1
|
Cao H, Tao Y, Jin R, Li P, Zhou H, Cheng J. Proteomics reveals the key transcription-related factors mediating obstructive nephropathy in pediatric patients and mice. Ren Fail 2025; 47:2443032. [PMID: 39743726 DOI: 10.1080/0886022x.2024.2443032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Obstructive nephropathy is one of the leading causes of kidney injury in infants and children. Increasing evidence has shown that transcription-related factors (TRFs), including transcription factors and cofactors, are associated with kidney diseases. However, a global landscape of dysregulated TRFs in pediatric patients with obstructive nephropathy is lacking. METHODS We mined the data from our previous proteomic study for the TRF profile in pediatric patients with obstructive nephropathy and unilateral ureteral obstruction (UUO) mice. Gene ontology (GO) analysis was performed to determine pathways that were enriched in the dysregulated TRFs. We then took advantage of kidney samples from patients and UUO mice to verify the selected TRFs by immunoblots. RESULTS The proteomes identified a total of 140 human TRFs with 28 upregulated and 1 downregulated, and 160 murine TRFs with 88 upregulated and 1 downregulated (fold change >2 or <0.5). These dysregulated TRFs were enriched in the inflammatory signalings, such as janus kinase/signal transducer and activator of transcription (JAK-STAT) and tumor necrosis factor (TNF) pathways. Of note, the transforming growth factor (TGF)-β signaling pathway, which is the master regulator of organ fibrosis, was enriched in both patients and mice. Cross-species analysis showed 16 key TRFs that might mediate obstructive nephropathy in patients and UUO mice. Moreover, we verified a significant dysregulation of three previously unexplored TRFs; prohibitin (PHB), regulatory factor X 1 (RFX1), and activity-dependent neuroprotector homeobox protein (ADNP), in patients and mice. CONCLUSIONS Our study uncovered key TRFs in the obstructed kidneys and provided additional molecular insights into obstructive nephropathy.
Collapse
Affiliation(s)
- Hualin Cao
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuandong Tao
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Ruyue Jin
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Pin Li
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Huixia Zhou
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Yuan Q, Xia X, Yuan Y, Chen Q, Feng X. A YAP-derived peptide blocks YAP-TEAD signaling and suppresses cell proliferation. Cell Signal 2025; 131:111738. [PMID: 40081550 DOI: 10.1016/j.cellsig.2025.111738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Yes-associated protein (YAP), a pivotal transcriptional co-activator in cell growth regulation, exerts its function through interactions with transcriptional factors like TEAD. Ectopic activation of YAP causes excessive cell proliferation, leading to multiple human diseases, including cancers. However, current pharmacological YAP inhibition lacks specificity and may have unintended effects, necessitating the development of direct YAP-derived inhibitors. In this study, we designed a novel YAP-derived peptide, TBDi, that specifically disrupted YAP-TEAD interaction and exhibited robust inhibition of TEAD activity. Mechanistically, TBDi directly binds to TEAD, blocking the physical interaction between YAP and TEAD. Transcriptomic analysis revealed that TBDi significantly altered gene expression profiles associated with TEAD activity, including downregulation of signature genes like CYR61 and CTGF. Functionally, TBDi emerged as a potent suppressor of cell proliferation, inhibiting cell proliferation to a degree comparable to YAP/TAZ knockdown. Altogether, our study not only identifies TBDi as a promising tool to block YAP-TEAD axis, but also offers insights for potential therapeutic interventions in diseases.
Collapse
Affiliation(s)
- Qiuyun Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, Zhejiang 310006, China
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Akao K, Sato T, Mishiro-Sato E, Mukai S, Ghani FI, Kondo-Ida L, Imaizumi K, Sekido Y. TEAD-Independent Cell Growth of Hippo-Inactive Mesothelioma Cells: Unveiling Resistance to TEAD Inhibitor K-975 through MYC Signaling Activation. Mol Cancer Ther 2025; 24:709-719. [PMID: 39686607 DOI: 10.1158/1535-7163.mct-24-0308] [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: 05/01/2024] [Revised: 09/30/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Inactivation of tumor-suppressive Hippo signaling pathway is frequently observed in mesothelioma, which leads to the activation of yes-associated protein (YAP) and TAZ (also known as WW domain-containing transcription regulator 1; YAP/TAZ) transcriptional coactivators. YAP/TAZ form complexes with TEAD family members, DNA-binding proteins, to activate transcription, which promotes cancer cell growth and proliferation. Recently developed TEAD inhibitors exhibit antitumor activity by inhibiting the formation of the transcription complex through binding to TEAD; however, the antitumor activity of TEAD inhibitors against mesothelioma remains to be fully elucidated. Here, we show that the TEAD inhibitor K-975 acts as a pan-TEAD inhibitor and selectively inhibits the binding of TEAD-binding proteins, especially YAP/TAZ, in mesothelioma cells. In studies using a panel of mesothelioma cell lines, K-975 showed a significant growth inhibitory effect on Hippo-inactivated mesothelioma cells, but some of these cell lines exhibited primary resistance to K-975. Differential gene expression analysis revealed that cells resistant to K-975 exhibited activation of MYC signaling in the presence of K-975, and cells overexpressed with MYC showed strong drug resistance, in vitro and in vivo. Our study revealed the features of a subset of mesothelioma cells that proliferate in a TEAD-independent manner and provides important insights for the successful development of therapeutic strategies for mesothelioma with Hippo pathway inactivation.
Collapse
Affiliation(s)
- Ken Akao
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tatsuhiro Sato
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Emi Mishiro-Sato
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Satomi Mukai
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Farhana Ishrat Ghani
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Lisa Kondo-Ida
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kazuyoshi Imaizumi
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshitaka Sekido
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Cancer Genetics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
4
|
Liu S, Chen Y, Zhou G, Sun C, Ma M, Huang R, Li X, Liang X, Shi C, Wu W, Yan X, Wang L, Han J. Uniform and controllable surface nano-structure on polyetheretherketone implants can regulate mechanical property to enhance soft tissue integration through Piezo1/TGF-β1 signaling axis. Mater Today Bio 2025; 31:101645. [PMID: 40151615 PMCID: PMC11946874 DOI: 10.1016/j.mtbio.2025.101645] [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: 12/17/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Polyetheretherketone (PEEK) has potential to repair the orbital floor bone defects following craniofacial trauma and orbital surgery. However, the inertness of the material impedes the soft tissue integration of implants, leading to complications such as implant migration and infection. Surface patterning modification on PEEK can promote the surface hydrophily to enhance better soft tissue integration, but it is difficult to obtain the uniform and controllable nano-structure. In this study, hot-pressing technology on PEEK implant was used to produce surface nanopores with a uniform diameter of 200, 500, 800 nm. Depending on the controllable craft, the surficial mechanical properties of PEEK can be regulated and assessed by finite element analysis. Furthermore, 500 nm interface has better mechanical properties to promote the proliferation, migration, and fibrosis of fibroblasts and achieved optimal integration effects in animal implantation experiments. To explore the mechanism of biological responses, transcriptomics and molecular biology experiments revealed that Piezo1/TGF-β1 axis played a critical role in the response of soft tissue cells to the mechanical stimulation of PEEK. Our study has established a novel modification technology for constructing uniform and controllable nanostructures on the surface of PEEK, thereby promoting the soft tissues integration with implants and improving the anchoring effect.
Collapse
Affiliation(s)
- Sida Liu
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
- The 940 Hospital of the Joint Logistic Support Force, 730050, Lanzhou, Gansu Province, China
| | - Yixuan Chen
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Gandong Zhou
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Changning Sun
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Minghai Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Rou Huang
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xing Li
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Xiao Liang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Changquan Shi
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054, Xi'an, Shaanxi Province, China
| | - Weiwei Wu
- School of Advanced Materials and Nanotechnology, Xidian University, 710126, Xi'an, Shaanxi Province, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, 710038, Xi'an, Shaanxi Province, China
| |
Collapse
|
5
|
Huang C, Li W, Shen C, Jiang B, Zhang K, Li X, Zhong W, Li Z, Chen Z, Chen C, Jian X, Liu X, Huang H, Yang L, Yu B. YAP1 facilitates the pathogenesis of psoriasis via modulating keratinocyte proliferation and inflammation. Cell Death Dis 2025; 16:186. [PMID: 40108109 PMCID: PMC11923178 DOI: 10.1038/s41419-025-07521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Psoriasis is an autoinflammatory skin disease characterized by the abnormal activation of epidermal keratinocytes. The Hippo-YAP pathway is an evolutionarily conserved pathway that plays important roles in organ size control and tumorigenesis. Recently, accumulating evidence demonstrated that YAP1, the core downstream component of Hippo-YAP pathway, was up-regulated in psoriasis patients, suggesting its possible role in psoriasis development. However, its precise function and mechanism in psoriasis pathogenesis are still not well-clarified. In the present study, we confirmed the up-regulation of YAP1 in psoriasis keratinocytes by measuring its expression in psoriatic patient skins, psoriatic-like cellular model, and IMQ-induced mouse model. Further functional studies showed that YAP1 promoted keratinocyte proliferation and inflammation in vitro. Meanwhile, VP, a selective YAP1 antagonist, inhibited keratinocyte proliferation and inflammatory factor production in a dose-dependent way. Moreover, intradermal injection of si-Yap1 or VP hindered psoriasis development by impeding epidermal hyperplasia and relieving systemic inflammatory response in the IMQ-induced mouse model. Therefore, our findings suggest that YAP1 plays a crucial role in psoriasis pathogenesis through modulating keratinocyte activation and may serve as a novel target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Cong Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
| | - Wenting Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- The Digestive and Reproductive System Cancers Precise Prevention Engineering Research Center of Jiangsu Province, Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai' an, Jiangsu, China
| | - Changbing Shen
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bin Jiang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Kaoyuan Zhang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiahong Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Weilong Zhong
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Zizhuo Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Zhenzhen Chen
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Chaofeng Chen
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xingling Jian
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiaoming Liu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Haiyan Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Lili Yang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
| |
Collapse
|
6
|
Cherkashina O, Tsitrina A, Abolin D, Morgun E, Kosykh A, Sabirov M, Vorotelyak E, Kalabusheva E. The Recovery of Epidermal Proliferation Pattern in Human Skin Xenograft. Cells 2025; 14:448. [PMID: 40136697 PMCID: PMC11941497 DOI: 10.3390/cells14060448] [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: 11/22/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Abnormalities in epidermal keratinocyte proliferation are a characteristic feature of a range of dermatological conditions. These include hyperproliferative states in psoriasis and dermatitis as well as hypoproliferative states in chronic wounds. This emphasises the importance of investigating the proliferation kinetics under conditions of healthy skin and identifying the key regulators of epidermal homeostasis, maintenance, and recovery following wound healing. Animal models contribute to our understanding of human epidermal self-renewal. Human skin xenografting overcomes the ethical limitations of studying human skin during regeneration. The application of this approach has allowed for the identification of a single population of stem cells and both slowly and rapidly cycling progenitors within the epidermal basal layer and the mapping of their location in relation to rete ridges and hair follicles. Furthermore, we have traced the dynamics of the proliferation pattern reorganization that occurs during epidermal regeneration, underlining the role of YAP activity in epidermal relief formation.
Collapse
Affiliation(s)
- Olga Cherkashina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
| | - Alexandra Tsitrina
- Ilse Katz Institute of Nanoscale Science, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Danila Abolin
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
| | - Elena Morgun
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anastasiya Kosykh
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Marat Sabirov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
| | - Ekaterina Vorotelyak
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
| | - Ekaterina Kalabusheva
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia (E.K.)
| |
Collapse
|
7
|
Yuan Q, Yuan Y, Peng Y, Xia X, Chen Q, Yu FX, Feng X. Distinct effects of Hippo-YAP/TAZ and YAP/TAZ-TEAD in epithelial maintenance and repair. Biochem Biophys Res Commun 2025; 751:151427. [PMID: 39903968 DOI: 10.1016/j.bbrc.2025.151427] [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: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
The maintenance of epithelial homeostasis is essential for preserving tissue architecture and function, and the transcriptional co-activators YAP/TAZ are central to this regulatory network. Although the Hippo-YAP/TAZ-TEAD axis is known to govern epithelial integrity, it remains unclear to what extent Hippo-controlled YAP/TAZ activity overlaps with, or diverges from, YAP/TAZ-TEAD-dependent transcriptional programs in maintaining epithelial homeostasis. Here, we address this question by employing two complementary mouse models: "SuperHippo," which suppresses YAP/TAZ activity through enhanced Hippo pathway engagement, and "TEADi," which selectively disrupts YAP/TAZ-TEAD interactions. Our results revealed that while both models led to increased epithelial thickness in skin epithelial, SuperHippo mice exhibited pronounced epithelial impairment in oral mucosa, and markedly delayed wound healing. In contrast, TEADi mice displayed tissue-specific phenotypes with minimal disruption to oral epithelium integrity or wound repair. These findings indicate that Hippo-mediated YAP/TAZ regulation may extend beyond TEAD-dependent transcription. Our work clarifies the distinct contributions of Hippo-YAP/TAZ signaling and YAP/TAZ-TEAD interaction to epithelial maintenance and provides a basis for the development of therapeutic strategies targeting YAP/TAZ in epithelial disorders.
Collapse
Affiliation(s)
- Qiuyun Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, 310006, Zhejiang, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
8
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
9
|
Sato K, Faraji F, Cervantes-Villagrana RD, Wu X, Koshizuka K, Ishikawa T, Iglesias-Bartolome R, Chen L, Miliani de Marval PL, Gwaltney SL, Adler B, Gutkind JS. Targeting YAP/TAZ-TEAD signaling as a therapeutic approach in head and neck squamous cell carcinoma. Cancer Lett 2025; 612:217467. [PMID: 39826667 PMCID: PMC12044704 DOI: 10.1016/j.canlet.2025.217467] [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: 12/05/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Genetic alterations in Hippo pathway and the consequent activation of YAP/TAZ-TEAD are frequently observed in HPV-negative head and neck squamous cell carcinoma (HNSCC) patients. These include loss-of-function mutation and/or copy number loss of FAT1, and amplification of YAP1 and WWTR1 (encoding TAZ), thus raising the possibility that HNSCC cells may be dependent on YAP/TAZ-TEAD-mediated transcriptional programs. In this regard, the recent development of small molecule TEAD inhibitors (smTEADi) provides an opportunity to therapeutically target Hippo pathway dysregulation in human malignancies. This prompted us to explore the potential benefit of pharmacologically targeting the YAP/TAZ-TEAD axis in this disease. Here, we provide the pre-clinical evidence for the antitumor activity of novel smTEADi, SW-682 in HPV-negative HNSCC. By the use of multiple complementary experimental approaches, including siRNA knockdown, expression of a genetically encoded TEAD inhibitor peptide (pTEADi), and SW-682, we revealed that disruption of YAP/TAZ-TEAD interaction suppresses YAP/TAZ-TEAD-dependent target gene transcription and growth of HNSCC tumors. HNSCC cells with genetic alterations in FAT1 were more sensitive to TEADi compared to FAT1-wild type cells. Mechanistically, TEADi suppressed cell cycle progression and promoted the expression of terminal differentiation gene programs, resulting in tumor growth inhibition. A HNSCC-specific TEADi target gene set was defined from RNA-seq data, which is highly expressed in HNSCC tissues and predicts poor prognosis of HPV-negative HNSCC patients. Our results underscore that YAP/TAZ-TEAD-mediated growth-promoting programs represent a vulnerability in HPV-negative HNSCC, thus providing a pre-clinical rationale for the future evaluation of YAP/TAZ-TEAD targeting strategies as a therapeutic approach for HPV-negative HNSCC patients.
Collapse
Affiliation(s)
- Kuniaki Sato
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Farhoud Faraji
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Otolaryngology-Head and Neck Surgery, UC San Diego Health, La Jolla, CA, USA
| | - Rodolfo Daniel Cervantes-Villagrana
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Xingyu Wu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Keiichi Koshizuka
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Tomohiko Ishikawa
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lei Chen
- SpringWorks Therapeutics, Inc., Stamford, CT, USA
| | | | | | | | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Krantz S, Bell B, Lund K, Parra NS, Ng Y, De Oliveira Rosa N, Mukhopadhyay S, Croix BS, Sarin KY, Weigert R, Raimondi F, Iglesias-Bartolome R. Dissection of Gαs and Hedgehog signaling crosstalk reveals therapeutic opportunities to target adenosine receptor 2b in Hedgehog-dependent tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639530. [PMID: 40060632 PMCID: PMC11888225 DOI: 10.1101/2025.02.21.639530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Basal cell carcinoma (BCC), the most common human cancer, is driven by hyperactivation of Hedgehog Smoothened (SMO) and GLI transcription. Gαs and protein kinase A (PKA) negatively regulate Hedgehog signaling, offering an alternative BCC development and treatment pathway. Here, using histology alongside bulk and single-cell RNA sequencing, we find that mouse BCC-like tumors that originate from Gαs pathway inactivation are strikingly similar to those driven by canonical Hedgehog SMO. Interestingly, mutations that reduce Gαs and PKA activity are present in human BCC. Tumors from Gαs pathway inactivation are independent of the canonical Hedgehog regulators SMO and GPR161, establishing them as an SMO-independent oncogenic Hedgehog signaling model. Finally, we demonstrate that activation of the Gαs-coupled adenosine 2B receptor counteracts oncogenic SMO, reducing Hedgehog signaling and tumor formation and offering a potential therapeutic strategy for BCC.
Collapse
Affiliation(s)
- Sarah Krantz
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Braden Bell
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Katherine Lund
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Natalia Salinas Parra
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Yeap Ng
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
- Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | | | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Texas, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States
| | - Kavita Y Sarin
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
- Intravital Microscopy Core, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | | | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
11
|
Peng Y, Yuan Q, Zhou S, Gan J, Shen Z, Xia X, Jiang Y, Chen Q, Yuan Y, He G, Wei Q, Feng X. FAK mediates mechanical signaling to maintain epithelial homeostasis through YAP/TAZ-TEADs. Histochem Cell Biol 2025; 163:31. [PMID: 39918604 DOI: 10.1007/s00418-025-02360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Epithelial homeostasis ensures that the epithelium can perform its normal physiological functions. Mechanical signaling response through integrin-mediated adhesions of the basement membrane (BM) is crucial for maintaining epithelial homeostasis. The essential mechanosensors YAP and the paralog TAZ (YAP/TAZ) have been shown to play a critical role in epithelial homeostasis, but the key regulator that mediates mechanical signaling to YAP/TAZ in maintaining epithelial homeostasis has not been fully understood. In this study, we noticed that mechanical signals correlated with YAP/TAZ activation and basal state maintenance in epithelial stem/progenitor cells through immunohistochemistry. Subsequently, we found that inhibition of focal adhesion kinase (FAK) suppressed YAP/TAZ activation in the human keratinocyte line HaCaT cells. Furthermore, inhibition of the interaction between YAP/TAZ and the transcriptional enhanced associate domains (TEADs) resulted in the differentiation of HaCaT cells. Finally, we used primary mouse epithelial cells to reconstruct the epithelium in vitro and found that FAK inhibition led to both a reduction in YAP/TAZ activity and an increase of differentiation in the basal layer cells. In conclusion, our findings reveal that FAK mediates mechanical signaling to maintain epithelial homeostasis via YAP/TAZ-TEADs.
Collapse
Affiliation(s)
- Yang Peng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiuyun Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuting Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jianguo Gan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhengzhong Shen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuchen Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, 310000, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Gu He
- Department of Dermatology and Venerology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China.
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases and Frontier Innovation Center for Dental Medicine Plus and Research Unit of Oral Carcinogenesis and Management and Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
El Yousfi Y, Fernández-Farrán FJ, Oliver FJ, López-Rivas A, Yerbes R. Regulation of ER stress-induced apoptotic and inflammatory responses via YAP/TAZ-mediated control of the TRAIL-R2/DR5 signaling pathway. Cell Death Discov 2025; 11:42. [PMID: 39904986 PMCID: PMC11794427 DOI: 10.1038/s41420-025-02335-w] [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: 10/23/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
In tumors, cancer cells are frequently exposed to adverse environmental conditions that result in endoplasmic reticulum (ER) stress. Mechanical signals emerging from extracellular matrix (ECM) rigidity and cell shape regulate the activity of transcriptional co-activators Yes-associated protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding motif (TAZ). However, the role of ECM rigidity and YAP/TAZ in tumor cell fate decisions under ER stress remains relatively unexplored. Our results suggest that the YAP/TAZ system plays an important role in the control of ER stress-induced cell death by mechanical signaling arising from ECM stiffness in tumor cells. Mechanistically, YAP/TAZ regulates apoptosis induced by ER stress in tumor cells by controlling the activation of the TRAIL-R2/DR5-mediated extrinsic apoptotic pathway through a dual mechanism. On the one hand, the YAP/TAZ system prevents intracellular TRAIL-R2/DR5 clustering in tumor cells. On the other hand, it inhibits cFLIP down-regulation in tumor cells experiencing ER stress. In addition, YAP/TAZ controls the expression of pro-inflammatory interleukin-8 (IL-8/CXCL8) in tumor cells undergoing ER stress by a TRAIL-R2/DR5/caspase-8-dependent mechanism. Although other mechanisms may also be involved in controlling cell death and inflammation in tumor cells facing environmental stress, our results support a model in which regulation of the subcellular localization and activity of the YAP/TAZ transcriptional co-activators could contribute to the microenvironmental control of cell fate decisions in tumor cells undergoing ER stress.
Collapse
Affiliation(s)
- Y El Yousfi
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - F J Fernández-Farrán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - F J Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, Centro de Investigación Biomédica en Red de Cáncer CIBERONC, Granada, Spain
| | - A López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain
| | - R Yerbes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, Seville, Spain.
- Medical Physiology and Biophysics Department, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS) (Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla), Seville, Spain.
| |
Collapse
|
13
|
Faraji F, Ramirez SI, Clubb LM, Sato K, Burghi V, Hoang TS, Officer A, Anguiano Quiroz PY, Galloway WMG, Mikulski Z, Medetgul-Ernar K, Marangoni P, Jones KB, Cao Y, Molinolo AA, Kim K, Sakaguchi K, Califano JA, Smith Q, Goren A, Klein OD, Tamayo P, Gutkind JS. YAP-driven malignant reprogramming of oral epithelial stem cells at single cell resolution. Nat Commun 2025; 16:498. [PMID: 39779672 PMCID: PMC11711616 DOI: 10.1038/s41467-024-55660-6] [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: 08/24/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Tumor initiation represents the first step in tumorigenesis during which normal progenitor cells undergo cell fate transition to cancer. Capturing this process as it occurs in vivo, however, remains elusive. Here we employ spatiotemporally controlled oncogene activation and tumor suppressor inhibition together with multiomics to unveil the processes underlying oral epithelial progenitor cell reprogramming into tumor initiating cells at single cell resolution. Tumor initiating cells displayed a distinct stem-like state, defined by aberrant proliferative, hypoxic, squamous differentiation, and partial epithelial to mesenchymal invasive gene programs. YAP-mediated tumor initiating cell programs included activation of oncogenic transcriptional networks and mTOR signaling, and recruitment of myeloid cells to the invasive front contributing to tumor infiltration. Tumor initiating cell transcriptional programs are conserved in human head and neck cancer and associated with poor patient survival. These findings illuminate processes underlying cancer initiation at single cell resolution, and identify candidate targets for early cancer detection and prevention.
Collapse
Affiliation(s)
- Farhoud Faraji
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego Health, La Jolla, CA, 92037, USA.
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA.
| | - Sydney I Ramirez
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego Health, La Jolla, CA, 92093, USA
- La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Lauren M Clubb
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Kuniaki Sato
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Valeria Burghi
- Department of Pharmacology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Thomas S Hoang
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Adam Officer
- University of California San Diego, Bioinformatics and Systems Biology Graduate Program, La Jolla, CA, 92093, USA
| | - Paola Y Anguiano Quiroz
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - William M G Galloway
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA, 92697, USA
| | | | - Kate Medetgul-Ernar
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Pauline Marangoni
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kyle B Jones
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Yuwei Cao
- University of California San Diego, Biomedical Sciences Graduate Program, La Jolla, CA, 92093, USA
| | - Alfredo A Molinolo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Joseph A Califano
- Department of Otolaryngology-Head and Neck Surgery, University of California San Diego Health, La Jolla, CA, 92037, USA
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA, 92697, USA
| | - Alon Goren
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, 90048, USA
| | - Pablo Tamayo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA
- Division of Medical Genetics, Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Center for Novel Therapeutics, University of California San Diego, La Jolla, CA, 92037, USA
| | - J Silvio Gutkind
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego Health, La Jolla, CA, 92037, USA.
- Department of Pharmacology, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| |
Collapse
|
14
|
Xu Q, Wen X, Huang C, Lin Z, Xu Z, Sun C, Li L, Zhang S, Song S, Lou J, Hou Z, Chen Y, Li X, Chen L. RRFERV stabilizes TEAD1 expression to mediate nasopharyngeal cancer radiation resistance rendering tumor cells vulnerable to ferroptosis. Int J Surg 2025; 111:450-466. [PMID: 39352125 PMCID: PMC11745583 DOI: 10.1097/js9.0000000000002099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/18/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) regulate various essential biological processes, including cell proliferation, differentiation, apoptosis, migration, and invasion. However, in nasopharyngeal carcinoma (NPC), the clinical significance and mechanisms of lncRNAs in malignant progression are unknown. METHODS RNA sequencing and bioinformatic analysis were used to determine the potential function of RRFERV (radiation-resistant but ferroptosis-vulnerable), and its biological effects were investigated using in vitro and in vivo experiments. Western blotting, quantitative real-time reverse transcription PCR, RNA immunoprecipitation (RIP) assays, and flow cytometry detected RRFERV expression. Ferroptosis and lipid peroxidation were added to evaluate the relationship between it and radiotherapy resistance. RESULTS LncRNA-RRFERV was both highly expressed in NPC tissues and radiation-resistant cells. RRFERV is associated with poor clinical outcomes of NPC patients and stabilizes TEAD1 by competitive binding with microRNA-615-5p and microRNA-1293. RRFERV-TEAD1 signaling axis leads to malignant progression and radiotherapy resistance of NPC. Furthermore, we observed that NPC radiotherapy-resistance cells exist in a fragile oxidative stress equilibrium, which makes them more sensitive to ferroptosis inducers. Surprisingly, we found that RRFERV-TEAD1 signaling axis also plays a key role in mediating the lipid peroxidation levels of NPC radiotherapy-resistance cells through transcriptional activation of ACSL4/TFRC. CONCLUSIONS RRFERV serves as an independent prognostic factor in NPC. During the malignant progression of NPC caused by high expression of RRFERV, ferroptosis can be induced to effectively kill cancer cells and reverse the radiotherapy resistance of NPC cells, suggesting a potential treatment approach for recurrent and refractory NPC.
Collapse
Affiliation(s)
- Qingqing Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Xin Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chenglong Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Zaishan Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Zhen Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Ciming Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Li Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Suixian Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Shuanghong Song
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Jiahao Lou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Zan Hou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Yuanyuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Xuan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| | - Lei Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co, Guangzhou, China
| |
Collapse
|
15
|
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
|
16
|
Pankratova MD, Riabinin AA, Butova EA, Selivanovskiy AV, Morgun EI, Ulianov SV, Vorotelyak EA, Kalabusheva EP. YAP/TAZ Signalling Controls Epidermal Keratinocyte Fate. Int J Mol Sci 2024; 25:12903. [PMID: 39684613 DOI: 10.3390/ijms252312903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
The paralogues Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) control cell proliferation and cell fate determination from embryogenesis to ageing. In the skin epidermis, these proteins are involved in both homeostatic cell renewal and injury-induced regeneration and also drive carcinogenesis and other pathologies. YAP and TAZ are usually considered downstream of the Hippo pathway. However, they are the central integrating link for the signalling microenvironment since they are involved in the interplay with signalling cascades induced by growth factors, cytokines, and physical parameters of the extracellular matrix. In this review, we summarise the evidence on how YAP and TAZ are activated in epidermal keratinocytes; how YAP/TAZ-mediated signalling cooperates with other signalling molecules at the plasma membrane, cytoplasmic, and nuclear levels; and how YAP/TAZ ultimately controls transcription programmes, defining epidermal cell fate.
Collapse
Affiliation(s)
- Maria D Pankratova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Andrei A Riabinin
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elizaveta A Butova
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Arseniy V Selivanovskiy
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Elena I Morgun
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Sergey V Ulianov
- Laboratory of Structural-Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ekaterina A Vorotelyak
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Ekaterina P Kalabusheva
- Cell Biology Laboratory, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
17
|
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
|
18
|
Branch B, Yuan Y, Cascone M, Raimondi F, Iglesias-Bartolome R. An improved TEAD dominant-negative protein inhibitor to study Hippo YAP1/TAZ-dependent transcription. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.03.615022. [PMID: 39502361 PMCID: PMC11537315 DOI: 10.1101/2024.10.03.615022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Hippo signaling is one of the top pathways altered in human cancer, and intensive focus has been devoted to developing therapies targeting Hippo-dependent transcription mediated by YAP1 and TAZ interaction with TEAD proteins. However, a significant challenge in evaluating the efficacy of these approaches is the lack of models that can precisely characterize the consequences of TEAD inhibition. To address this gap, our laboratory developed a strategy that utilizes a fluorescently traceable, dominant-negative protein named TEADi. TEADi specifically blocks the nuclear interactions of TEAD with YAP1 and TAZ, enabling precise dissection of Hippo TEAD-dependent and independent effects on cell fate. In this study, we aimed to enhance TEADi effectiveness by altering post-transcriptional modification sites within its TEAD-binding domains (TBDs). We demonstrate that a D93E mutation in the YAP1 TBD significantly increases TEADi inhibitory capacity. Additionally, we find that TBDs derived from VGLL4 and YAP1 are insufficient to block TAZ-induced TEAD activity, revealing crucial differences in YAP1 and TAZ displacement mechanisms by dominant-negative TBDs. Structural differences in YAP1 and TAZ TBDs were also identified, which may contribute to the distinct binding of these proteins to TEAD. Our findings expand our understanding of TEAD regulation and highlight the potential of an optimized TEADi as a more potent, specific, and versatile tool for studying TEAD-transcriptional activity.
Collapse
Affiliation(s)
- Briana Branch
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
- Department of Cellular, Molecular, Developmental Biology and Biophysics Graduate Program, Johns Hopkins University, Baltimore, Maryland, United States
| | - Yao Yuan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| | | | | | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
19
|
Cui YY, Yang YH, Zheng JY, Ma HH, Han X, Liao CS, Zhou M. Elevated neutrophil extracellular trap levels in periodontitis: Implications for keratinization and barrier function in gingival epithelium. J Clin Periodontol 2024; 51:1210-1221. [PMID: 38839576 DOI: 10.1111/jcpe.14025] [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: 08/26/2023] [Revised: 04/18/2024] [Accepted: 05/19/2024] [Indexed: 06/07/2024]
Abstract
AIM To explore the levels of neutrophil extracellular traps (NETs) in patients with periodontitis and examine their effects on keratinization, barrier function of human gingival keratinocytes (HGKs) and the associated mechanisms. MATERIALS AND METHODS Saliva, gingival crevicular fluid (GCF), clinical periodontal parameters and gingival specimens were collected from 10 healthy control subjects and 10 patients with stage II-IV periodontitis to measure the NET levels. Subsequently, mRNA and protein levels of keratinization and barrier indicators, as well as intracellular calcium and epithelial barrier permeability, were analysed in HGKs after NET stimulation. RESULTS The study showed that NET levels significantly elevated in patients with periodontitis, across multiple specimens including saliva, GCF and gingival tissues. Stimulation of HGKs with NETs resulted in a decrease in the expressions of involucrin, cytokeratin 10, zonula occludens 1 and E-cadherin, along with decreased intracellular calcium levels and increased epithelial barrier permeability. Furthermore, the inhibition of keratinization by NETs is ERK-KLF4-dependent. CONCLUSIONS This study indicates that NETs impair the barrier function of HGKs and suppress keratinization through ERK/KLF4 axis. These findings provide potential targets for therapeutic approaches in periodontitis to address impaired gingival keratinization.
Collapse
Affiliation(s)
- Ya-Yun Cui
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Periodontology, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Yi-Heng Yang
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia-Yi Zheng
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui-Hui Ma
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xue Han
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Periodontology, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Chong-Shan Liao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Periodontology, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Min Zhou
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Periodontology, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Pinelli M, Makdissi S, Scur M, Parsons BD, Baker K, Otley A, MacIntyre B, Nguyen HD, Kim PK, Stadnyk AW, Di Cara F. Peroxisomal cholesterol metabolism regulates yap-signaling, which maintains intestinal epithelial barrier function and is altered in Crohn's disease. Cell Death Dis 2024; 15:536. [PMID: 39069546 DOI: 10.1038/s41419-024-06925-x] [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: 01/08/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Intestinal epithelial cells line the luminal surface to establish the intestinal barrier, where the cells play essential roles in the digestion of food, absorption of nutrients and water, protection from microbial infections, and maintaining symbiotic interactions with the commensal microbial populations. Maintaining and coordinating all these functions requires tight regulatory signaling, which is essential for intestinal homeostasis and organismal health. Dysfunction of intestinal epithelial cells, indeed, is linked to gastrointestinal disorders such as irritable bowel syndrome, inflammatory bowel disease, and gluten-related enteropathies. Emerging evidence suggests that peroxisome metabolic functions are crucial in maintaining intestinal epithelial cell functions and intestinal epithelium regeneration and, therefore, homeostasis. Here, we investigated the molecular mechanisms by which peroxisome metabolism impacts enteric health using the fruit fly Drosophila melanogaster and murine model organisms and clinical samples. We show that peroxisomes control cellular cholesterol, which in turn regulates the conserved yes-associated protein-signaling and contributes to intestinal epithelial structure and epithelial barrier function. Moreover, analysis of intestinal organoid cultures derived from biopsies of patients affected by Crohn's Disease revealed that the dysregulation of peroxisome number, excessive cellular cholesterol, and inhibition of Yap-signaling are markers of disease and could be novel diagnostic and/or therapeutic targets for treating Crohn's Disease. Our studies provided mechanistic insights on peroxisomal signaling in intestinal epithelial cell functions and identified cholesterol as a novel metabolic regulator of yes-associated protein-signaling in tissue homeostasis.
Collapse
Affiliation(s)
- Marinella Pinelli
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Stephanie Makdissi
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Michal Scur
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Brendon D Parsons
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Anthony Otley
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Brad MacIntyre
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Huong D Nguyen
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Peter K Kim
- The Hospital for Sick Children, Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Andrew W Stadnyk
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Francesca Di Cara
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
- Department of Pediatrics, Dalhousie University, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada.
| |
Collapse
|
21
|
Hu Q, Zhang B, Jing Y, Ma S, Hu L, Li J, Zheng Y, Xin Z, Peng J, Wang S, Cheng B, Qu J, Zhang W, Liu GH, Wang S. Single-nucleus transcriptomics uncovers a geroprotective role of YAP in primate gingival aging. Protein Cell 2024; 15:612-632. [PMID: 38577810 PMCID: PMC11259548 DOI: 10.1093/procel/pwae017] [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: 12/09/2023] [Accepted: 02/01/2024] [Indexed: 04/06/2024] Open
Abstract
Aging has a profound impact on the gingiva and significantly increases its susceptibility to periodontitis, a worldwide prevalent inflammatory disease. However, a systematic characterization and comprehensive understanding of the regulatory mechanism underlying gingival aging is still lacking. Here, we systematically dissected the phenotypic characteristics of gingiva during aging in primates and constructed the first single-nucleus transcriptomic landscape of gingival aging, by which a panel of cell type-specific signatures were elucidated. Epithelial cells were identified as the most affected cell types by aging in the gingiva. Further analyses pinpointed the crucial role of YAP in epithelial self-renew and homeostasis, which declined during aging in epithelial cells, especially in basal cells. The decline of YAP activity during aging was confirmed in the human gingival tissues, and downregulation of YAP in human primary gingival keratinocytes recapitulated the major phenotypic defects observed in the aged primate gingiva while overexpression of YAP showed rejuvenation effects. Our work provides an in-depth understanding of gingival aging and serves as a rich resource for developing novel strategies to combat aging-associated gingival diseases, with the ultimate goal of advancing periodontal health and promoting healthy aging.
Collapse
Affiliation(s)
- Qinchao Hu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510060, China
| | - Bin Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaobin Jing
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- International Center for Aging and Cancer, Hainan Medical University, Haikou 571199, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Lei Hu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Yandong Zheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijuan Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jianmin Peng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510060, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510060, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
22
|
Pedro MP, Lund K, Kang SWS, Chen T, Stuelten CH, Porat-Shliom N, Iglesias-Bartolome R. GPCR Screening Reveals that the Metabolite Receptor HCAR3 Regulates Epithelial Proliferation, Migration, and Cellular Respiration. J Invest Dermatol 2024; 144:1311-1321.e7. [PMID: 38103827 PMCID: PMC11116076 DOI: 10.1016/j.jid.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Epithelial cells in the skin and other tissues rely on signals from their environment to maintain homeostasis and respond to injury, and GPCRs play a critical role in this communication. A better understanding of the GPCRs expressed in epithelial cells will contribute to understanding the relationship between cells and their niche and could lead to developing new therapies to modulate cell fate. This study used human primary keratinocytes as a model to investigate the specific GPCRs regulating epithelial cell proliferation and differentiation. We identified 3 key receptors-HCAR3, LTB4R, and GPR137-and found that knockdown of these receptors led to changes in numerous gene networks that are important for maintaining cell identity and promoting proliferation while inhibiting differentiation. Our study also revealed that the metabolite receptor HCAR3 regulates keratinocyte migration and cellular metabolism. Knockdown of HCAR3 led to reduced keratinocyte migration and respiration, which could be attributed to altered metabolite use and aberrant mitochondrial morphology caused by the absence of the receptor. This study contributes to understanding the complex interplay between GPCR signaling and epithelial cell fate decisions.
Collapse
Affiliation(s)
- M Pilar Pedro
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Lund
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sun Woo Sophie Kang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ting Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina H Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Natalie Porat-Shliom
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
23
|
Kim JY, Quan T. Emerging Perspectives of YAP/TAZ in Human Skin Epidermal and Dermal Aging. Ann Dermatol 2024; 36:135-144. [PMID: 38816974 PMCID: PMC11148314 DOI: 10.5021/ad.23.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/25/2024] [Accepted: 02/18/2024] [Indexed: 06/01/2024] Open
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the Hippo signaling pathway, which plays a central role in tissue homeostasis, organ development, and regeneration. While the dysregulation of YAP/TAZ has been linked to various human diseases, their involvement in the aging of human skin has only recently begun to manifest. In the skin, the YAP/TAZ effectors emerge as central regulators in maintaining homeostasis of epidermal stem cells and dermal extracellular matrix, and thus intimately linked to skin aging processes. This review underscores recent molecular breakthroughs highlighting how age-related decline of YAP/TAZ activity impacts human epidermal and dermal aging. Gaining insight into the evolving roles of YAP/TAZ in human skin aging presents a promising avenue for the development of innovative therapeutic approaches aimed at enhancing skin health and addressing age-related skin conditions.
Collapse
Affiliation(s)
- Jun Young Kim
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Xia L, Lin H, Cao H, Lian J. Tenascin C as a novel zinc finger protein 750 target regulating the immunogenicity via DNA damage in lung squamous cell carcinoma. BMC Cancer 2024; 24:561. [PMID: 38711034 PMCID: PMC11071264 DOI: 10.1186/s12885-024-12285-8] [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: 01/18/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
Modulation of DNA damage repair in lung squamous cell carcinoma (LUSC) can result in the generation of neoantigens and heightened immunogenicity. Therefore, understanding DNA damage repair mechanisms holds significant clinical relevance for identifying targets for immunotherapy and devising therapeutic strategies. Our research has unveiled that the tumor suppressor zinc finger protein 750 (ZNF750) in LUSC binds to the promoter region of tenascin C (TNC), leading to reduced TNC expression. This modulation may impact the malignant behavior of tumor cells and is associated with patient prognosis. Additionally, single-cell RNA sequencing (scRNA-seq) of LUSC tissues has demonstrated an inverse correlation between ZNF750/TNC expression levels and immunogenicity. Manipulation of the ZNF750-TNC axis in vitro within LUSC cells has shown differential sensitivity to CD8+ cells, underscoring its pivotal role in regulating cellular immunogenicity. Further transcriptome sequencing analysis, DNA damage repair assay, and single-strand break analyses have revealed the involvement of the ZNF750-TNC axis in determining the preference for homologous recombination (HR) repair or non-homologous end joining (NHEJ) repair of DNA damage. with involvement of the Hippo/ERK signaling pathway. In summary, this study sheds light on the ZNF750-TNC axis's role in DNA damage repair regulation in LUSC, laying a groundwork for future translational research in immune cell therapy for LUSC.
Collapse
Affiliation(s)
- Lu Xia
- Xiamen Cell Therapy Research Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, CN, China.
| | - Hexin Lin
- Department of Gastrointestinal Oncology Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, CN, China
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350000, CN, China
| | - Huifen Cao
- Institute of Genomics, School of Medicine, Huaqiao University, Xiamen, 361000, CN, China.
| | - Jiabian Lian
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, CN, China.
| |
Collapse
|
25
|
Kim HJ, Choi Y, Lee Y, Hwangbo M, Kim J. OTUD6A orchestrates complex modulation of TEAD4-mediated transcriptional programs. FEBS Lett 2024; 598:1045-1060. [PMID: 38594215 DOI: 10.1002/1873-3468.14861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 04/11/2024]
Abstract
TEAD transcription factors play a central role in the Hippo signaling pathway. In this study, we focused on transcriptional enhancer factor TEF-3 (TEAD4), exploring its regulation by the deubiquitinase OTU domain-containing protein 6A (OTUD6A). We identified OTUD6A as a TEAD4-interacting deubiquitinase, positively influencing TEAD-driven transcription without altering TEAD4 stability. Structural analyses revealed specific interaction domains: the N-terminal domain of OTUD6A and the YAP-binding domain of TEAD4. Functional assays demonstrated the positive impact of OTUD6A on the transcription of YAP-TEAD target genes. Despite no impact on TEAD4 nuclear localization, OTUD6A selectively modulated nuclear interactions, enhancing YAP-TEAD4 complex formation while suppressing VGLL4 (transcription cofactor vestigial-like protein 4)-TEAD4 interaction. Critically, OTUD6A facilitated YAP-TEAD4 complex binding to target gene promoters. Our study unveils the regulatory landscape of OTUD6A on TEAD4, providing insights into diseases regulated by YAP-TEAD complexes.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yunsik Choi
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Yuri Lee
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Mi Hwangbo
- Department of Life Sciences, Sogang University, Seoul, Korea
| | - Jongchan Kim
- Department of Life Sciences, Sogang University, Seoul, Korea
| |
Collapse
|
26
|
Liu Y, Wang Y, Xu C, Zhang Y, Wang Y, Qin J, Lan HY, Wang L, Huang Y, Mak KK, Zheng Z, Xia Y. Activation of the YAP/KLF5 transcriptional cascade in renal tubular cells aggravates kidney injury. Mol Ther 2024; 32:1526-1539. [PMID: 38414248 PMCID: PMC11081877 DOI: 10.1016/j.ymthe.2024.02.031] [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: 09/13/2023] [Revised: 01/11/2024] [Accepted: 02/24/2024] [Indexed: 02/29/2024] Open
Abstract
The Hippo/YAP pathway plays a critical role in tissue homeostasis. Our previous work demonstrated that renal tubular YAP activation induced by double knockout (dKO) of the upstream Hippo kinases Mst1 and Mst2 promotes tubular injury and renal inflammation under basal conditions. However, the importance of tubular YAP activation remains to be established in injured kidneys in which many other injurious pathways are simultaneously activated. Here, we show that tubular YAP was already activated 6 h after unilateral ureteral obstruction (UUO). Tubular YAP deficiency greatly attenuated tubular cell overproliferation, tubular injury, and renal inflammation induced by UUO or cisplatin. YAP promoted the transcription of the transcription factor KLF5. Consistent with this, the elevated expression of KLF5 and its target genes in Mst1/2 dKO or UUO kidneys was blocked by ablation of Yap in tubular cells. Inhibition of KLF5 prevented tubular cell overproliferation, tubular injury, and renal inflammation in Mst1/2 dKO kidneys. Therefore, our results demonstrate that tubular YAP is a key player in kidney injury. YAP and KLF5 form a transcriptional cascade, where tubular YAP activation induced by kidney injury promotes KLF5 transcription. Activation of this cascade induces tubular cell overproliferation, tubular injury, and renal inflammation.
Collapse
Affiliation(s)
- Yang Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yu Wang
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Chunhua Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yu Zhang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinzhong Qin
- The Key Laboratory of Model Animal for Disease Study of the Ministry of Education, Model Animal Research Center, Nanjing University, Nanjing, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Kingston Kinglun Mak
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| | - Zhihua Zheng
- Department of Nephrology, Center of Nephrology and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Guangdong-Hong Kong Joint Laboratory for Immune and Genetic Kidney Disease, The Chinese University of Hong Kong, Hong Kong, China; Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
27
|
Guo Z, Guo L. Abnormal activation of RFC3, A YAP1/TEAD downstream target, promotes gastric cancer progression. Int J Clin Oncol 2024; 29:442-455. [PMID: 38383698 DOI: 10.1007/s10147-024-02478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 01/11/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a malignant tumor with a high mortality rate, and thus, it is necessary to explore molecular mechanisms underlying its progression. While replication factor C subunit 3 (RFC3) has been demonstrated to function as an oncogene in many cancers, its role in GC remains unclear. METHODS Tumor tissues were collected from clinical GC patients, and the expression of RFC3 was analyzed. NCI-N87 and HGC-27 cells were infected with lentivirus sh-RFC3 to knock down RFC3 expression. RFC3 expression levels were determined, in addition to cell biological behaviors both in vitro and in vivo. The relationship between RFC3 and the YAP1/TEAD signaling pathway was detected by dual luciferase reporter assay. RESULTS RFC3 was upregulated in GC tumor tissues. RFC3 knockdown inhibited cell proliferation, promoted cell apoptosis of GC cells, and suppressed cell migration and invasion. Moreover, depleted RFC3 suppressed tumor growth and metastasis in vivo. Mechanistically, the YAP1/TEAD axis activated RFC3 expression transcriptionally by binding to the RFC3 promoter. CONCLUSIONS RFC3 was transcriptional activated by the YAP1/TEAD signaling pathway, thus promoting GC progression. RFC3 may be a promising therapeutic target for GC.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Operating Room, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, People's Republic of China
| | - Lin Guo
- Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China.
| |
Collapse
|
28
|
Zhong Z, Jiao Z, Yu FX. The Hippo signaling pathway in development and regeneration. Cell Rep 2024; 43:113926. [PMID: 38457338 DOI: 10.1016/j.celrep.2024.113926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.
Collapse
Affiliation(s)
- Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
29
|
Longmate WM, Norton E, Duarte GA, Wu L, DiPersio MR, Lamar JM, DiPersio CM. Keratinocyte integrin α3β1 induces expression of the macrophage stimulating factor, CSF-1, through a YAP/TEAD-dependent mechanism. Matrix Biol 2024; 127:48-56. [PMID: 38340968 PMCID: PMC10923166 DOI: 10.1016/j.matbio.2024.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3β1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3β1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3β1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3β1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3β1 regulates Csf1 expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3β1-deficient wounds in vivo display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3β1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3β1 as a potential target of wound therapy.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Giesse Albeche Duarte
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Lei Wu
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA
| | - Mathieu R DiPersio
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
30
|
Kanai R, Norton E, Stern P, Hynes RO, Lamar JM. Identification of a Gene Signature That Predicts Dependence upon YAP/TAZ-TEAD. Cancers (Basel) 2024; 16:852. [PMID: 38473214 PMCID: PMC10930532 DOI: 10.3390/cancers16050852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Targeted therapies are effective cancer treatments when accompanied by accurate diagnostic tests that can help identify patients that will respond to those therapies. The YAP/TAZ-TEAD axis is activated and plays a causal role in several cancer types, and TEAD inhibitors are currently in early-phase clinical trials in cancer patients. However, a lack of a reliable way to identify tumors with YAP/TAZ-TEAD activation for most cancer types makes it difficult to determine which tumors will be susceptible to TEAD inhibitors. Here, we used a combination of RNA-seq and bioinformatic analysis of metastatic melanoma cells to develop a YAP/TAZ gene signature. We found that the genes in this signature are TEAD-dependent in several melanoma cell lines, and that their expression strongly correlates with YAP/TAZ activation in human melanomas. Using DepMap dependency data, we found that this YAP/TAZ signature was predictive of melanoma cell dependence upon YAP/TAZ or TEADs. Importantly, this was not limited to melanoma because this signature was also predictive when tested on a panel of over 1000 cancer cell lines representing numerous distinct cancer types. Our results suggest that YAP/TAZ gene signatures like ours may be effective tools to predict tumor cell dependence upon YAP/TAZ-TEAD, and thus potentially provide a means to identify patients likely to benefit from TEAD inhibitors.
Collapse
Affiliation(s)
- Ryan Kanai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| | - Patrick Stern
- Koch Institute for Integrative Cancer Research, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Richard O. Hynes
- Department of Biology, Koch Institute for Integrative Cancer Research, and Howard Hughes Medical Institute, at Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - John M. Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (R.K.); (E.N.)
| |
Collapse
|
31
|
Kodali N, Kumar KD, Schwartz RA. The role of scoliosis on the comorbidity and demographics of neurofibromatosis type 1 patients: A retrospective analysis of the National Inpatient Sample database. Exp Dermatol 2024; 33:e14996. [PMID: 38284196 DOI: 10.1111/exd.14996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 01/30/2024]
Abstract
Neurofibromatosis type 1 (NF1) is the most common neurocutaneous syndrome in the United States, affecting every 1 in 3000 individuals. NF1 occurs due to non-functional mutations in the NF1 gene, which expresses neurofibromin, a protein involved in tumour suppression. As a result, NF1 typically presents with non-cancerous neoplasm masses called neurofibromas across the body. Out of all NF1 abnormalities, the most common skeletal abnormality seen in around 10%-30% of NF1 patients is scoliosis, an improver curvature of the spine. However, there is a lack of research on the effects of scoliosis on demographics and morbidities of NF1 patients. We performed a national analysis to investigate the complex relationship between NF1 and scoliosis on patients' demographics and comorbidities. We conducted a retrospective cross-sectional analysis of the 2017 US National Inpatient Sample database using univariable Chi-square analysis and multivariable binary logistic regression analysis to determine the interplay of NF1 and scoliosis on patients' demographics and comorbidities. Our query resulted in 4635 total NF1 patients, of which 475 (10.25%) had scoliosis and 4160 (89.75%) did not. Demographic analysis showed that NF1 patients with scoliosis were typically younger, female and white compared to NF1 patients without scoliosis. Comorbidity analysis showed that NF1 patients with scoliosis were more likely to develop malignant brain neoplasms, epilepsy, hydrocephalus, pigmentation disorders, hypothyroidism, diabetes with chronic complications and coagulopathy disorders. NF1 patients with scoliosis were less likely to develop congestive heart failure, pulmonary circulation disease, peripheral vascular disease, paralysis, chronic pulmonary disease, lymphoma and psychosis. NF1 patients with scoliosis were predominantly younger, female, white patients. The presence of scoliosis in NF1 patients increases the risks for certain brain neoplasms and disorders but serves a protective effect against some pulmonary and cardiac complications.
Collapse
Affiliation(s)
- Nilesh Kodali
- Department of Dermatology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Keshav D Kumar
- Department of Dermatology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Robert A Schwartz
- Department of Dermatology, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
32
|
Mukhopadhyay S, Huang HY, Lin Z, Ranieri M, Li S, Sahu S, Liu Y, Ban Y, Guidry K, Hu H, Lopez A, Sherman F, Tan YJ, Lee YT, Armstrong AP, Dolgalev I, Sahu P, Zhang T, Lu W, Gray NS, Christensen JG, Tang TT, Velcheti V, Khodadadi-Jamayran A, Wong KK, Neel BG. Genome-Wide CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy. Cancer Res 2023; 83:4095-4111. [PMID: 37729426 PMCID: PMC10841254 DOI: 10.1158/0008-5472.can-23-2729] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
UNLABELLED Non-small lung cancers (NSCLC) frequently (∼30%) harbor KRAS driver mutations, half of which are KRASG12C. KRAS-mutant NSCLC with comutated STK11 and/or KEAP1 is particularly refractory to conventional, targeted, and immune therapy. Development of KRASG12C inhibitors (G12Ci) provided a major therapeutic advance, but resistance still limits their efficacy. To identify genes whose deletion augments efficacy of the G12Cis adagrasib (MRTX-849) or adagrasib plus TNO155 (SHP2i), we performed genome-wide CRISPR/Cas9 screens on KRAS/STK11-mutant NSCLC lines. Recurrent, potentially targetable, synthetic lethal (SL) genes were identified, including serine-threonine kinases, tRNA-modifying and proteoglycan synthesis enzymes, and YAP/TAZ/TEAD pathway components. Several SL genes were confirmed by siRNA/shRNA experiments, and the YAP/TAZ/TEAD pathway was extensively validated in vitro and in mice. Mechanistic studies showed that G12Ci treatment induced gene expression of RHO paralogs and activators, increased RHOA activation, and evoked ROCK-dependent nuclear translocation of YAP. Mice and patients with acquired G12Ci- or G12Ci/SHP2i-resistant tumors showed strong overlap with SL pathways, arguing for the relevance of the screen results. These findings provide a landscape of potential targets for future combination strategies, some of which can be tested rapidly in the clinic. SIGNIFICANCE Identification of synthetic lethal genes with KRASG12C using genome-wide CRISPR/Cas9 screening and credentialing of the ability of TEAD inhibition to enhance KRASG12C efficacy provides a roadmap for combination strategies. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hsin-Yi Huang
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Ziyan Lin
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Michela Ranieri
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Soumyadip Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yingzhuo Liu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Ban
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Kayla Guidry
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alfonso Lopez
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Fiona Sherman
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Jer Tan
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yeuan Ting Lee
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Amanda P. Armstrong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Igor Dolgalev
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Wenchao Lu
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | | | - Tracy T. Tang
- Vivace Therapeutics, Inc., San Mateo, California, United States
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| |
Collapse
|
33
|
Edwards AC, Stalnecker CA, Morales AJ, Taylor KE, Klomp JE, Klomp JA, Waters AM, Sudhakar N, Hallin J, Tang TT, Olson P, Post L, Christensen JG, Cox AD, Der CJ. TEAD Inhibition Overcomes YAP1/TAZ-Driven Primary and Acquired Resistance to KRASG12C Inhibitors. Cancer Res 2023; 83:4112-4129. [PMID: 37934103 PMCID: PMC10821578 DOI: 10.1158/0008-5472.can-23-2994] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Primary/intrinsic and treatment-induced acquired resistance limit the initial response rate to and long-term efficacy of direct inhibitors of the KRASG12C mutant in cancer. To identify potential mechanisms of resistance, we applied a CRISPR/Cas9 loss-of-function screen and observed loss of multiple components of the Hippo tumor suppressor pathway, which acts to suppress YAP1/TAZ-regulated gene transcription. YAP1/TAZ activation impaired the antiproliferative and proapoptotic effects of KRASG12C inhibitor (G12Ci) treatment in KRASG12C-mutant cancer cell lines. Conversely, genetic suppression of YAP1/WWTR1 (TAZ) enhanced G12Ci sensitivity. YAP1/TAZ activity overcame KRAS dependency through two distinct TEAD transcription factor-dependent mechanisms, which phenocopy KRAS effector signaling. First, TEAD stimulated ERK-independent transcription of genes normally regulated by ERK (BIRC5, CDC20, ECT2, FOSL1, and MYC) to promote progression through the cell cycle. Second, TEAD caused activation of PI3K-AKT-mTOR signaling to overcome apoptosis. G12Ci treatment-induced acquired resistance was also caused by YAP1/TAZ-TEAD activation. Accordingly, concurrent treatment with pharmacologic inhibitors of TEAD synergistically enhanced KRASG12C inhibitor antitumor activity in vitro and prolonged tumor suppression in vivo. In summary, these observations reveal YAP1/TAZ-TEAD signaling as a crucial driver of primary and acquired resistance to KRAS inhibition and support the use of TEAD inhibitors to enhance the antitumor efficacy of KRAS-targeted therapies. SIGNIFICANCE YAP1/TAZ-TEAD activation compensates for loss of KRAS effector signaling, establishing a mechanistic basis for concurrent inhibition of TEAD to enhance the efficacy of KRASG12C-selective inhibitor treatment of KRASG12C-mutant cancers. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- A. Cole Edwards
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Clint A. Stalnecker
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexis Jean Morales
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Khalilah E. Taylor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jennifer E. Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey A. Klomp
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew M. Waters
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Jill Hallin
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Leonard Post
- Vivace Therapeutics, Inc., San Mateo, California
| | | | - Adrienne D. Cox
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Channing J. Der
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
34
|
Claude-Taupin A, Isnard P, Bagattin A, Kuperwasser N, Roccio F, Ruscica B, Goudin N, Garfa-Traoré M, Regnier A, Turinsky L, Burtin M, Foretz M, Pontoglio M, Morel E, Viollet B, Terzi F, Codogno P, Dupont N. The AMPK-Sirtuin 1-YAP axis is regulated by fluid flow intensity and controls autophagy flux in kidney epithelial cells. Nat Commun 2023; 14:8056. [PMID: 38052799 PMCID: PMC10698145 DOI: 10.1038/s41467-023-43775-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/13/2023] [Indexed: 12/07/2023] Open
Abstract
Shear stress generated by urinary fluid flow is an important regulator of renal function. Its dysregulation is observed in various chronic and acute kidney diseases. Previously, we demonstrated that primary cilium-dependent autophagy allows kidney epithelial cells to adapt their metabolism in response to fluid flow. Here, we show that nuclear YAP/TAZ negatively regulates autophagy flux in kidney epithelial cells subjected to fluid flow. This crosstalk is supported by a primary cilium-dependent activation of AMPK and SIRT1, independently of the Hippo pathway. We confirm the relevance of the YAP/TAZ-autophagy molecular dialog in vivo using a zebrafish model of kidney development and a unilateral ureteral obstruction mouse model. In addition, an in vitro assay simulating pathological accelerated flow observed at early stages of chronic kidney disease (CKD) activates YAP, leading to a primary cilium-dependent inhibition of autophagic flux. We confirm this YAP/autophagy relationship in renal biopsies from patients suffering from diabetic kidney disease (DKD), the leading cause of CKD. Our findings demonstrate the importance of YAP/TAZ and autophagy in the translation of fluid flow into cellular and physiological responses. Dysregulation of this pathway is associated with the early onset of CKD.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France.
| | - Pierre Isnard
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Alessia Bagattin
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | | | - Federica Roccio
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Biagina Ruscica
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Nicolas Goudin
- Structure Fédérative de Recherche Necker, US24-UMS3633, Paris, France
| | | | - Alice Regnier
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Lisa Turinsky
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Martine Burtin
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Marc Foretz
- Institut Cochin, Inserm U1016 - CNRS UMR8104 - Université Paris Cité, 75014, Paris, France
| | - Marco Pontoglio
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Etienne Morel
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Benoit Viollet
- Institut Cochin, Inserm U1016 - CNRS UMR8104 - Université Paris Cité, 75014, Paris, France
| | - Fabiola Terzi
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Patrice Codogno
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Nicolas Dupont
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France.
| |
Collapse
|
35
|
Guo Z, Guo L. YAP/TEAD-induced PRIM1 contributes to the progression and poor prognosis of gastric carcinoma. Transl Oncol 2023; 38:101791. [PMID: 37741096 PMCID: PMC10541473 DOI: 10.1016/j.tranon.2023.101791] [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: 01/17/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Gastric carcinoma has a poor prognosis and low survival rate. PRIM1 is closely associated with the origin of DNA replication and serves as a carcinogenic factor in multiple tumors. This study aimed to explore the functions of PRIM1 in the progression of gastric carcinoma. The luciferase reporter assay examined the regulatory effect of YAP1/TEAD4 on PRIM1. A xenograft tumor mouse model was constructed to observe cancer cell proliferation in vivo. The upregulation of PRIM1 was found in gastric carcinoma cells and tissues, and it was associated with poor prognosis. Silencing PRIM1 inhibited cell proliferation, arrested the cell cycle, and upregulated Cdc25, Cyclin B, and Cdc2 expression. In addition, apoptosis was increased upon PRIM1 knockdown, accompanied by increased protein levels of cleaved caspase-3 and caspase-8. In vivo, knockdown of PRIM1 suppressed the growth of xenograft tumors formed by gastric carcinoma cells. Moreover, PRIM1 silencing elevated the chemosensitivity of gastric carcinoma cells. By investigating molecular events downstream of the Hippo signaling pathway, we found that PRIM1 was a target gene of the YAP1/TEAD4 transcriptional regulatory complex. PRIM1 represents a novel target for gastric carcinoma therapeutic approaches.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Operating Room, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Lin Guo
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
36
|
Yu S, Feng W, Zeng J, Zhou S, Peng Y, Zhang P. GALNT12 promotes fibrosarcoma growth by accelerating YAP1 nuclear localization. Oncol Lett 2023; 26:543. [PMID: 38020290 PMCID: PMC10660188 DOI: 10.3892/ol.2023.14131] [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: 02/22/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Fibrosarcoma is a highly malignant type of soft tissue sarcoma that currently lacks effective treatment options. Polypeptide N-acetylgalactosaminyltransferase 12 (GALNT12) belongs to the uridine diphosphate N-acetylgalactosamine gene family, which is involved in numerous biological processes of diseases, such as tumor progression. Its upregulated expression is closely associated with the development of colorectal cancer. However, research on the role of GALNT12 in fibrosarcoma is currently limited. The present study aimed to assess the expression and biological function of GALNT12 in fibrosarcoma. Patient data and tissue samples were collected and public datasets were obtained from the Gene Expression Omnibus (GSE24369 and GSE21124). Immunofluorescence assays were performed to observe the cellular localization of GALNT12. GALNT12 expression was measured using reverse transcription-quantitative PCR, western blotting and immunohistochemistry. Small interfering RNAs were constructed to knock down GALNT12 expression in HT-1080 cells. Cell Counting Kit-8 and EdU assays were used to assess fibrosarcoma cell proliferation. Wound healing and Transwell assays were used to detect migration. Gene set enrichment analysis was performed to identify key pathways. Paired and unpaired Student's t-test, Fisher's exact test and one-way ANOVA (followed by Tukey's Honest Significant Difference test) were used to analyze the data. It was demonstrated that GALNT12 expression was upregulated in both fibrosarcoma cell lines and tissue samples and predicted poor patient prognosis. In vitro experiments demonstrated that high GALNT12 expression levels significantly increased HT-1080 cell proliferation and migration. Furthermore, it was demonstrated that high GALNT12 expression levels were closely associated with the yes1 associated transcriptional regulator (YAP1) signaling pathway. Knockdown of GALNT12 inhibited YAP1 nuclear translocation, which affected activation of key downstream genes including AMOTL2, BIRC5 and CYR61. Therefore, the present study demonstrated that GALNT12 promoted fibrosarcoma progression. GALNT12 could be a potential biomarker for this disease and may potentially provide new ideas for targeted therapy of fibrosarcoma in the future.
Collapse
Affiliation(s)
- Site Yu
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jizhang Zeng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Situo Zhou
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yinghua Peng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Pihong Zhang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
37
|
Zhuang X, Liu T, Wei L, Gao Y, Gao J. RNA sequencing reveals the mechanism of FTO in inhibiting inflammation and excessive proliferation of lipopolysaccharide-induced human glomerular mesangial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3835-3846. [PMID: 37358794 DOI: 10.1007/s00210-023-02570-x] [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: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Chronic glomerulonephritis (CGN) is a leading cause of end-stage renal disease in China; thus, there is an urgent need for effective therapeutic targets and strategies for CGN treatment. However, studies on CGN pathogenesis are limited. In this study, we found that the fat mass and obesity-associated protein (FTO) was significantly decreased in the lipopolysaccharide (LPS)-induced human glomerular mesangial cells (HGMCs) (P < 0.01) and kidney tissues of CGN patients (P < 0.05). Moreover, double-labeling immunofluorescence and flow cytometry assays demonstrated that the overexpression of FTO could inhibit inflammation and excessive proliferation of HGMCs. Furthermore, RNA-sequencing (RNA-seq) and real-time quantitative polymerase chain reaction (RT-qPCR) analyses revealed that FTO overexpression induced differential expression of 269 genes (absolute fold change ≥ 2 and P-value < 0.05), including 143 upregulated and 126 downregulated genes. Further functional analysis of these differentially expressed genes by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses suggested that FTO possibly mediates its inhibitory function by regulating the mammalian target of rapamycin (mTOR) signaling pathway and substance metabolism. Lastly, analysis of the PPI network and further identification of the top 10 hub genes (RPS15, RPS18, RPL18A, GNB2L1, RPL19, EEF1A1, RPS25, FAU, UBA52, and RPS6) indicated that FTO mediates its function by affecting the ribosomal proteins. Therefore, in this study, we elucidated the important role of FTO in the regulation of inflammation and excessive proliferation of HGMCs, suggesting FTO administration as a suitable therapeutic intervention for CGN.
Collapse
Affiliation(s)
- Xingxing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University, No. 64 North Chaohu Road, Chaohu, 238000, Anhui, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Tao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Liangbing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Yachen Gao
- Department of Nephropathy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China
| | - Jiarong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, No. 117 Meishan Road, Hefei, 230012, Anhui, China.
| |
Collapse
|
38
|
Shi H, Zou Y, Zhong W, Li Z, Wang X, Yin Y, Li D, Liu Y, Li M. Complex roles of Hippo-YAP/TAZ signaling in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:15311-15322. [PMID: 37608027 DOI: 10.1007/s00432-023-05272-2] [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: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND The Hippo signaling pathway is an evolutionarily conserved signaling module that controls organ size in different species, and the disorder of the Hippo pathway can induce liver cancer in organisms, especially hepatocellular carcinoma (HCC). The exact mechanism that causes cancer is still unknown. Recent studies have shown that it is a classical kinase cascade that phosphorylates the Mst1/2-sav1 complex and activates the phosphorylation of the Lats1/2-mob1A/B complex for inactivating Yap and Taz. These kinases and scaffolds are regarded as primary regulators of the Hippo pathway, and help in activating a variety of carcinogenic processes. Among them, Yap/Taz is seen to be the main effector molecule, which is downstream of the Hippo pathway, and its abnormal activation is related to a variety of human cancers including liver cancer. Currently, since Yap/Taz plays a variety of roles in cancer promotion and tumor regeneration, the Hippo pathway has emerged as an attractive target in recent drug development research. METHODS We collect and review relevant literature in web of Science and Pubmed. CONCLUSION This review highlights the important roles of Yap/Taz in activating Hippo pathway in liver cancer. The recent findings on the crosstalks between the Hippo and other cancer associated pathways and moleculars are also discussed. In this review, we summarized and discussed recent breakthroughs in our understanding of how key components of the Hippo-YAP/TAZ pathway influence the hepatocellular carcinoma, including their effects on tumor occurrence and development, their roles in regulating metastasis, and their function in chemotherapy resistance. Further, the molecular mechanism and roles in regulating cross talk between Hippo-YAP/TAZ pathway and other cancer-associated pathways or oncogenes/cancer suppressor genes were summarized and discussed. More, many other inducers and inhibitors of this signaling cascade and available experimental therapies against the YAP/TAZ/TEAD axis were discussed. Targeting this pathway for cancer therapy may have great significance in the treatment of hepatocellular carcinoma. Graphical summary of the complex role of Hippo-YAP/TAZ signaling in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hewen Shi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Zou
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Weiwei Zhong
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhaoying Li
- Traditional Chinese Medicine Research Center, Shandong Public Health Clinical Center, Jinan, 250102, People's Republic of China
| | - Xiaoxue Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| | - Minjing Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
39
|
El Yousfi Y, Mora-Molina R, López-Rivas A, Yerbes R. Role of the YAP/TAZ-TEAD Transcriptional Complex in the Metabolic Control of TRAIL Sensitivity by the Mevalonate Pathway in Cancer Cells. Cells 2023; 12:2370. [PMID: 37830584 PMCID: PMC10571597 DOI: 10.3390/cells12192370] [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: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Different studies have reported that inhibiting the mevalonate pathway with statins may increase the sensitivity of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), although the signaling mechanism leading to this sensitization remains largely unknown. We investigated the role of the YAP (Yes-associated protein)/TAZ (transcriptional co-activator with PDZ-binding motif)-TEAD (TEA/ATTS domain) transcriptional complex in the metabolic control of TRAIL sensitivity by the mevalonate pathway. We show that depleting nuclear YAP/TAZ in tumor cells, either via treatment with statins or by silencing YAP/TAZ expression with siRNAs, facilitates the activation of apoptosis by TRAIL. Furthermore, the blockage of TEAD transcriptional activity either pharmacologically or through the ectopic expression of a disruptor of the YAP/TAZ interaction with TEAD transcription factors, overcomes the resistance of tumor cells to the induction of apoptosis by TRAIL. Our results show that the mevalonate pathway controls cellular the FLICE-inhibitory protein (cFLIP) expression in tumor cells. Importantly, inhibiting the YAP/TAZ-TEAD signaling pathway induces cFLIP down-regulation, leading to a marked sensitization of tumor cells to apoptosis induction by TRAIL. Our data suggest that a combined strategy of targeting TEAD activity and selectively activating apoptosis signaling by agonists of apoptotic TRAIL receptors could be explored as a potential therapeutic approach in cancer treatment.
Collapse
Affiliation(s)
- Younes El Yousfi
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Rocío Mora-Molina
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Abelardo López-Rivas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
| | - Rosario Yerbes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, 41092 Seville, Spain; (Y.E.Y.); (R.M.-M.); (A.L.-R.)
- Medical Physiology and Biophysics Department, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS) (Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla), 41013 Seville, Spain
| |
Collapse
|
40
|
Cai S, Wang X, Xu R, Liang Z, Zhu Q, Chen M, Lin Z, Li C, Duo T, Tong X, Li E, He Z, Liu X, Chen Y, Mo D. KLF4 regulates skeletal muscle development and regeneration by directly targeting P57 and Myomixer. Cell Death Dis 2023; 14:612. [PMID: 37723138 PMCID: PMC10507053 DOI: 10.1038/s41419-023-06136-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: 05/25/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023]
Abstract
Krüppel-like factor 4 (KLF4) is an evolutionarily conserved zinc finger-containing transcription factor that regulates diverse cellular processes such as cell proliferation, apoptosis, and differentiation. Our previous study showed that KLF4 expression is upregulated in skeletal muscle ontogeny during embryonic development in pigs, suggesting its importance for skeletal muscle development and muscle function. We revealed here that KLF4 plays a critical role in skeletal muscle development and regeneration. Specific knockout of KLF4 in skeletal muscle impaired muscle formation further affecting physical activity and also defected skeletal muscle regeneration. In vitro, KLF4 was highly expressed in proliferating myoblasts and early differentiated cells. KLF4 knockdown promoted myoblast proliferation and inhibited myoblast fusion, while its overexpression showed opposite results. Mechanically, in proliferating myoblasts, KLF4 inhibits myoblast proliferation through regulating cell cycle arrest protein P57 by directly targeting its promoter; while in differentiated myoblasts, KLF4 promotes myoblast fusion by transcriptionally activating Myomixer. Our study provides mechanistic information for skeletal muscle development, reduced muscle strength and impaired regeneration after injury and unveiling the mechanism of KLF4 in myogenic regulation.
Collapse
Affiliation(s)
- Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Xiaoyu Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Rong Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Meilin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Zhuhu Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Chenggan Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Xian Tong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Enru Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
41
|
Rogg M, Maier JI, Helmstädter M, Sammarco A, Kliewe F, Kretz O, Weißer L, Van Wymersch C, Findeisen K, Koessinger AL, Tsoy O, Baumbach J, Grabbert M, Werner M, Huber TB, Endlich N, Schilling O, Schell C. A YAP/TAZ-ARHGAP29-RhoA Signaling Axis Regulates Podocyte Protrusions and Integrin Adhesions. Cells 2023; 12:1795. [PMID: 37443829 PMCID: PMC10340513 DOI: 10.3390/cells12131795] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Glomerular disease due to podocyte malfunction is a major factor in the pathogenesis of chronic kidney disease. Identification of podocyte-specific signaling pathways is therefore a prerequisite to characterizing relevant disease pathways and developing novel treatment approaches. Here, we employed loss of function studies for EPB41L5 (Yurt) as a central podocyte gene to generate a cell type-specific disease model. Loss of Yurt in fly nephrocytes caused protein uptake and slit diaphragm defects. Transcriptomic and proteomic analysis of human EPB41L5 knockout podocytes demonstrated impaired mechanotransduction via the YAP/TAZ signaling pathway. Further analysis of specific inhibition of the YAP/TAZ-TEAD transcription factor complex by TEADi led to the identification of ARGHAP29 as an EPB41L5 and YAP/TAZ-dependently expressed podocyte RhoGAP. Knockdown of ARHGAP29 caused increased RhoA activation, defective lamellipodia formation, and increased maturation of integrin adhesion complexes, explaining similar phenotypes caused by loss of EPB41L5 and TEADi expression in podocytes. Detection of increased levels of ARHGAP29 in early disease stages of human glomerular disease implies a novel negative feedback loop for mechanotransductive RhoA-YAP/TAZ signaling in podocyte physiology and disease.
Collapse
Affiliation(s)
- Manuel Rogg
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jasmin I. Maier
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alena Sammarco
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489 Greifswald, Germany (N.E.)
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lisa Weißer
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Clara Van Wymersch
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Karla Findeisen
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Anna L. Koessinger
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany; (O.T.)
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany; (O.T.)
- Department of Mathematics and Computer Science, University of Southern Denmark, 5230 Odense, Denmark
| | - Markus Grabbert
- Department of Urology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Martin Werner
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489 Greifswald, Germany (N.E.)
| | - Oliver Schilling
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79106 Freiburg, Germany
| | - Christoph Schell
- Institute of Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
42
|
Hile GA, Coit P, Xu B, Victory AM, Gharaee-Kermani M, Estadt SN, Maz MP, Martens JWS, Wasikowski R, Dobry C, Tsoi LC, Iglesias-Bartolome R, Berthier CC, Billi AC, Gudjonsson JE, Sawalha AH, Kahlenberg JM. Regulation of Photosensitivity by the Hippo Pathway in Lupus Skin. Arthritis Rheumatol 2023; 75:1216-1228. [PMID: 36704840 PMCID: PMC10313771 DOI: 10.1002/art.42460] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/05/2022] [Accepted: 01/24/2023] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Photosensitivity is one of the most common manifestations of systemic lupus erythematosus (SLE), yet its pathogenesis is not well understood. The normal-appearing epidermis of patients with SLE exhibits increased ultraviolet B (UVB)-driven cell death that persists in cell culture. Here, we investigated the role of epigenetic modification and Hippo signaling in enhanced UVB-induced apoptosis seen in SLE keratinocytes. METHODS We analyzed DNA methylation in cultured keratinocytes from SLE patients compared to keratinocytes from healthy controls (n = 6/group). Protein expression was validated in cultured keratinocytes using immunoblotting and immunofluorescence. An immortalized keratinocyte line overexpressing WWC1 was generated via lentiviral vector. WWC1-driven changes were inhibited using a large tumor suppressor kinase 1/2 (LATS1/2) inhibitor (TRULI) and small interfering RNA (siRNA). The interaction between the Yes-associated protein (YAP) and the transcriptional enhancer associate domain (TEAD) was inhibited by overexpression of an N/TERT cell line expressing a tetracycline-inducible green fluorescent protein-tagged protein that inhibits YAP-TEAD binding (TEADi). Apoptosis was assessed using cleaved caspase 3/7 and TUNEL staining. RESULTS Hippo signaling was the top differentially methylated pathway in SLE versus control keratinocytes. SLE keratinocytes (n = 6) showed significant hypomethylation (Δβ = -0.153) and thus overexpression of the Hippo regulator WWC1 (P = 0.002). WWC1 overexpression increased LATS1/2 kinase activation, leading to YAP cytoplasmic retention and altered proapoptotic transcription in SLE keratinocytes. Accordingly, UVB-mediated apoptosis in keratinocytes could be enhanced by WWC1 overexpression or YAP-TEAD inhibition, mimicking SLE keratinocytes. Importantly, inhibition of LATS1/2 with either the chemical inhibitor TRULI or siRNA effectively eliminated enhanced UVB-apoptosis in SLE keratinocytes. CONCLUSION Our work unravels a novel driver of photosensitivity in SLE: overactive Hippo signaling in SLE keratinocytes restricts YAP transcriptional activity, leading to shifts that promote UVB apoptosis.
Collapse
Affiliation(s)
- Grace A. Hile
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | - Patrick Coit
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh; Pittsburgh, USA
| | - Bin Xu
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| | | | - Mehrnaz Gharaee-Kermani
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| | - Shannon N. Estadt
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
| | - Mitra P. Maz
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
| | | | - Rachael Wasikowski
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
| | - Craig Dobry
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
- Department of Biostatistics, University of Michigan; Ann Arbor, MI 48109, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, USA
| | - Celine C. Berthier
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
- Department of Biostatistics, University of Michigan; Ann Arbor, MI 48109, USA
| | - Allison C. Billi
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | | | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh; Pittsburgh, USA
| | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| |
Collapse
|
43
|
Singh DR, Nelson SE, Pawelski AS, Kansra AS, Fogarty SA, Bristol JA, Ohashi M, Johannsen EC, Kenney SC. Epstein-Barr virus LMP1 protein promotes proliferation and inhibits differentiation of epithelial cells via activation of YAP and TAZ. Proc Natl Acad Sci U S A 2023; 120:e2219755120. [PMID: 37155846 PMCID: PMC10193989 DOI: 10.1073/pnas.2219755120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/28/2023] [Indexed: 05/10/2023] Open
Abstract
Latent Epstein-Barr virus (EBV) infection promotes undifferentiated nasopharyngeal carcinomas (NPCs) in humans, but the mechanism(s) for this effect has been difficult to study because EBV cannot transform normal epithelial cells in vitro and the EBV genome is often lost when NPC cells are grown in culture. Here we show that the latent EBV protein, LMP1 (Latent membrane protein 1), induces cellular proliferation and inhibits spontaneous differentiation of telomerase-immortalized normal oral keratinocytes (NOKs) in growth factor-deficient conditions by increasing the activity of the Hippo pathway effectors, YAP (Yes-associated protein) and TAZ (Transcriptional coactivator with PDZ-binding motif). We demonstrate that LMP1 enhances YAP and TAZ activity in NOKs both by decreasing Hippo pathway-mediated serine phosphorylation of YAP and TAZ and increasing Src kinase-mediated Y357 phosphorylation of YAP. Furthermore, knockdown of YAP and TAZ is sufficient to reduce proliferation and promote differentiation in EBV-infected NOKs. We find that YAP and TAZ are also required for LMP1-induced epithelial-to-mesenchymal transition. Importantly, we demonstrate that ibrutinib (an FDA-approved BTK inhibitor that blocks YAP and TAZ activity through an off-target effect) restores spontaneous differentiation and inhibits proliferation of EBV-infected NOKs at clinically relevant doses. These results suggest that LMP1-induced YAP and TAZ activity contributes to the development of NPC.
Collapse
Affiliation(s)
- Deo R. Singh
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Scott E. Nelson
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Abigail S. Pawelski
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Alisha S. Kansra
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Stuart A. Fogarty
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Jillian A. Bristol
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Makoto Ohashi
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Eric C. Johannsen
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| | - Shannon C. Kenney
- Department of Oncology, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, WIMR II, Madison, WI53705
| |
Collapse
|
44
|
Feng Z, Tao S, Huang Z, Zheng B, Kong X, Xiang Y, Zhang Q, Song H, Xu Z, Wei X, Zhao F, Chen J. The deubiquitinase UCHL1 negatively controls osteoclastogenesis by regulating TAZ/NFATC1 signalling. Int J Biol Sci 2023; 19:2319-2332. [PMID: 37215988 PMCID: PMC10197889 DOI: 10.7150/ijbs.82152] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 04/17/2023] [Indexed: 05/24/2023] Open
Abstract
The ubiquitin‒proteasome system (UPS) plays a key role in maintaining protein homeostasis and bone remodelling. However, the role of deubiquitinating enzymes (DUBs) in bone resorption is still not well defined. Here, we identified the deubiquitinase ubiquitin C-terminal hydrolase 1 (UCHL1) as a negative regulator of osteoclastogenesis by using the GEO database, proteomic analysis, and RNAi. Osteoclast-specific UCHL1 conditional knockout mice exhibited a severe osteoporosis phenotype in an ovariectomized model. Mechanistically, UCHL1 deubiquitinated and stabilized the transcriptional coactivator with PDZ-binding motif (TAZ) at the K46 residue, thereby inhibiting osteoclastogenesis. The TAZ protein underwent K48-linked polyubiquitination, which was degraded by UCHL1. As a substrate of UCHL1, TAZ regulates NFATC1 through a nontranscriptional coactivator function by competing with calcineurin A (CNA) for binding to NFATC1, which inhibits NFATC1 dephosphorylation and nuclear transport to impede osteoclastogenesis. Moreover, overexpression of UCHL1 locally alleviated acute and chronic bone loss. These findings suggest that activating UCHL1 may serve as a novel therapeutic approach targeting bone loss in various bone pathological states.
Collapse
Affiliation(s)
- Zhenhua Feng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Siyue Tao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhaobo Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Bingjie Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangxi Kong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Yufeng Xiang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Qibin Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Haixin Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhikun Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiaoan Wei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Fengdong Zhao
- ✉ Corresponding authors: Jian Chen () and Fengdong Zhao ()
| | - Jian Chen
- ✉ Corresponding authors: Jian Chen () and Fengdong Zhao ()
| |
Collapse
|
45
|
Li Y, Xiong Z, Jiang Y, Zhou H, Yi L, Hu Y, Zhai X, Liu J, Tian F, Chen Y. Klf4 deficiency exacerbates myocardial ischemia/reperfusion injury in mice via enhancing ROCK1/DRP1 pathway-dependent mitochondrial fission. J Mol Cell Cardiol 2023; 174:115-132. [PMID: 36509022 DOI: 10.1016/j.yjmcc.2022.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022]
Abstract
RATIONAL Excessive mitochondrial fission is considered key process involved in myocardial ischemia/reperfusion (I/R) injury. However, the upstream mechanism remains largely unclear. Decreased level of Kruppel Like Factor 4 (KLF4) has been implicated in the pathogenesis of mitochondrial dysfunction and heart's adaption to stress. However, the role of Klf4 in I/R process is not fully elucidated. This study aims to investigate how Klf4 regulates mitochondrial dynamics and further clarify its underlying mechanism during cardiac I/R injury. METHODS Loss-of-function and gain-of-function strategies were applied to investigate the role of Klf4 in cardiac I/R injury via genetic ablation or intra-myocardial adenovirus injection. Mitochondrial dynamics was analyzed by confocal microscopy in vitro and transmission electron microscopy in vivo. Chromatin immunoprecipitation and luciferase reporter assay were performed to explore the underlying mechanisms. RESULTS KLF4 was downregulated in I/R heart. Cardiac-specific Klf4 knockout significantly exacerbated cardiac dysfunction in I/R mice. Mechanistically, Klf4 deficiency aggravated mitochondrial apoptosis, reduced ATP generation and boosted ROS overproduction via enhancing DRP1-dependent mitochondrial fission. ROCK1 was identified as a kinase regulating DRP1 activity at Ser616. Klf4 deficiency upregulated the expression of ROCK1 at transcriptional level, thus increasing S616-DRP1-mediated mitochondrial fission during I/R. Finally, reconstitution of Klf4 inhibited mitochondrial fission, restored mitochondrial function and alleviated I/R injury. CONCLUSION Our study provides the first evidence that Klf4 deficiency exacerbates myocardial I/R injury through regulating ROCK1 expression at transcriptional level to induce DRP1-mediated mitochondrial fission. Targeting mitochondrial dynamics by restoring Klf4 might be potentially cardio-protective strategies attenuating I/R injury.
Collapse
Affiliation(s)
- Yueyang Li
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhenyu Xiong
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yufan Jiang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Zhou
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Yi
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingyun Hu
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaofeng Zhai
- The Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jie Liu
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Feng Tian
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
46
|
Wang T, Li K, Liu H, Luo E. Focusing on Hippo Pathway in Stem Cells of Oral Origin, Enamel Formation and Periodontium Regeneration. Organogenesis 2022; 18:2082236. [PMID: 35786361 PMCID: PMC9897286 DOI: 10.1080/15476278.2022.2082236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hippo pathway is a cellular regulatory pathway composed of core molecules such as MST1/2, LATS1/2, SAV1, MOB1A/B and downstream YAP/TAZ. Fully involved in regulating cell proliferation, differentiation, migration and apoptosis, the Hippo pathway is critical in regulating stem cells of oral origin, for instance, DPSCs and PDLSCs, enamel formation and periodontium regeneration. Here, we summarized the Hippo pathway involved in these progresses and concluded crosstalks of the Hippo pathway with BCL-2, ERK1/2, ROCK, TGF-β/BMP and Wnt/β-catenin pathways, hoping to provide foundation for further clinical therapy.
Collapse
Affiliation(s)
- Tianyi Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kehan Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China,CONTACT En Luo State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu, Sichuan610041, China
| |
Collapse
|
47
|
Jaiswal A, Singh R. Loss of Epidermal Homeostasis Underlies the Development of Squamous Cell Carcinoma. Stem Cell Rev Rep 2022; 19:667-679. [PMID: 36520410 DOI: 10.1007/s12015-022-10486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Squamous cell carcinoma (SCC) is one of the most common skin cancers. To develop targeted therapies for SCC, a comprehensive understanding of the disease through a systems approach is required. Here, we have collated and analyzed the literature on SCC and pathways that maintain skin homeostasis. Since, the loss of the Notch and the overactivation of the Wnt pathways in the epidermis cause SCC, we focused on these two pathways. We found that the two pathways are critical in maintaining epidermal homeostasis. Further, we found that the cancer stem cell (CSC) marker CD44 causes the transcription of SOX2, another CSC marker of SCC, activates the Wnt pathway, and blocks the Notch pathway. Similarly, the Wnt pathway causes the transcription of CD44 and SOX2 and blocks the Notch pathway. In this paper, we have discussed how the notch and the Wnt pathways affect epidermal homeostasis and the three CSCs (CD44, SOX2, and LGR6) affect the two pathways, linking the CSCs with epidermal homeostasis.
Collapse
|
48
|
Wang Z, Liu M, Lei H, Xiao S, Zheng Y. TEAD1 Silencing Regulates Cell Proliferation and Resistance to 5-Fluorouracil in Cutaneous Squamous Cell Carcinoma. Clin Cosmet Investig Dermatol 2022; 15:2685-2692. [PMID: 36536757 PMCID: PMC9759115 DOI: 10.2147/ccid.s386547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2023]
Abstract
PURPOSE Cutaneous squamous cell carcinoma (cSCC) is a skin malignant tumor account for approximately one-third of all nonmelanoma skin cancers. Studies have shown that TEA domain transcription factor 1 (TEAD1) is discovered to be involved in the pathogenesis of some human cancers, but to our knowledge its role in cSCC has not been reported. PATIENTS AND METHODS Samples from 16 cSCC patients and 27 healthy individuals were obtained for immunohistochemical staining of TEAD1. The expressions of TEAD1 in SCL-1, HSC-1 cells compared with the primary neonatal human epithelial keratinocytes were detected by Western blot and RT-qPCR. Proliferation and cell cycle of TEAD1 knockdown in cSCC cell lines were examined by MTT and flow cytometry analysis. Annexin V/PI and JC-1 staining were used to determine the cell apoptosis. RESULTS The expression of TEAD1 decreased significantly in cSCC compared to its expression in normal skin tissues and cell lines. Down-regulation of TEAD1 in cSCC cell lines promoted cell growth via regulation of the G2/M progression. Additionally, silence of TEAD1 also protected cells against 5-Fluorouracil-induced apoptosis and decreased the expression of apoptosis-related protein (p53). CONCLUSION Our results suggested that TEAD1 expression is down-regulated and functioned as a tumor suppressor in cSCC and that it may serve as a biomarker or therapeutic target of cSCC.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Dermatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Meng Liu
- Department of Dermatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Hao Lei
- Department of Dermatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Shengxiang Xiao
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yan Zheng
- Department of Dermatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
49
|
Fu M, Hu Y, Lan T, Guan KL, Luo T, Luo M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther 2022; 7:376. [PMID: 36347846 PMCID: PMC9643504 DOI: 10.1038/s41392-022-01191-9] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022] Open
Abstract
As an evolutionarily conserved signalling network, the Hippo pathway plays a crucial role in the regulation of numerous biological processes. Thus, substantial efforts have been made to understand the upstream signals that influence the activity of the Hippo pathway, as well as its physiological functions, such as cell proliferation and differentiation, organ growth, embryogenesis, and tissue regeneration/wound healing. However, dysregulation of the Hippo pathway can cause a variety of diseases, including cancer, eye diseases, cardiac diseases, pulmonary diseases, renal diseases, hepatic diseases, and immune dysfunction. Therefore, therapeutic strategies that target dysregulated Hippo components might be promising approaches for the treatment of a wide spectrum of diseases. Here, we review the key components and upstream signals of the Hippo pathway, as well as the critical physiological functions controlled by the Hippo pathway. Additionally, diseases associated with alterations in the Hippo pathway and potential therapies targeting Hippo components will be discussed.
Collapse
Affiliation(s)
- Minyang Fu
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Tianxia Lan
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Ting Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| | - Min Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| |
Collapse
|
50
|
Wang J, Li S, Zhang X, Zhu N, Yiminniyaze R, Dong L, Li C, Gulinuer W, Xia J, Li J, Zhou D, Liu X, Zhang Y, Zhang Y, Li S. Protein tyrosine phosphatase PTPL1 suppresses lung cancer through Src/ERK/YAP1 signaling. Thorac Cancer 2022; 13:3042-3051. [PMID: 36193770 PMCID: PMC9626330 DOI: 10.1111/1759-7714.14657] [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: 05/31/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND To reveal the function of protein tyrosine phosphatase-L1 (PTPL1) in lung adenocarcinoma. METHODS Lung cancer cell lines were transfected with short hairpin RNA against PTPL1 (shPTPL1 group) or negative control (shmock group). Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting were used to verify the transfection efficacy. Cell proliferation was analyzed by ethynyldeoxyuridine (EdU), Cell counting kit 8 (CCK8), and colony formation assay after PTPL1 or PTPL1 and yes-associated protein (YAP1) knockdown. The effect of PTPL1 on tumor growth was examined in a xenograft lung cancer model. RESULTS PTPL1 was downregulated in various types of lung cancer cell lines. The EdU, CCK8, colony formation assays and investigation using a xenograft lung cancer model indicated that PTPL1 knockdown increased the proliferation of lung cancer cells. Mechanistically, PTPL1 knockdown induced the activation of the Proto-oncogene tyrosine-protein kinase SRC (Src)/Extracellular regulated MAP kinase (ERK) pathway and thereby promoted yes-associated protein (YAP1) nuclear translocation and activation. CONCLUSIONS In our study, PTPL1 played a crucial suppressive role in the pathogenesis of lung cancer potentially through counteracting the Src/ERK/YAP1 pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Shuanghui Li
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiujuan Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Ning Zhu
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Ruzetuoheti Yiminniyaze
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Liang Dong
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Chengwei Li
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Wumaier Gulinuer
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Jingwen Xia
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Jing Li
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Daibing Zhou
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Xinning Liu
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Youzhi Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Shengqing Li
- Department of Pulmonary and Critical Care Medicine, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|