-CM diminished the TGF-β1-induced nuclear expressions of YAP and TAZ, while concurrently enhancing the cytoplasmic presence of phosphorylated YAP. Furthermore, UC-MSC-CM reduced TGF-β1-induced phosphorylation of Smad2. These findings suggest that UC-MSC-CM may inhibit TGF-β1-induced fibrogenic activation in HEFs by targeting the Rho-mediated MRTF/SRF and YAP/TAZ pathways, as well as the Smad2 pathway. This indicates its potential as a stem cell therapy for esophageal fibrosis.
Collapse
Affiliation(s)
- Yoon Jeong Choi
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
- Institute of Basic Medical Sciences, CHA University School of Medicine, Seongnam, 13496, South Korea
| | - Jee Hyun Kim
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
| | - Yeonju Lee
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
| | - Hee Jang Pyeon
- R&D Division, CHA Biotech Co., Ltd, Seongnam, 13488, South Korea
| | - In Kyung Yoo
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea.
| | - Jun Hwan Yoo
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea.
- Institute of Basic Medical Sciences, CHA University School of Medicine, Seongnam, 13496, South Korea.
| |
Collapse
|
34
|
Müller L, Gutschner T, Hatzfeld M. A feedback loop between plakophilin 4 and YAP signaling regulates keratinocyte differentiation. iScience 2024; 27:110762. [PMID: 39286493 PMCID: PMC11402648 DOI: 10.1016/j.isci.2024.110762] [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: 03/27/2024] [Revised: 07/12/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
The Hippo signaling pathway is an important regulator of organ growth and differentiation, and its deregulation contributes to the development of cancer. The activity of its downstream targets YAP/TAZ depends on adherens junctions. Plakophilin 4 (PKP4) is a cell-type specific adherens junction protein expressed in the proliferating cells of the epidermis. Here, we show that PKP4 diminishes proliferation as well as differentiation. Depletion of PKP4 increased proliferation but at the same time induced premature epidermal differentiation. PKP4 interacted with several Hippo pathway components, including the transcriptional co-activators YAP/TAZ, and promoted nuclear YAP localization and target gene expression. In differentiated keratinocytes, PKP4 recruited LATS and YAP to cell junctions where YAP is transcriptionally inactive. YAP depletion, on the other hand, reduced PKP4 levels and keratinocyte adhesion indicative of a feedback mechanism controlling adhesion, proliferation, and differentiation by balancing YAP functions.
Collapse
Affiliation(s)
- Lisa Müller
- Institute of Molecular Medicine, Section for Pathochemistry, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120 Halle, Germany
- Institute of Molecular Medicine, Section for RNA Biology and Pathogenesis, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120 Halle, Germany
| | - Tony Gutschner
- Institute of Molecular Medicine, Section for RNA Biology and Pathogenesis, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120 Halle, Germany
| | - Mechthild Hatzfeld
- Institute of Molecular Medicine, Section for Pathochemistry, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Research Center, Kurt-Mothes-Str. 3A, 06120 Halle, Germany
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Jafarinia H, Khalilimeybodi A, Barrasa-Fano J, Fraley SI, Rangamani P, Carlier A. Insights gained from computational modeling of YAP/TAZ signaling for cellular mechanotransduction. NPJ Syst Biol Appl 2024; 10:90. [PMID: 39147782 PMCID: PMC11327324 DOI: 10.1038/s41540-024-00414-9] [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: 02/29/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.
Collapse
Affiliation(s)
- Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Stephanie I Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA.
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
38
|
Guo P, Li B, Dong W, Zhou H, Wang L, Su T, Carl C, Zheng Y, Hong Y, Deng H, Pan D. PI4P-mediated solid-like Merlin condensates orchestrate Hippo pathway regulation. Science 2024; 385:eadf4478. [PMID: 39116228 PMCID: PMC11956869 DOI: 10.1126/science.adf4478] [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: 10/25/2022] [Revised: 10/11/2023] [Accepted: 06/10/2024] [Indexed: 08/10/2024]
Abstract
Despite recent studies implicating liquid-like biomolecular condensates in diverse cellular processes, many biomolecular condensates exist in a solid-like state, and their function and regulation are less understood. We show that the tumor suppressor Merlin, an upstream regulator of the Hippo pathway, localizes to both cell junctions and medial apical cortex in Drosophila epithelia, with the latter forming solid-like condensates that activate Hippo signaling. Merlin condensation required phosphatidylinositol-4-phosphate (PI4P)-mediated plasma membrane targeting and was antagonistically controlled by Pez and cytoskeletal tension through plasma membrane PI4P regulation. The solid-like material properties of Merlin condensates are essential for physiological function and protect the condensates against external perturbations. Collectively, these findings uncover an essential role for solid-like condensates in normal physiology and reveal regulatory mechanisms for their formation and disassembly.
Collapse
Affiliation(s)
- Pengfei Guo
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Bing Li
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Wei Dong
- Department of Cell Biology, University of Pittsburgh; Pittsburgh, PA 15261, USA
| | - Huabin Zhou
- Department of Biophysics, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Li Wang
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Ting Su
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Christopher Carl
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh; Pittsburgh, PA 15261, USA
| | - Hua Deng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center; Dallas, TX 75390, USA
| |
Collapse
|
39
|
Miyamoto S. Untangling the role of RhoA in the heart: protective effect and mechanism. Cell Death Dis 2024; 15:579. [PMID: 39122698 PMCID: PMC11315981 DOI: 10.1038/s41419-024-06928-8] [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: 01/08/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
RhoA (ras homolog family member A) is a small G-protein that transduces intracellular signaling to regulate a broad range of cellular functions such as cell growth, proliferation, migration, and survival. RhoA serves as a proximal downstream effector of numerous G protein-coupled receptors (GPCRs) and is also responsive to various stresses in the heart. Upon its activation, RhoA engages multiple downstream signaling pathways. Rho-associated coiled-coil-containing protein kinase (ROCK) is the first discovered and best characterized effector or RhoA, playing a major role in cytoskeletal arrangement. Many other RhoA effectors have been identified, including myocardin-related transcription factor A (MRTF-A), Yes-associated Protein (YAP) and phospholipase Cε (PLCε) to regulate transcriptional and post-transcriptional processes. The role of RhoA signaling in the heart has been increasingly studied in last decades. It was initially suggested that RhoA signaling pathway is maladaptive in the heart, but more recent studies using cardiac-specific expression or deletion of RhoA have revealed that RhoA activation provides cardioprotection against stress through various mechanisms including the novel role of RhoA in mitochondrial quality control. This review summarizes recent advances in understanding the role of RhoA in the heart and its signaling pathways to prevent progression of heart disease.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
40
|
Wu J, Zhang Q, Yang Z, Xu Y, Liu X, Wang X, Peng J, Xiao J, Wang Y, Shang Z, Wang N, Li L, Zhang R, Zhang W, Zhang J, Zeng Z, Wu J. CD248-expressing cancer-associated fibroblasts induce non-small cell lung cancer metastasis via Hippo pathway-mediated extracellular matrix stiffness. J Cell Mol Med 2024; 28:e70025. [PMID: 39164826 PMCID: PMC11335579 DOI: 10.1111/jcmm.70025] [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: 11/02/2023] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
Metastasis is a crucial stage in tumour progression, and cancer-associated fibroblasts (CAFs) support metastasis through their participation in extracellular matrix (ECM) stiffness. CD248 is a possible biomarker for non-small cell lung cancer (NSCLC)-derived CAFs, but its role in mediating ECM stiffness to promote NSCLC metastasis is unknown. We investigated the significance of CD248+ CAFs in activating the Hippo axis and promoting connective tissue growth factor (CTGF) expression, which affects the stromal collagen I environment and improves ECM stiffness, thereby facilitating NSCLC metastasis. In this study, we found that higher levels of CD248 in CAFs induced the formation of collagen I, which in turn increased extracellular matrix stiffness, thereby enabling NSCLC cell infiltration and migration. Hippo axis activation by CD248+ CAFs induces CTGF expression, which facilitates the formation of the collagen I milieu in the stromal matrix. In a tumour lung metastasis model utilizing fibroblast-specific CD248 gene knockout mice, CD248 gene knockout mice showed a significantly reduced ability to develop tumour lung metastasis compared to that of WT mice. Our findings demonstrate that CD248+ CAFs activate the Hippo pathway, thereby inducing CTGF expression, which in turn facilitates the collagen I milieu of the stromal matrix, which promotes NSCLC metastasis.
Collapse
Affiliation(s)
- Jiangwei Wu
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Qiaoling Zhang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Zeyang Yang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Yujun Xu
- Department of BiologyGuizhou Medical UniversityGuiyangChina
| | - Xinlei Liu
- Guizhou Prenatal Diagnsis CenterThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xuanying Wang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Jiangying Peng
- Department of Pharmaceutical AnalysisZunyi Medical UniversityZunyiChina
| | - Jing Xiao
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Yun Wang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Zhenling Shang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Nianxue Wang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Long Li
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Department of Thoracic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui Zhang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Wei Zhang
- Department of Biochemistry and Molecular BiologyJilin Medical UniversityJilinChina
| | - Jian Zhang
- Department of Thoracic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zhu Zeng
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Jieheng Wu
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anChina
- Tumor Immunotherapy Technology Engineering Research CenterGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
41
|
Wang Y, Yu FX. Angiomotin family proteins in the Hippo signaling pathway. Bioessays 2024; 46:e2400076. [PMID: 38760875 DOI: 10.1002/bies.202400076] [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: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
The Motin family proteins (Motins) are a class of scaffolding proteins consisting of Angiomotin (AMOT), AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). Motins play a pivotal role in angiogenesis, tumorigenesis, and neurogenesis by modulating multiple cellular signaling pathways. Recent findings indicate that Motins are components of the Hippo pathway, a signaling cascade involved in development and cancer. This review discusses how Motins are integrated into the Hippo signaling network, as either upstream regulators or downstream effectors, to modulate cell proliferation and migration. The repression of YAP/TAZ by Motins contributes to growth inhibition, whereas subcellular localization of Motins and their interactions with actin fibers are critical in regulating cell migration. The net effect of Motins on cell proliferation and migration may contribute to their diverse biological functions.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Graham K, Lienau P, Bader B, Prechtl S, Naujoks J, Lesche R, Weiske J, Kuehnlenz J, Brzezinka K, Potze L, Zanconato F, Nicke B, Montebaur A, Bone W, Golfier S, Kaulfuss S, Kopitz C, Pilari S, Steuber H, Hayat S, Kamburov A, Steffen A, Schlicker A, Buchgraber P, Braeuer N, Font NA, Heinrich T, Kuhnke L, Nowak-Reppel K, Stresemann C, Steigemann P, Walter AO, Blotta S, Ocker M, Lakner A, von Nussbaum F, Mumberg D, Eis K, Piccolo S, Lange M. Discovery of YAP1/TAZ pathway inhibitors through phenotypic screening with potent anti-tumor activity via blockade of Rho-GTPase signaling. Cell Chem Biol 2024; 31:1247-1263.e16. [PMID: 38537632 DOI: 10.1016/j.chembiol.2024.02.013] [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: 09/18/2023] [Revised: 01/08/2024] [Accepted: 02/27/2024] [Indexed: 07/21/2024]
Abstract
This study describes the identification and target deconvolution of small molecule inhibitors of oncogenic Yes-associated protein (YAP1)/TAZ activity with potent anti-tumor activity in vivo. A high-throughput screen (HTS) of 3.8 million compounds was conducted using a cellular YAP1/TAZ reporter assay. Target deconvolution studies identified the geranylgeranyltransferase-I (GGTase-I) complex as the direct target of YAP1/TAZ pathway inhibitors. The small molecule inhibitors block the activation of Rho-GTPases, leading to subsequent inactivation of YAP1/TAZ and inhibition of cancer cell proliferation in vitro. Multi-parameter optimization resulted in BAY-593, an in vivo probe with favorable PK properties, which demonstrated anti-tumor activity and blockade of YAP1/TAZ signaling in vivo.
Collapse
Affiliation(s)
- Keith Graham
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Philip Lienau
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Benjamin Bader
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefan Prechtl
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Jan Naujoks
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Ralf Lesche
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Joerg Weiske
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Julia Kuehnlenz
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Krzysztof Brzezinka
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Lisette Potze
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Francesca Zanconato
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Barbara Nicke
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Anna Montebaur
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Wilhelm Bone
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sven Golfier
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefan Kaulfuss
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Charlotte Kopitz
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sabine Pilari
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Holger Steuber
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Sikander Hayat
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Atanas Kamburov
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Andreas Steffen
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Andreas Schlicker
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Philipp Buchgraber
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Nico Braeuer
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Nuria Aiguabella Font
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Tobias Heinrich
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Lara Kuhnke
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Katrin Nowak-Reppel
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Carlo Stresemann
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Patrick Steigemann
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Annette O Walter
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Simona Blotta
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Matthias Ocker
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Ashley Lakner
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Franz von Nussbaum
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany
| | - Dominik Mumberg
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Knut Eis
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy; IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Martin Lange
- Bayer AG, Pharmaceuticals, Research & Development, Muellerstr. 178, 13353 Berlin, Germany; Nuvisan ICB GmbH, Muellerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
43
|
Marvin JC, Liu EJ, Chen HH, Shiovitz DA, Andarawis-Puri N. Proteins Derived From MRL/MpJ Tendon Provisional Extracellular Matrix and Secretome Promote Pro-Regenerative Tenocyte Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602500. [PMID: 39026846 PMCID: PMC11257490 DOI: 10.1101/2024.07.08.602500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tendinopathies are prevalent musculoskeletal conditions that have no effective therapies to attenuate scar formation. In contrast to other adult mammals, the tendons of Murphy Roths Large (MRL/MpJ) mice possess a superior healing capacity following acute and overuse injuries. Here, we hypothesized that the application of biological cues derived from the local MRL/MpJ tendon environment would direct otherwise scar-mediated tenocytes towards a pro-regenerative MRL/MpJ-like phenotype. We identified soluble factors enriched in the secretome of MRL/MpJ tenocytes using bioreactor systems and quantitative proteomics. We then demonstrated that the combined administration of structural and soluble constituents isolated from decellularized MRL/MpJ tendon provisional ECM (dPECM) and the secretome stimulate scar-mediated rodent tenocytes towards enhanced mechanosensitivity, proliferation, intercellular communication, and ECM deposition associated with MRL/MpJ cell behavior. Our findings highlight key biological mechanisms that drive MRL/MpJ tenocyte activity and their interspecies utility to be harnessed for therapeutic strategies that promote pro-regenerative healing outcomes. Teaser Proteins enriched in a super-healer mouse strain elicit interspecies utility in promoting pro-regenerative tenocyte behavior.
Collapse
|
44
|
Yılmaz D, Marques FC, Fischer Y, Zimmermann S, Hwang G, Atkins PR, Mathavan N, Singh A, de Souza PP, Kuhn GA, Wehrle E, Müller R. Elucidating the mechano-molecular dynamics of TRAP activity using CRISPR/Cas9 mediated fluorescent reporter mice. Heliyon 2024; 10:e32949. [PMID: 39021958 PMCID: PMC11252717 DOI: 10.1016/j.heliyon.2024.e32949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/22/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
Osteoclasts are essential for bone remodeling by adapting their resorptive activity in response to their mechanical in vivo environment. However, the molecular mechanisms underlying this process remain unclear. Here, we demonstrated the role of tartrate-resistant acid phosphatase (TRAP, Acp5), a key enzyme secreted by osteoclasts, in bone remodeling and mechanosensitivity. Using CRISPR/Cas9 reporter mice, we demonstrated bone cell reporter (BCRIbsp/Acp5) mice feature fluorescent TRAP-deficient osteoclasts and examined their activity during mechanically driven trabecular bone remodeling. Although BCRIbsp/Acp5 mice exhibited trabecular bone impairments and reduced resorption capacity in vitro, RNA sequencing revealed unchanged levels of key osteoclast-associated genes such as Ctsk, Mmp9, and Calcr. These findings, in conjunction with serum carboxy-terminal collagen crosslinks (CTX) and in vivo mechanical loading outcomes collectively indicated an unaltered bone resorption capacity of osteoclasts in vivo. Furthermore, we demonstrated similar mechanoregulation during trabecular bone remodeling in BCRIbsp/Acp5 and wild-type (WT) mice. Hence, this study provides valuable insights into the dynamics of TRAP activity in the context of bone remodeling and mechanosensation.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | | | | | - Gaonhae Hwang
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Penny R. Atkins
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- Department of Orthopaedics, University of Utah, 590 Wakara Way, Salt Lake City, USA
| | | | - Amit Singh
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Pedro P.C. de Souza
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia, Brazil
| | - Gisela A. Kuhn
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
45
|
Fowler A, Knaus KR, Khuu S, Khalilimeybodi A, Schenk S, Ward SR, Fry AC, Rangamani P, McCulloch AD. Network model of skeletal muscle cell signalling predicts differential responses to endurance and resistance exercise training. Exp Physiol 2024; 109:939-955. [PMID: 38643471 PMCID: PMC11140181 DOI: 10.1113/ep091712] [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: 12/06/2023] [Accepted: 03/20/2024] [Indexed: 04/22/2024]
Abstract
Exercise-induced muscle adaptations vary based on exercise modality and intensity. We constructed a signalling network model from 87 published studies of human or rodent skeletal muscle cell responses to endurance or resistance exercise in vivo or simulated exercise in vitro. The network comprises 259 signalling interactions between 120 nodes, representing eight membrane receptors and eight canonical signalling pathways regulating 14 transcriptional regulators, 28 target genes and 12 exercise-induced phenotypes. Using this network, we formulated a logic-based ordinary differential equation model predicting time-dependent molecular and phenotypic alterations following acute endurance and resistance exercises. Compared with nine independent studies, the model accurately predicted 18/21 (85%) acute responses to resistance exercise and 12/16 (75%) acute responses to endurance exercise. Detailed sensitivity analysis of differential phenotypic responses to resistance and endurance training showed that, in the model, exercise regulates cell growth and protein synthesis primarily by signalling via mechanistic target of rapamycin, which is activated by Akt and inhibited in endurance exercise by AMP-activated protein kinase. Endurance exercise preferentially activates inflammation via reactive oxygen species and nuclear factor κB signalling. Furthermore, the expected preferential activation of mitochondrial biogenesis by endurance exercise was counterbalanced in the model by protein kinase C in response to resistance training. This model provides a new tool for investigating cross-talk between skeletal muscle signalling pathways activated by endurance and resistance exercise, and the mechanisms of interactions such as the interference effects of endurance training on resistance exercise outcomes.
Collapse
Affiliation(s)
- Annabelle Fowler
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Katherine R. Knaus
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Stephanie Khuu
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Simon Schenk
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Samuel R. Ward
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew C. Fry
- Department of Health, Sport and Exercise SciencesUniversity of KansasLawrenceKansasUSA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew D. McCulloch
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
46
|
Zhu M, Meglicki M, Lamba A, Wang P, Royer C, Turner K, Jauhar MA, Jones C, Child T, Coward K, Na J, Zernicka-Goetz M. Tead4 and Tfap2c generate bipotency and a bistable switch in totipotent embryos to promote robust lineage diversification. Nat Struct Mol Biol 2024; 31:964-976. [PMID: 38789684 PMCID: PMC11189297 DOI: 10.1038/s41594-024-01311-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/09/2024] [Indexed: 05/26/2024]
Abstract
The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.
Collapse
Affiliation(s)
- Meng Zhu
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Maciej Meglicki
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Adiyant Lamba
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Peizhe Wang
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Christophe Royer
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Karen Turner
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, UK
| | - Muhammad Abdullah Jauhar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Celine Jones
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Tim Child
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kevin Coward
- Nuffield Department of Women's and Reproductive Health, Level 3, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jie Na
- Centre for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
47
|
Lin G, Xia A, Qiao J, Zhang H, Chen P, Zhou P, Hu Q, Xiang Z, Zhang S, Li L, Yang S. Identification of a new class of activators of the Hippo pathway with antitumor activity in vitro and in vivo. Biochem Pharmacol 2024; 224:116217. [PMID: 38641306 DOI: 10.1016/j.bcp.2024.116217] [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: 11/24/2023] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
The Hippo pathway is a key regulator of tissue growth, organ size, and tumorigenesis. Activating the Hippo pathway by gene editing or pharmaceutical intervention has been proven to be a new therapeutic strategy for treatment of the Hippo pathway-dependent cancers. To now, a number of compounds that directly target the downstream effector proteins of Hippo pathway, including YAP and TEADs, have been disclosed, but very few Hippo pathway activators are reported. Here, we discovered a new class of Hippo pathway activator, YL-602, which inhibited CTGF expression in cells irrespective of cell density and the presence of serum. Mechanistically, YL-602 activates the Hippo pathway via MST1/2, which is different from known activators of Hippo pathway. In vitro, YL-602 significantly induced tumor cell apoptosis and inhibited colony formation of tumor cells. In vivo, oral administration of YL-602 substantially suppressed the growth of cancer cells by activation of Hippo pathway. Overall, YL-602 could be a promising lead compound, and deserves further investigation for its mechanism of action and therapeutic applications.
Collapse
Affiliation(s)
- Guifeng Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Anjie Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingxin Qiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hailin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pei Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pei Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyu Xiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
48
|
Yang S, Golkaram M, Oh S, Oh Y, Cho Y, Yoe J, Ju S, Lalli MA, Park SY, Lee Y, Jang J. ETV4 is a mechanical transducer linking cell crowding dynamics to lineage specification. Nat Cell Biol 2024; 26:903-916. [PMID: 38702503 PMCID: PMC11178500 DOI: 10.1038/s41556-024-01415-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: 02/21/2023] [Accepted: 04/03/2024] [Indexed: 05/06/2024]
Abstract
Dynamic changes in mechanical microenvironments, such as cell crowding, regulate lineage fates as well as cell proliferation. Although regulatory mechanisms for contact inhibition of proliferation have been extensively studied, it remains unclear how cell crowding induces lineage specification. Here we found that a well-known oncogene, ETS variant transcription factor 4 (ETV4), serves as a molecular transducer that links mechanical microenvironments and gene expression. In a growing epithelium of human embryonic stem cells, cell crowding dynamics is translated into ETV4 expression, serving as a pre-pattern for future lineage fates. A switch-like ETV4 inactivation by cell crowding derepresses the potential for neuroectoderm differentiation in human embryonic stem cell epithelia. Mechanistically, cell crowding inactivates the integrin-actomyosin pathway and blocks the endocytosis of fibroblast growth factor receptors (FGFRs). The disrupted FGFR endocytosis induces a marked decrease in ETV4 protein stability through ERK inactivation. Mathematical modelling demonstrates that the dynamics of cell density in a growing human embryonic stem cell epithelium precisely determines the spatiotemporal ETV4 expression pattern and, consequently, the timing and geometry of lineage development. Our findings suggest that cell crowding dynamics in a stem cell epithelium drives spatiotemporal lineage specification using ETV4 as a key mechanical transducer.
Collapse
Affiliation(s)
- Seungbok Yang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Mahdi Golkaram
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Seyoun Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yujeong Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yoonjae Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jeehyun Yoe
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sungeun Ju
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Matthew A Lalli
- Seaver Autism Center for Research and Treatment at Mount Sinai, New York, NY, USA
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jiwon Jang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
49
|
Koçak G, Uyulgan S, Polatlı E, Sarı V, Kahveci B, Bursali A, Binokay L, Reçber T, Nemutlu E, Mardinoğlu A, Karakülah G, Utine CA, Güven S. Generation of Anterior Segment of the Eye Cells from hiPSCs in Microfluidic Platforms. Adv Biol (Weinh) 2024; 8:e2400018. [PMID: 38640945 DOI: 10.1002/adbi.202400018] [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: 01/10/2024] [Revised: 03/10/2024] [Indexed: 04/21/2024]
Abstract
Ophthalmic diseases affect many people, causing partial or total loss of vision and a reduced quality of life. The anterior segment of the eye accounts for nearly half of all visual impairment that can lead to blindness. Therefore, there is a growing demand for ocular research and regenerative medicine that specifically targets the anterior segment to improve vision quality. This study aims to generate a microfluidic platform for investigating the formation of the anterior segment of the eye derived from human induced pluripotent stem cells (hiPSC) under various spatial-mechanoresponsive conditions. Microfluidic platforms are developed to examine the effects of dynamic conditions on the generation of hiPSCs-derived ocular organoids. The differentiation protocol is validated, and mechanoresponsive genes are identified through transcriptomic analysis. Several culture strategies is implemented for the anterior segment of eye cells in a microfluidic chip. hiPSC-derived cells showed anterior eye cell characteristics in mRNA and protein expression levels under dynamic culture conditions. The expression levels of yes-associated protein and transcriptional coactivator PDZ binding motif (YAP/TAZ) and PIEZO1, varied depending on the differentiation and growth conditions of the cells, as well as the metabolomic profiles under dynamic culture conditions.
Collapse
Affiliation(s)
- Gamze Koçak
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Sude Uyulgan
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Elifsu Polatlı
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Vedat Sarı
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Burak Kahveci
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Ahmet Bursali
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
| | - Leman Binokay
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Sıhhiye, Ankara, 06100, Türkiye
| | - Adil Mardinoğlu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Canan Aslı Utine
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Department of Ophthalmology, Dokuz Eylül University Hospital, Dokuz Eylül University, Izmir, 35340, Türkiye
| | - Sinan Güven
- Izmir Biomedicine and Genome Center, Izmir, 35340, Türkiye
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, 35340, Türkiye
- Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylül University, Izmir, 35340, Türkiye
| |
Collapse
|
50
|
Park S, Ryu WJ, Kim TY, Hwang Y, Han HJ, Lee JD, Kim GM, Sohn J, Kim SK, Kim MH, Kim J. Overcoming BRAF and CDK4/6 inhibitor resistance by inhibiting MAP3K3-dependent protection against YAP lysosomal degradation. Exp Mol Med 2024; 56:987-1000. [PMID: 38622197 PMCID: PMC11059244 DOI: 10.1038/s12276-024-01210-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 04/17/2024] Open
Abstract
Transcriptional programs governed by YAP play key roles in conferring resistance to various molecular-targeted anticancer agents. Strategies aimed at inhibiting YAP activity have garnered substantial interest as a means to overcome drug resistance. However, despite extensive research into the canonical Hippo-YAP pathway, few clinical agents are currently available to counteract YAP-associated drug resistance. Here, we present a novel mechanism of YAP stability regulation by MAP3K3 that is independent of Hippo kinases. Furthermore, we identified MAP3K3 as a target for overcoming anticancer drug resistance. Depletion of MAP3K3 led to a substantial reduction in the YAP protein level in melanoma and breast cancer cells. Mass spectrometry analysis revealed that MAP3K3 phosphorylates YAP at serine 405. This MAP3K3-mediated phosphorylation event hindered the binding of the E3 ubiquitin ligase FBXW7 to YAP, thereby preventing its p62-mediated lysosomal degradation. Robust YAP activation was observed in CDK4/6 inhibitor-resistant luminal breast cancer cells. Knockdown or pharmacological inhibition of MAP3K3 effectively suppressed YAP activity and restored CDK4/6 inhibitor sensitivity. Similarly, elevated MAP3K3 expression supported the prosurvival activity of YAP in BRAF inhibitor-resistant melanoma cells. Inhibition of MAP3K3 decreased YAP-dependent cell proliferation and successfully restored BRAF inhibitor sensitivity. In conclusion, our study reveals a previously unrecognized mechanism for the regulation of YAP stability, suggesting MAP3K3 inhibition as a promising strategy for overcoming resistance to CDK4/6 and BRAF inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Sanghyun Park
- Department of Dermatology, Chonnam National University Medical School, Gwangju, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Won-Ji Ryu
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Yeong Kim
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yumi Hwang
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Ju Han
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Dong Lee
- Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Gun Min Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea.
| | - Min Hwan Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|