1
|
Lao Z, Chen X, Pan B, Fang B, Yang W, Qian Y. Pharmacological regulators of Hippo pathway: Advances and challenges of drug development. FASEB J 2025; 39:e70438. [PMID: 40100056 DOI: 10.1096/fj.202401895rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
The Hippo signaling pathway is crucial in regulating organ size, tumor progression, tissue regeneration, and bone homeostasis. Inactivation of the Hippo pathway results in the nuclear translocation and activation of YAP/TAZ. This activation not only promotes tumor progression but also enhances tissue regeneration, wound healing, and maintenance of bone stability Although its discovery occurred over two decades ago, developing effective inhibitors or activators for the Hippo pathway remains challenging. Recently, however, the pace of advancements in developing Hippo signaling-related agonists and antagonists has accelerated, with some drugs that target TEAD advancing to clinical trials and showing promise for treating related diseases. This review summarizes the progress in research on Hippo signaling-related agonists and inhibitors, offering an in-depth analysis of their regulatory mechanisms, pharmacological properties, and potential in vivo applications.
Collapse
Affiliation(s)
- Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Fang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
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
|
3
|
Wang K, Lu J, Song C, Qiao M, Li Y, Chang M, Bao H, Qiu Y, Qian B. Extracellular Vesicles Derived from Ligament Tissue Transport Interleukin-17A to Mediate Ligament-To-Bone Crosstalk in Ankylosing Spondylitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406876. [PMID: 39308181 PMCID: PMC11633500 DOI: 10.1002/advs.202406876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/09/2024] [Indexed: 10/12/2024]
Abstract
Pathological new bone formation is a critical feature of the progression of ankylosing spondylitis (AS), and spine ankylosis is a distinctive feature of this condition. Ligaments are the primary regions of pathological new bone formation in AS. Here, it is demonstrated that ligament tissue-derived extracellular vesicles (EVs) and their interleukin-17A (IL-17A) cargo mediate the communication between the tissue and other cells. The investigation revealed that IL-17A in EVs can activate the JAK-STAT3 pathway, thereby stimulating the expression of MMP14 in AS ligament. Overexpression of MMP14 can lead to changes in the cytoskeleton and mechanical signaling of mesenchymal stem cells and other cells. These alterations in cellular cytoskeleton and mechanical signaling at ligament sites in patients with AS or in stem cells treated with EVs can result in pathological new bone formation. Finally, inhibiting IL-17A activity and EV endocytosis effectively controlled inflammation and pathological new bone formation. Overall, these data suggest that ligament-derived EVs and the enclosed IL-17A have a potential role in driving pathological new bone formation in AS, and targeting EVs may therefore emerge as a novel approach to delaying ectopic ossification in AS.
Collapse
Affiliation(s)
- Kaiyang Wang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Jingshun Lu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Chenyu Song
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Mu Qiao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yao Li
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Menghan Chang
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Hongda Bao
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Yong Qiu
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| | - Bang‐Ping Qian
- Division of Spine SurgeryDepartment of Orthopedic SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityZhongshan Road 321Nanjing210008China
| |
Collapse
|
4
|
Huang C, Zhong Q, Lian W, Kang T, Hu J, Lei M. Ankrd1 inhibits the FAK/Rho-GTPase/F-actin pathway by downregulating ITGA6 transcriptional to regulate myoblast functions. J Cell Physiol 2024; 239:e31359. [PMID: 38988048 DOI: 10.1002/jcp.31359] [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: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
Skeletal muscle constitutes the largest percentage of tissue in the animal body and plays a pivotal role in the development of normal life activities in the organism. However, the regulation mechanism of skeletal muscle growth and development remains largely unclear. This study investigated the effects of Ankrd1 on the proliferation and differentiation of C2C12 myoblasts. Here, we identified Ankrd1 as a potential regulator of muscle cell development, and found that Ankrd1 knockdown resulted in the proliferation ability decrease but the differentiation level increase of C2C12 cells. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyzes as well as RNA-seq results showed that Ankrd1 knockdown activated focal adhesion kinase (FAK)/F-actin signal pathway with most genes significantly enriched in this pathway upregulated. The integrin subunit Itga6 promoter activity is increased when Ankrd1 knockdown, as demonstrated by a dual-luciferase reporter assay. This study revealed the molecular mechanism by which Ankrd1 knockdown enhanced FAK phosphorylation activity through the alteration of integrin subunit levels, thus activating FAK/Rho-GTPase/F-actin signal pathway, eventually promoting myoblast differentiation. Our data suggested that Ankrd1 might serve as a potential regulator of muscle cell development. Our findings provide new insights into skeletal muscle growth and development and valuable references for further study of human muscle-related diseases.
Collapse
Affiliation(s)
- Cheng Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiqi Zhong
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Weisi Lian
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Tingting Kang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Jinling Hu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Minggang Lei
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education, Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
- National Engineering Research Center for Livestock, Huazhong Agricultural University, Wuhan, Hubei, China
- Department of Pig Production, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| |
Collapse
|
5
|
Meng X, Zhu Y, Tan H, Daraqel B, Ming Y, Li X, Yang G, He X, Song J, Zheng L. The cytoskeleton dynamics-dependent LINC complex in periodontal ligament stem cells transmits mechanical stress to the nuclear envelope and promotes YAP nuclear translocation. Stem Cell Res Ther 2024; 15:284. [PMID: 39243052 PMCID: PMC11380336 DOI: 10.1186/s13287-024-03884-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/14/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Periodontal ligament stem cells (PDLSCs) are important seed cells in tissue engineering and clinical applications. They are the priority receptor cells for sensing various mechanical stresses. Yes-associated protein (YAP) is a recognized mechanically sensitive transcription factor. However, the role of YAP in regulating the fate of PDLSCs under tension stress (TS) and its underlying mechanism is still unclear. METHODS The effects of TS on the morphology and fate of PDLSCs were investigated using fluorescence staining, transmission electron microscopy, flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR). Then qRT-PCR, western blotting, immunofluorescence staining and gene knockdown experiments were performed to investigate the expression and distribution of YAP and its correlation with PDLSCs proliferation. The effects of cytoskeleton dynamics on YAP nuclear translocation were subsequently explored by adding cytoskeleton inhibitors. The effect of cytoskeleton dynamics on the expression of the LINC complex was proved through qRT-PCR and western blotting. After destroying the LINC complex by adenovirus, the effects of the LINC complex on YAP nuclear translocation and PDLSCs proliferation were investigated. Mitochondria-related detections were then performed to explore the role of mitochondria in YAP nuclear translocation. Finally, the in vitro results were verified by constructing orthodontic tooth movement models in Sprague-Dawley rats. RESULTS TS enhanced the polymerization and stretching of F-actin, which upregulated the expression of the LINC complex. This further strengthened the pull on the nuclear envelope, enlarged the nuclear pore, and facilitated YAP's nuclear entry, thus enhancing the expression of proliferation-related genes. In this process, mitochondria were transported to the periphery of the nucleus along the reconstructed microtubules. They generated ATP to aid YAP's nuclear translocation and drove F-actin polymerization to a certain degree. When the LINC complex was destroyed, the nuclear translocation of YAP was inhibited, which limited PDLSCs proliferation, impeded periodontal tissue remodeling, and hindered tooth movement. CONCLUSIONS Our study confirmed that appropriate TS could promote PDLSCs proliferation and periodontal tissue remodeling through the mechanically driven F-actin/LINC complex/YAP axis, which could provide theoretical guidance for seed cell expansion and for promoting healthy and effective tooth movement in clinical practice.
Collapse
Affiliation(s)
- Xuehuan Meng
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Ye Zhu
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Hao Tan
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Baraa Daraqel
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
- Oral Health Research and Promotion Unit, Al-Quds University, Jerusalem, Palestine
| | - Ye Ming
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xiang Li
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Guoyin Yang
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Xinyi He
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| | - Leilei Zheng
- College of Stomatology, Chongqing Medical University, Chongqing, 401147, China.
- Chongqing Key Laboratory of Oral Diseases, Chongqing, 401147, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China.
| |
Collapse
|
6
|
Jeong W, Kwon H, Park SK, Lee IS, Jho EH. Retinoic acid-induced protein 14 links mechanical forces to Hippo signaling. EMBO Rep 2024; 25:4033-4061. [PMID: 39160347 PMCID: PMC11387738 DOI: 10.1038/s44319-024-00228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024] Open
Abstract
Cells sense and respond to various mechanical forces from the extracellular matrix primarily by modulating the actin cytoskeleton. Mechanical forces can be translated into biochemical signals in a process called mechanotransduction. Yes-associated protein (YAP) is an effector of Hippo signaling and a mediator of mechanotransduction, but how mechanical forces regulate Hippo signaling is still an open question. We propose that retinoic acid-induced protein 14 (RAI14) responds to mechanical forces and regulates Hippo signaling. RAI14 positively regulates the activity of YAP. RAI14 interacts with NF2, a key component of the Hippo pathway, and the interaction occurs on filamentous actin. When mechanical forces are kept low in cells, NF2 dissociates from RAI14 and filamentous actin, resulting in increased interactions with LATS1 and activation of the Hippo pathway. Clinical data show that tissue stiffness and expression of RAI14 and YAP are upregulated in tumor tissues and that RAI14 is strongly associated with adverse outcome in patients with gastric cancer. Our data suggest that RAI14 links mechanotransduction with Hippo signaling and mediates Hippo-related biological functions such as cancer progression.
Collapse
Affiliation(s)
- Wonyoung Jeong
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Hyeryun Kwon
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - In-Seob Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea.
| |
Collapse
|
7
|
Wu J, Zhang Q, Yang Z, Xu Y, Liu X, Wang X, Peng J, Xiao J, Wang Y, Shang Z, Wang N, Li L, Zhang R, Zhang W, Zhang J, Zeng Z, Wu J. CD248-expressing cancer-associated fibroblasts induce non-small cell lung cancer metastasis via Hippo pathway-mediated extracellular matrix stiffness. J Cell Mol Med 2024; 28:e70025. [PMID: 39164826 PMCID: PMC11335579 DOI: 10.1111/jcmm.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024] Open
Abstract
Metastasis is a crucial stage in tumour progression, and cancer-associated fibroblasts (CAFs) support metastasis through their participation in extracellular matrix (ECM) stiffness. CD248 is a possible biomarker for non-small cell lung cancer (NSCLC)-derived CAFs, but its role in mediating ECM stiffness to promote NSCLC metastasis is unknown. We investigated the significance of CD248+ CAFs in activating the Hippo axis and promoting connective tissue growth factor (CTGF) expression, which affects the stromal collagen I environment and improves ECM stiffness, thereby facilitating NSCLC metastasis. In this study, we found that higher levels of CD248 in CAFs induced the formation of collagen I, which in turn increased extracellular matrix stiffness, thereby enabling NSCLC cell infiltration and migration. Hippo axis activation by CD248+ CAFs induces CTGF expression, which facilitates the formation of the collagen I milieu in the stromal matrix. In a tumour lung metastasis model utilizing fibroblast-specific CD248 gene knockout mice, CD248 gene knockout mice showed a significantly reduced ability to develop tumour lung metastasis compared to that of WT mice. Our findings demonstrate that CD248+ CAFs activate the Hippo pathway, thereby inducing CTGF expression, which in turn facilitates the collagen I milieu of the stromal matrix, which promotes NSCLC metastasis.
Collapse
Affiliation(s)
- Jiangwei Wu
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Qiaoling Zhang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Zeyang Yang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Yujun Xu
- Department of BiologyGuizhou Medical UniversityGuiyangChina
| | - Xinlei Liu
- Guizhou Prenatal Diagnsis CenterThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xuanying Wang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Jiangying Peng
- Department of Pharmaceutical AnalysisZunyi Medical UniversityZunyiChina
| | - Jing Xiao
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Yun Wang
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Zhenling Shang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Nianxue Wang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
| | - Long Li
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Department of Thoracic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui Zhang
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Wei Zhang
- Department of Biochemistry and Molecular BiologyJilin Medical UniversityJilinChina
| | - Jian Zhang
- Department of Thoracic SurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zhu Zeng
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
| | - Jieheng Wu
- Department of ImmunologyGuizhou Medical UniversityGuiyangChina
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical EngineeringGuizhou Medical UniversityGuiyangChina
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical SciencesGuizhou Medical UniversityGuiyangChina
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anChina
- Tumor Immunotherapy Technology Engineering Research CenterGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
8
|
Wang Y, Yu FX. Angiomotin family proteins in the Hippo signaling pathway. Bioessays 2024; 46:e2400076. [PMID: 38760875 DOI: 10.1002/bies.202400076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
The Motin family proteins (Motins) are a class of scaffolding proteins consisting of Angiomotin (AMOT), AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). Motins play a pivotal role in angiogenesis, tumorigenesis, and neurogenesis by modulating multiple cellular signaling pathways. Recent findings indicate that Motins are components of the Hippo pathway, a signaling cascade involved in development and cancer. This review discusses how Motins are integrated into the Hippo signaling network, as either upstream regulators or downstream effectors, to modulate cell proliferation and migration. The repression of YAP/TAZ by Motins contributes to growth inhibition, whereas subcellular localization of Motins and their interactions with actin fibers are critical in regulating cell migration. The net effect of Motins on cell proliferation and migration may contribute to their diverse biological functions.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Lin KY, Gujar MR, Lin J, Ding WY, Huang J, Gao Y, Tan YS, Teng X, Christine LSL, Kanchanawong P, Toyama Y, Wang H. Astrocytes control quiescent NSC reactivation via GPCR signaling-mediated F-actin remodeling. SCIENCE ADVANCES 2024; 10:eadl4694. [PMID: 39047090 PMCID: PMC11268418 DOI: 10.1126/sciadv.adl4694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine filamentous actin (F-actin) structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of myocardin-related transcription factor, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G protein-coupled receptor Smog, G protein αq subunit, Rho1 guanosine triphosphatase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand folded gastrulation to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, an NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.
Collapse
Affiliation(s)
- Kun-Yang Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Mahekta R. Gujar
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jiaen Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Wei Yung Ding
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Jiawen Huang
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Yang Gao
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Ye Sing Tan
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Xiang Teng
- Mechanobiology Institute, Level 5, T-lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Low Siok Lan Christine
- Mechanobiology Institute, Level 5, T-lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, Level 5, T-lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, Level 5, T-lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| |
Collapse
|
10
|
Hu L, Liu D, Zheng D, Lu J, Yuan X, Li Y, Shi F, Shi X, He QY, Li Q, Zhang CZ. Pan-Cancer Proteomics Analysis Reveals Wiskott-Aldrich Syndrome Protein as a Potential Regulator of Programmed Death-Ligand 1. J Proteome Res 2024; 23:2195-2205. [PMID: 38661673 DOI: 10.1021/acs.jproteome.4c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The programmed death-ligand 1 (PD-L1) is a key mediator of immunosuppression in the tumor microenvironment. The expression of PD-L1 in cancer cells is useful for the clinical determination of an immune checkpoint blockade (ICB). However, the regulatory mechanism of the PD-L1 abundance remains incompletely understood. Here, we integrated the proteomics of 52 patients with solid tumors and examined immune cell infiltration to reveal PD-L1-related regulatory modules. Wiskott-Aldrich syndrome protein (WASP) was identified as a potential regulator of PD-L1 transcription. In two independent cohorts containing 164 cancer patients, WASP expression was significantly associated with PD-L1. High WASP expression contributed to immunosuppressive cell composition, including cells positive for immune checkpoints (PD1, CTLA4, TIGIT, and TIM3), FoxP3+ Treg cells, and CD163+ tumor-associated macrophages. Overexpression of WASP increased, whereas knockdown of WASP decreased the protein level of PD-L1 in cancer cells without alteration of PD-L1 protein stability. The WASP-mediated cell migration and invasion were markedly attenuated by the silence of PD-L1. Collectively, our data suggest that WASP is a potential regulator of PD-L1 and the WASP/PD-L1 axis is responsible for cell migration and an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Liling Hu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Danya Liu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Dandan Zheng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiangli Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiaoyi Yuan
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yuying Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Fujin Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xinyu Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuli Li
- Department of Head and Neck, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chris Zhiyi Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
11
|
Nguyen TD, Rao MK, Dhyani SP, Banks JM, Winek MA, Michalkiewicz J, Lee MY. Nucleoporin93 limits Yap activity to prevent endothelial cell senescence. Aging Cell 2024; 23:e14095. [PMID: 38348753 PMCID: PMC11019141 DOI: 10.1111/acel.14095] [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: 11/21/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 02/27/2024] Open
Abstract
As the innermost lining of the vasculature, endothelial cells (ECs) are constantly subjected to systemic inflammation and particularly vulnerable to aging. Endothelial health is hence vital to prevent age-related vascular disease. Healthy ECs rely on the proper localization of transcription factors via nuclear pore complexes (NPCs) to govern cellular behavior. Emerging studies report NPC degradation with natural aging, suggesting impaired nucleocytoplasmic transport in age-associated EC dysfunction. We herein identify nucleoporin93 (Nup93), a crucial structural NPC protein, as an indispensable player in vascular protection. Endothelial Nup93 protein levels are significantly reduced in the vasculature of aged mice, paralleling observations of Nup93 loss when using in vitro models of EC senescence. The loss of Nup93 in human ECs induces cell senescence and promotes the expression of inflammatory adhesion molecules, where restoring Nup93 protein in senescent ECs reverses features of endothelial aging. Mechanistically, we find that both senescence and loss of Nup93 impair endothelial NPC transport, leading to nuclear accumulation of Yap and downstream inflammation. Pharmacological studies indicate Yap hyperactivation as the primary consequence of senescence and Nup93 loss in ECs. Collectively, our findings indicate that the maintenance of endothelial Nup93 is a key determinant of EC health, where aging targets endothelial Nup93 levels to impair NPC function as a novel mechanism of EC senescence and vascular aging.
Collapse
Affiliation(s)
- Tung D. Nguyen
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
- The Center for Cardiovascular ResearchThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Mihir K. Rao
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Shaiva P. Dhyani
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Justin M. Banks
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Michael A. Winek
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Julia Michalkiewicz
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
- The Center for Cardiovascular ResearchThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| | - Monica Y. Lee
- Department of Physiology and BiophysicsThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
- The Center for Cardiovascular ResearchThe University of Illinois at Chicago – College of MedicineChicagoIllinoisUSA
| |
Collapse
|
12
|
Li K, Zhang J, Lyu H, Yang J, Wei W, Wang Y, Luo H, Zhang Y, Jiang X, Yi H, Wang M, Zhang C, Wu K, Xiao L, Wen W, Xu H, Li G, Wan Y, Yang F, Yang R, Fu X, Qin B, Zhou Z, Zhang H, Lee M. CSN6-SPOP-HMGCS1 Axis Promotes Hepatocellular Carcinoma Progression via YAP1 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306827. [PMID: 38308184 PMCID: PMC11005689 DOI: 10.1002/advs.202306827] [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: 09/18/2023] [Revised: 01/15/2024] [Indexed: 02/04/2024]
Abstract
Cholesterol metabolism has important roles in maintaining membrane integrity and countering the development of diseases such as obesity and cancers. Cancer cells sustain cholesterol biogenesis for their proliferation and microenvironment reprograming even when sterols are abundant. However, efficacy of targeting cholesterol metabolism for cancer treatment is always compromised. Here it is shown that CSN6 is elevated in HCC and is a positive regulator of hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) of mevalonate (MVA) pathway to promote tumorigenesis. Mechanistically, CSN6 antagonizes speckle-type POZ protein (SPOP) ubiquitin ligase to stabilize HMGCS1, which in turn activates YAP1 to promote tumor growth. In orthotopic liver cancer models, targeting CSN6 and HMGCS1 hinders tumor growth in both normal and high fat diet. Significantly, HMGCS1 depletion improves YAP inhibitor efficacy in patient derived xenograft models. The results identify a CSN6-HMGCS1-YAP1 axis mediating tumor outgrowth in HCC and propose a therapeutic strategy of targeting non-alcoholic fatty liver diseases- associated HCC.
Collapse
|
13
|
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
|
14
|
Lin KY, Gujar MR, Lin J, Ding WY, Huang J, Gao Y, Tan YS, Teng X, Christine LSL, Kanchanawong P, Toyama Y, Wang H. Astrocytes control quiescent NSC reactivation via GPCR signaling-mediated F-actin remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584337. [PMID: 38903085 PMCID: PMC11188063 DOI: 10.1101/2024.03.11.584337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine F-actin structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of Mrtf, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G-protein-coupled receptor (GPCR) Smog, G-protein αq subunit, Rho1 GTPase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand Fog to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, a NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.
Collapse
|
15
|
Tokamov SA, Buiter S, Ullyot A, Scepanovic G, Williams AM, Fernandez-Gonzalez R, Horne-Badovinac S, Fehon RG. Cortical tension promotes Kibra degradation via Par-1. Mol Biol Cell 2024; 35:ar2. [PMID: 37903240 PMCID: PMC10881160 DOI: 10.1091/mbc.e23-06-0246] [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: 06/26/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/01/2023] Open
Abstract
The Hippo pathway is an evolutionarily conserved regulator of tissue growth. Multiple Hippo signaling components are regulated via proteolytic degradation. However, how these degradation mechanisms are themselves modulated remains unexplored. Kibra is a key upstream pathway activator that promotes its own ubiquitin-mediated degradation upon assembling a Hippo signaling complex. Here, we demonstrate that Hippo complex-dependent Kibra degradation is modulated by cortical tension. Using classical genetic, osmotic, and pharmacological manipulations of myosin activity and cortical tension, we show that increasing cortical tension leads to Kibra degradation, whereas decreasing cortical tension increases Kibra abundance. Our study also implicates Par-1 in regulating Kib abundance downstream of cortical tension. We demonstrate that Par-1 promotes ubiquitin-mediated Kib degradation in a Hippo complex-dependent manner and is required for tension-induced Kib degradation. Collectively, our results reveal a previously unknown molecular mechanism by which cortical tension affects Hippo signaling and provide novel insights into the role of mechanical forces in growth control.
Collapse
Affiliation(s)
- Sherzod A. Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Stephan Buiter
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Anne Ullyot
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Audrey Miller Williams
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomedical Engineering and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
16
|
Deng H, Jia Q, Ming X, Sun Y, Lu Y, Liu L, Zhou J. Hippo pathway in intestinal diseases: focusing on ferroptosis. Front Cell Dev Biol 2023; 11:1291686. [PMID: 38130953 PMCID: PMC10734691 DOI: 10.3389/fcell.2023.1291686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The incidence of intestinal diseases, such as inflammatory bowel disease, gastric cancer, and colorectal cancer, has steadily increased over the past decades. The Hippo pathway is involved in cell proliferation, tissue and organ damage, energy metabolism, tumor formation, and other physiologic processes. Ferroptosis is a form of programmed cell death characterized by the accumulation of iron and lipid peroxides. The Hippo pathway and ferroptosis are associated with various intestinal diseases; however, the crosstalk between them is unclear. This review elaborates on the current research on the Hippo pathway and ferroptosis in the context of intestinal diseases. We summarized the connection between the Hippo pathway and ferroptosis to elucidate the underlying mechanism by which these pathways influence intestinal diseases. We speculate that a mutual regulatory mechanism exists between the Hippo pathway and ferroptosis and these two pathways interact in several ways to regulate intestinal diseases.
Collapse
Affiliation(s)
- Hongwei Deng
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Qiuting Jia
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Xin Ming
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yuxin Sun
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- School of Basic Medicine, Southwest Medical University, Luzhou, China
| | - Yuxuan Lu
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Li Liu
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Zhou
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
17
|
Nguyen TD, Rao MK, Dhyani SP, Banks JM, Winek MA, Michalkiewicz J, Lee MY. Nucleoporin93 (Nup93) Limits Yap Activity to Prevent Endothelial Cell Senescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566598. [PMID: 38014013 PMCID: PMC10680655 DOI: 10.1101/2023.11.10.566598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Endothelial cells (ECs) form the innermost lining of the vasculature and serve a pivotal role in preventing age-related vascular disease. Endothelial health relies on the proper nucleocytoplasmic shuttling of transcription factors via nuclear pore complexes (NPCs). Emerging studies report NPC degradation with natural aging, suggesting impaired nucleocytoplasmic transport in age-related EC dysfunction. We herein identify nucleoporin93 (Nup93), a crucial structural NPC protein, as an indispensable player for vascular protection. Endothelial Nup93 protein levels are significantly reduced in the vasculature of aged mice, paralleling observations of Nup93 loss when using in vitro models of endothelial aging. Mechanistically, we find that loss of Nup93 impairs NPC transport, leading to the nuclear accumulation of Yap and downstream inflammation. Collectively, our findings indicate maintenance of endothelial Nup93 as a key determinant of EC health, where aging targets endothelial Nup93 levels to impair NPC function as a novel mechanism for EC senescence and vascular aging.
Collapse
|
18
|
Tokamov SA, Nouri N, Rich A, Buiter S, Glotzer M, Fehon RG. Apical polarity and actomyosin dynamics control Kibra subcellular localization and function in Drosophila Hippo signaling. Dev Cell 2023; 58:1864-1879.e4. [PMID: 37729921 PMCID: PMC10591919 DOI: 10.1016/j.devcel.2023.08.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/02/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
The Hippo pathway is an evolutionarily conserved regulator of tissue growth that integrates inputs from both polarity and actomyosin networks. An upstream activator of the Hippo pathway, Kibra, localizes at the junctional and medial regions of the apical cortex in epithelial cells, and medial accumulation promotes Kibra activity. Here, we demonstrate that cortical Kibra distribution is controlled by a tug-of-war between apical polarity and actomyosin dynamics. We show that while the apical polarity network, in part via atypical protein kinase C (aPKC), tethers Kibra at the junctional cortex to silence its activity, medial actomyosin flows promote Kibra-mediated Hippo complex formation at the medial cortex, thereby activating the Hippo pathway. This study provides a mechanistic understanding of the relationship between the Hippo pathway, polarity, and actomyosin cytoskeleton, and it offers novel insights into how fundamental features of epithelial tissue architecture can serve as inputs into signaling cascades that control tissue growth, patterning, and morphogenesis.
Collapse
Affiliation(s)
- Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Nicki Nouri
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Ashley Rich
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Stephan Buiter
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
19
|
Ran X, Müller S, Brunssen C, Huhle R, Scharffenberg M, Schnabel C, Koch T, Gama de Abreu M, Morawietz H, Ferreira JMC, Wittenstein J. Modulation of the hippo-YAP pathway by cyclic stretch in rat type 2 alveolar epithelial cells-a proof-of-concept study. Front Physiol 2023; 14:1253810. [PMID: 37877098 PMCID: PMC10591329 DOI: 10.3389/fphys.2023.1253810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/28/2023] [Indexed: 10/26/2023] Open
Abstract
Background: Mechanical ventilation (MV) is a life supporting therapy but may also cause lung damage. This phenomenon is known as ventilator-induced lung injury (VILI). A potential pathomechanisms of ventilator-induced lung injury may be the stretch-induced production and release of cytokines and pro-inflammatory molecules from the alveolar epithelium. Yes-associated protein (YAP) might be regulated by mechanical forces and involved in the inflammation cascade. However, its role in stretch-induced damage of alveolar cells remains poorly understood. In this study, we explored the role of YAP in the response of alveolar epithelial type II cells (AEC II) to elevated cyclic stretch in vitro. We hypothesize that Yes-associated protein activates its downstream targets and regulates the interleukin-6 (IL-6) expression in response to 30% cyclic stretch in AEC II. Methods: The rat lung L2 cell line was exposed to 30% cyclic equibiaxial stretch for 1 or 4 h. Non-stretched conditions served as controls. The cytoskeleton remodeling and cell junction integrity were evaluated by F-actin and Pan-cadherin immunofluorescence, respectively. The gene expression and protein levels of IL-6, Yes-associated protein, Cysteine-rich angiogenic inducer 61 (Cyr61/CCN1), and connective tissue growth factor (CTGF/CCN2) were studied by real-time polymerase chain reaction (RT-qPCR) and Western blot, respectively. Verteporfin (VP) was used to inhibit Yes-associated protein activation. The effects of 30% cyclic stretch were assessed by two-way ANOVA. Statistical significance as accepted at p < 0.05. Results: Cyclic stretch of 30% induced YAP nuclear accumulation, activated the transcription of Yes-associated protein downstream targets Cyr61/CCN1 and CTGF/CCN2 and elevated IL-6 expression in AEC II after 1 hour, compared to static control. VP (2 µM) inhibited Yes-associated protein activation in response to 30% cyclic stretch and reduced IL-6 protein levels. Conclusion: In rat lung L2 AEC II, 30% cyclic stretch activated YAP, and its downstream targets Cyr61/CCN1 and CTGF/CCN2 and proinflammatory IL-6 expression. Target activation was blocked by a Yes-associated protein inhibitor. This novel YAP-dependent pathway could be involved in stretch-induced damage of alveolar cells.
Collapse
Affiliation(s)
- Xi Ran
- Department of Intensive Care Medicine, Chongqing General Hospital, Changqing, China
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Sabine Müller
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Robert Huhle
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Martin Scharffenberg
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Christian Schnabel
- Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Thea Koch
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Marcelo Gama de Abreu
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
- Department of Intensive Care and Resuscitation, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Outcomes Research, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Jorge M. C. Ferreira
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Jakob Wittenstein
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
20
|
Bai Y, Zhao F, Wu T, Chen F, Pang X. Actin polymerization and depolymerization in developing vertebrates. Front Physiol 2023; 14:1213668. [PMID: 37745245 PMCID: PMC10515290 DOI: 10.3389/fphys.2023.1213668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Development is a complex process that occurs throughout the life cycle. F-actin, a major component of the cytoskeleton, is essential for the morphogenesis of tissues and organs during development. F-actin is formed by the polymerization of G-actin, and the dynamic balance of polymerization and depolymerization ensures proper cellular function. Disruption of this balance results in various abnormalities and defects or even embryonic lethality. Here, we reviewed recent findings on the structure of G-actin and F-actin and the polymerization of G-actin to F-actin. We also focused on the functions of actin isoforms and the underlying mechanisms of actin polymerization/depolymerization in cellular and organic morphogenesis during development. This information will extend our understanding of the role of actin polymerization in the physiologic or pathologic processes during development and may open new avenues for developing therapeutics for embryonic developmental abnormalities or tissue regeneration.
Collapse
Affiliation(s)
- Yang Bai
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Feng Zhao
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingting Wu
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Fangchun Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
21
|
Oliva-Vilarnau N, Vorrink SU, Büttner FA, Heinrich T, Sensbach J, Koscielski I, Wienke D, Petersson C, Perrin D, Lauschke VM. Comparative analysis of YAP/TEAD inhibitors in 2D and 3D cultures of primary human hepatocytes reveals a novel non-canonical mechanism of CYP induction. Biochem Pharmacol 2023; 215:115755. [PMID: 37607620 DOI: 10.1016/j.bcp.2023.115755] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
Induction of cytochrome P450 (CYP) genes constitutes an important cause of drug-drug interactions and preclinical evaluation of induction liability is mandatory for novel drug candidates. YAP/TEAD signaling has emerged as an attractive target for various oncological indications and multiple chemically distinct YAP/TEAD inhibitors are rapidly progressing towards clinical stages. Here, we tested the liability for CYP induction of a diverse set of YAP/TEAD inhibitors with different modes of action and TEAD isoform selectivity profiles in monolayers and 3D spheroids of primary human hepatocytes (PHH). We found that YAP/TEAD inhibition resulted in broad induction of CYPs in 2D monolayers, whereas, if at all, only marginal induction was seen in spheroid culture. Comprehensive RNA-Seq indicated that YAP/TEAD signaling was increased in 2D culture compared to spheroids, which was paralleled by elevated activities of the interacting transcription factors LXR and ESRRA, likely at least in part due to altered mechanosensing. Inhibition of this YAP/TEAD hyperactivation resulted in an overall reduction of hepatocyte dedifferentiation marked by increased hepatic functionality, including CYPs. These results thus demonstrate that the observed induction is due to on-target effects of the compounds rather than direct activation of xenobiotic sensing nuclear receptors. Combined, the presented data link hepatocyte dedifferentiation to YAP/TEAD dysregulation, reveal a novel non-canonical pathway of CYP induction and highlight the advantage of organotypic 3D cultures to predict clinically relevant pharmacokinetic properties, particularly for atypical induction mechanisms.
Collapse
Affiliation(s)
- Nuria Oliva-Vilarnau
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Florian A Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany
| | - Timo Heinrich
- Department of Medicinal Chemistry and Drug Design, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Janike Sensbach
- Department of Chemical and Pre-Clinical Safety, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Isabel Koscielski
- Department of Chemical and Pre-Clinical Safety, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dirk Wienke
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Carl Petersson
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Dominique Perrin
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; HepaPredict AB, Stockholm, Sweden; Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany; University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
22
|
Mokhtari RB, Ashayeri N, Baghaie L, Sambi M, Satari K, Baluch N, Bosykh DA, Szewczuk MR, Chakraborty S. The Hippo Pathway Effectors YAP/TAZ-TEAD Oncoproteins as Emerging Therapeutic Targets in the Tumor Microenvironment. Cancers (Basel) 2023; 15:3468. [PMID: 37444578 DOI: 10.3390/cancers15133468] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Various cancer cell-associated intrinsic and extrinsic inputs act on YAP/TAZ proteins to mediate the hyperactivation of the TEAD transcription factor-based transcriptome. This YAP/TAZ-TEAD activity can override the growth-limiting Hippo tumor-suppressor pathway that maintains normal tissue homeostasis. Herein, we provide an integrated summary of the contrasting roles of YAP/TAZ during normal tissue homeostasis versus tumor initiation and progression. In addition to upstream factors that regulate YAP/TAZ in the TME, critical insights on the emerging functions of YAP/TAZ in immune suppression and abnormal vasculature development during tumorigenesis are illustrated. Lastly, we discuss the current methods that intervene with the YAP/TAZ-TEAD oncogenic signaling pathway and the emerging applications of combination therapies, gut microbiota, and epigenetic plasticity that could potentiate the efficacy of chemo/immunotherapy as improved cancer therapeutic strategies.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Neda Ashayeri
- Division of Hematology and Oncology, Department of Pediatrics, Ali-Asghar Children Hospital, Iran University of Medical Science, Tehran 1449614535, Iran
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Manpreet Sambi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Kosar Satari
- Division of Hematology and Oncology, Department of Pediatrics, Ali-Asghar Children Hospital, Iran University of Medical Science, Tehran 1449614535, Iran
| | - Narges Baluch
- Department of Immunology and Allergy, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Dmitriy A Bosykh
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Sayan Chakraborty
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
23
|
Ibar C, Chinthalapudi K, Heissler SM, Irvine KD. Competition between myosin II and β H-spectrin regulates cytoskeletal tension. eLife 2023; 12:RP84918. [PMID: 37367948 DOI: 10.7554/elife.84918] [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] [Indexed: 06/28/2023] Open
Abstract
Spectrins are membrane cytoskeletal proteins generally thought to function as heterotetramers comprising two α-spectrins and two β-spectrins. They influence cell shape and Hippo signaling, but the mechanism by which they influence Hippo signaling has remained unclear. We have investigated the role and regulation of the Drosophila β-heavy spectrin (βH-spectrin, encoded by the karst gene) in wing imaginal discs. Our results establish that βH-spectrin regulates Hippo signaling through the Jub biomechanical pathway due to its influence on cytoskeletal tension. While we find that α-spectrin also regulates Hippo signaling through Jub, unexpectedly, we find that βH-spectrin localizes and functions independently of α-spectrin. Instead, βH-spectrin co-localizes with and reciprocally regulates and is regulated by myosin. In vivo and in vitro experiments support a model in which βH-spectrin and myosin directly compete for binding to apical F-actin. This competition can explain the influence of βH-spectrin on cytoskeletal tension and myosin accumulation. It also provides new insight into how βH-spectrin participates in ratcheting mechanisms associated with cell shape change.
Collapse
Affiliation(s)
- Consuelo Ibar
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, United States
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, United States
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, United States
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, United States
| |
Collapse
|
24
|
Xia L, Shen Y, Liu S, Du J. Iron overload triggering ECM-mediated Hippo/YAP pathway in follicle development: a hypothetical model endowed with therapeutic implications. Front Endocrinol (Lausanne) 2023; 14:1174817. [PMID: 37223010 PMCID: PMC10200985 DOI: 10.3389/fendo.2023.1174817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/12/2023] [Indexed: 05/25/2023] Open
Abstract
Disruption of iron homeostasis plays a negative role in follicle development. The dynamic changes in follicle growth are dependent on Hippo/YAP signaling and mechanical forces. However, little is known about the liaison between iron overload and the Hippo/YAP signalling pathway in term of folliculogenesis. Here, based on the available evidence, we established a hypothesized model linking excessive iron, extracellular matrix (ECM), transforming growth factor-β (TGF-β) and Hippo/Yes-associated protein (YAP) signal regarding follicle development. Hypothetically, the TGF-β signal and iron overload may play a synergistic role in ECM production via YAP. We speculate that the dynamic homeostasis of follicular iron interacts with YAP, increasing the risk of ovarian reserve loss and may enhance the sensitivity of follicles to accumulated iron. Hence, therapeutic interventions targeting iron metabolism disorders, and Hippo/YAP signal may alter the consequences of the impaired developmental process based on our hypothesis, which provides potential targets and inspiration for further drug discovery and development applied to clinical treatment.
Collapse
Affiliation(s)
- Lingjin Xia
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Yupei Shen
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Du
- National Health Commission of the People's Republic of China (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Huang Y, Gui J, Myllymäki SM, Mikkola ML, Shimmi O. Coordination of tissue homeostasis and growth by the Scribble-α-Catenin-Septate junction complex. iScience 2023; 26:106490. [PMID: 37096043 PMCID: PMC10122046 DOI: 10.1016/j.isci.2023.106490] [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: 12/20/2021] [Revised: 02/27/2023] [Accepted: 03/18/2023] [Indexed: 04/26/2023] Open
Abstract
Maintaining apicobasal polarity (ABP) is crucial for epithelial integrity and homeostasis during tissue development. Although intracellular mechanisms underlying ABP establishment have been well studied, it remains to be addressed how the ABP coordinates tissue growth and homeostasis. By studying Scribble, a key ABP determinant, we address molecular mechanisms underlying ABP-mediated growth control in the Drosophila wing imaginal disc. Our data reveal that genetic and physical interactions between Scribble, Septate junction complex and α-Catenin appear to be key for sustaining ABP-mediated growth control. Cells with conditional scribble knockdown instigate the loss of α-Catenin, ultimately leading to the formation of neoplasia accompanying with activation of Yorkie. In contrast, cells expressing wild type scribble progressively restore ABP in scribble hypomorphic mutant cells in a non-autonomous manner. Our findings provide unique insights into cellular communication among optimal and sub-optimal cells to regulate epithelial homeostasis and growth.
Collapse
Affiliation(s)
- Yunxian Huang
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Jinghua Gui
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | | | - Marja L. Mikkola
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
- Institute of Molecular and Cell Biology, University of Tartu, 51010 Tartu, Estonia
- Corresponding author
| |
Collapse
|
26
|
Nguyen MT, Lee W. Induction of miR-665-3p Impairs the Differentiation of Myogenic Progenitor Cells by Regulating the TWF1-YAP1 Axis. Cells 2023; 12:cells12081114. [PMID: 37190023 DOI: 10.3390/cells12081114] [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: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Actin dynamics are known to orchestrate various myogenic processes in progenitor cells. Twinfilin-1 (TWF1) is an actin-depolymerizing factor that plays a crucial role in the differentiation of myogenic progenitor cells. However, little is known about the mechanisms underlying the epigenetic regulation of TWF1 expression and impaired myogenic differentiation in the background of muscle wasting. This study investigated how miR-665-3p affects TWF1 expression, actin filaments' modulation, proliferation, and myogenic differentiation in progenitor cells. Palmitic acid, the most prevalent saturated fatty acid (SFA) in food, suppressed TWF1 expression and inhibited the myogenic differentiation of C2C12 cells while increasing the level of miR-665-3p expression. Interestingly, miR-665-3p inhibited TWF1 expression by targeting TWF1 3'UTR directly. In addition, miR-665-3p accumulated filamentous actin (F-actin) and enhanced the nuclear translocation of Yes-associated protein 1 (YAP1), consequently promoting cell cycle progression and proliferation. Furthermore, miR-665-3p suppressed the expressions of myogenic factors, i.e., MyoD, MyoG, and MyHC, and consequently impaired myoblast differentiation. In conclusion, this study suggests that SFA-inducible miR-665-3p suppresses TWF1 expression epigenetically and inhibits myogenic differentiation by facilitating myoblast proliferation via the F-actin/YAP1 axis.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
27
|
Nguyen MT, Lee W. Mir-302a/TWF1 Axis Impairs the Myogenic Differentiation of Progenitor Cells through F-Actin-Mediated YAP1 Activation. Int J Mol Sci 2023; 24:ijms24076341. [PMID: 37047312 PMCID: PMC10094299 DOI: 10.3390/ijms24076341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Actin cytoskeleton dynamics have been found to regulate myogenesis in various progenitor cells, and twinfilin-1 (TWF1), an actin-depolymerizing factor, plays a vital role in actin dynamics and myoblast differentiation. Nevertheless, the molecular mechanisms underlying the epigenetic regulation and biological significance of TWF1 in obesity and muscle wasting have not been explored. Here, we investigated the roles of miR-302a in TWF1 expression, actin filament modulation, proliferation, and myogenic differentiation in C2C12 progenitor cells. Palmitic acid, the most prevalent saturated fatty acid (SFA) in the diet, decreased the expression of TWF1 and impeded myogenic differentiation while increasing the miR-302a levels in C2C12 myoblasts. Interestingly, miR-302a inhibited TWF1 expression directly by targeting its 3′UTR. Furthermore, ectopic expression of miR-302a promoted cell cycle progression and proliferation by increasing the filamentous actin (F-actin) accumulation, which facilitated the nuclear translocation of Yes-associated protein 1 (YAP1). Consequently, by suppressing the expressions of myogenic factors, i.e., MyoD, MyoG, and MyHC, miR-302a impaired myoblast differentiation. Hence, this study demonstrated that SFA-inducible miR-302a suppresses TWF1 expression epigenetically and impairs myogenic differentiation by facilitating myoblast proliferation via F-actin-mediated YAP1 activation.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
28
|
Golubev DA, Zemskaya NV, Gorbunova AA, Kukuman DV, Moskalev A, Shaposhnikov MV. Studying the Geroprotective Properties of YAP/TAZ Signaling Inhibitors on Drosophila melanogaster Model. Int J Mol Sci 2023; 24:ijms24066006. [PMID: 36983079 PMCID: PMC10058302 DOI: 10.3390/ijms24066006] [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: 12/01/2022] [Revised: 02/28/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the main downstream effectors of the evolutionarily conserved Hippo signaling pathway. YAP/TAZ are implicated in the transcriptional regulation of target genes that are involved in a wide range of key biological processes affecting tissue homeostasis and play dual roles in the aging process, depending on the cellular and tissue context. The aim of the present study was to investigate whether pharmacological inhibitors of Yap/Taz increase the lifespan of Drosophila melanogaster. Real-time qRT-PCR was performed to measure the changes in the expression of Yki (Yorkie, the Drosophila homolog of YAP/TAZ) target genes. We have revealed a lifespan-increasing effect of YAP/TAZ inhibitors that was mostly associated with decreased expression levels of the wg and E2f1 genes. However, further analysis is required to understand the link between the YAP/TAZ pathway and aging.
Collapse
Affiliation(s)
- Denis A Golubev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Nadezhda V Zemskaya
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Anastasia A Gorbunova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Daria V Kukuman
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Mikhail V Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| |
Collapse
|
29
|
Zhang H, Mao Z, Yang Z, Nakamura F. Identification of Filamin A Mechanobinding Partner III: SAV1 Specifically Interacts with Filamin A Mechanosensitive Domain 21. Biochemistry 2023; 62:1197-1208. [PMID: 36857526 DOI: 10.1021/acs.biochem.2c00665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Filamin A (FLNA) cross-links actin filaments and mediates mechanotransduction by force-induced conformational changes of its domains. FLNA's mechanosensitive immunoglobulin-like repeats (R) interact with each other to create cryptic binding sites, which can be exposed by physiologically relevant mechanical forces. Using the FLNA mechanosensing domains as an affinity ligand followed by stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics, we recently identified smoothelin and fimbacin as FLNA mechanobinding proteins. Here, using the mechanosensing domain as an affinity ligand and two labeled amino acids, we identify salvador homologue 1 (SAV1), a component of the Hippo pathway kinase cascade, as a new FLNA mechanobinding partner. We demonstrate that SAV1 specifically interacts with the cryptic C-D cleft of FLNA R21 and map the FLNA-binding site on SAV1. We show that point mutations on the R21 C strand block the SAV1 interaction and find that SAV1 contains a FLNA-binding motif in the central region (116Phe-124Val). Point mutations F116A and T118A (FT/AA) disrupt the interaction. A proximity ligation assay reveals that their interaction occurs in the cytosol in an actin polymerization-dependent manner. Although SAV1 is typically found in the cytosol, disrupting the interaction between SAV1 and FLNA causes SAV1 to diffuse to the nucleus and YAP1 to diffuse to the cytosol in an inverse relationship. These results suggest that FLNA mediates regulation of the Hippo pathway through actin polymerization-dependent interaction with SAV1.
Collapse
Affiliation(s)
- Huaguan Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Zhenfeng Mao
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Ziwei Yang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
30
|
Maejima Y, Zablocki D, Nah J, Sadoshima J. The role of the Hippo pathway in autophagy in the heart. Cardiovasc Res 2023; 118:3320-3330. [PMID: 35150237 DOI: 10.1093/cvr/cvac014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/07/2022] [Indexed: 01/25/2023] Open
Abstract
The Hippo pathway, an evolutionarily conserved signalling mechanism, controls organ size and tumourigenesis. Increasing lines of evidence suggest that autophagy, an important mechanism of lysosome-mediated cellular degradation, is regulated by the Hippo pathway, which thereby profoundly affects cell growth and death responses in various cell types. In the heart, Mst1, an upstream component of the Hippo pathway, not only induces apoptosis but also inhibits autophagy through phosphorylation of Beclin 1. YAP/TAZ, transcription factor co-factors and the terminal effectors of the Hippo pathway, affect autophagy through transcriptional activation of TFEB, a master regulator of autophagy and lysosomal biogenesis. The cellular abundance of YAP is negatively regulated by autophagy and suppression of autophagy induces accumulation of YAP, which, in turn, acts as a feedback mechanism to induce autophagosome formation. Thus, the Hippo pathway and autophagy regulate each other, thereby profoundly affecting cardiomyocyte survival and death. This review discusses the interaction between the Hippo pathway and autophagy and its functional significance during stress conditions in the heart and the cardiomyocytes therein.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA.,Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| | - Jihoon Nah
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave., MSB G-609, Newark, NJ 07103, USA
| |
Collapse
|
31
|
Genome Editing and Cardiac Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:37-52. [DOI: 10.1007/978-981-19-5642-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
32
|
Bhavnagari H, Raval A, Shah F. Deciphering Potential Role of Hippo Signaling Pathway in Breast Cancer: A Comprehensive Review. Curr Pharm Des 2023; 29:3505-3518. [PMID: 38141194 DOI: 10.2174/0113816128274418231215054210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/25/2023]
Abstract
Breast cancer is a heterogeneous disease and a leading malignancy around the world. It is a vital cause of untimely mortality among women. Drug resistance is the major challenge for effective cancer therapeutics. In contrast, cancer stem cells (CSCs) are one of the reasons for drug resistance, tumor progression, and metastasis. The small population of CSCs present in each tumor has the ability of self-renewal, differentiation, and tumorigenicity. CSCs are often identified and enriched using a variety of cell surface markers (CD44, CD24, CD133, ABCG2, CD49f, LGR5, SSEA-3, CD70) that exert their functions by different regulatory networks, i.e., Notch, Wnt/β-catenin, hedgehog (Hh), and Hippo signaling pathways. Particularly the Hippo signaling pathway is the emerging and very less explored cancer stem cell pathway. Here, in this review, the Hippo signaling molecules are elaborated with respect to their ability of stemness as epigenetic modulators and how these molecules can be targeted for better cancer treatment and to overcome drug resistance.
Collapse
Affiliation(s)
- Hunayna Bhavnagari
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Apexa Raval
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Franky Shah
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| |
Collapse
|
33
|
Hwang SH, Yang Y, Jung JH, Kim Y. Oleic acid from cancer-associated fibroblast promotes cancer cell stemness by stearoyl-CoA desaturase under glucose-deficient condition. Cancer Cell Int 2022; 22:404. [PMID: 36514170 PMCID: PMC9746202 DOI: 10.1186/s12935-022-02824-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) coordinate the malignancy of cancer cells via secretory materials. Reprogrammed lipid metabolism and signaling play critical roles in cancer biology. Oleic acid (OA) serves as a source of energy under glucose-deficient conditions, but its function in cancer progression remains unclear. The present study investigated that CAFs in xenografted tumors had higher amounts of fatty acids, particularly OA, compared to normal fibroblasts, and promoted the cancer cell stemness in lung adenocarcinoma cells under glucose-deficient condition. METHODS Xenografts were established in immunodeficient mice by injection of NCI-H460 (H460) cells. Lipids and fatty acids were evaluated using the BODIPY staining and fatty-acid methyl esters analysis. The expression levels of markers for lipid metabolism and cancer stemness were determined by western blot, flow cytometry, and real-time PCR. Cancer cell subclones against stearoyl-CoA desaturase (SCD) were produced by lentiviral vector and CRISPR/cas9 systems. The expression of SCD was examined immunochemically in human adenocarcinoma tissues, and its clinical relevance to survival rate in lung adenocarcinoma patients was assessed by Kaplan-Meier analysis. RESULTS Transferred CAF-derived OA through lipid transporter upregulated SCD in cancer cells under glucose-deficient conditions, resulting in enhanced lipid metabolism and autophagosome maturation. By OA treatment under glucose deficient condition, cancer cell stemness was significantly enhanced through sequential activation of SCD, F-actin polymerization and nuclear translocation of yes-associated protein. These findings were confirmed by experiments using chemical inhibitors, SCD-overexpressing cells and SCD-knockout (KO) cells. When xenografted, SCD-overexpressing cells produced larger tumors compared with parental cells, while SCD-KO cells generated much smaller tumors. Analysis of tumor tissue microarray from lung adenocarcinoma patients revealed that SCD expression was the marker for poor prognosis involving tumor grade, clinical stage and survival rate. CONCLUSION Our data indicate that CAFs-derived OA activated lipid metabolism in lung adenocarcinoma cells under glucose-deficient conditions, subsequently enhancing stemness and progression toward malignancy.
Collapse
Affiliation(s)
- Sung-Hyun Hwang
- grid.31501.360000 0004 0470 5905Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea ,grid.31501.360000 0004 0470 5905BK21 Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea ,grid.412480.b0000 0004 0647 3378Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam, 13620 Republic of Korea
| | - Yeseul Yang
- grid.31501.360000 0004 0470 5905BK21 Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea ,grid.31501.360000 0004 0470 5905Laboratory of Clinical Pathology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea
| | - Jae-Ha Jung
- grid.31501.360000 0004 0470 5905BK21 Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea ,grid.31501.360000 0004 0470 5905Laboratory of Clinical Pathology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea
| | - Yongbaek Kim
- grid.31501.360000 0004 0470 5905BK21 Future Veterinary Medicine Leading Education and Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea ,grid.31501.360000 0004 0470 5905Laboratory of Clinical Pathology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826 Republic of Korea
| |
Collapse
|
34
|
Hu L, Brichalli W, Li N, Chen S, Cheng Y, Liu Q, Xiong Y, Yu J. Myotubularin functions through actomyosin to interact with the Hippo pathway. EMBO Rep 2022; 23:e55851. [PMID: 36285521 PMCID: PMC9724681 DOI: 10.15252/embr.202255851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway is an evolutionarily conserved developmental pathway that controls organ size by integrating diverse regulatory inputs, including actomyosin-mediated cytoskeletal tension. Despite established connections between the actomyosin cytoskeleton and the Hippo pathway, the upstream regulation of actomyosin in the Hippo pathway is less defined. Here, we identify the phosphoinositide-3-phosphatase Myotubularin (Mtm) as a novel upstream regulator of actomyosin that functions synergistically with the Hippo pathway during growth control. Mechanistically, Mtm regulates membrane phospholipid PI(3)P dynamics, which, in turn, modulates actomyosin activity through Rab11-mediated vesicular trafficking. We reveal PI(3)P dynamics as a novel mode of upstream regulation of actomyosin and establish Rab11-mediated vesicular trafficking as a functional link between membrane lipid dynamics and actomyosin activation in the context of growth control. Our study also shows that MTMR2, the human counterpart of Drosophila Mtm, has conserved functions in regulating actomyosin activity and tissue growth, providing new insights into the molecular basis of MTMR2-related peripheral nerve myelination and human disorders.
Collapse
Affiliation(s)
- Liang Hu
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Wyatt Brichalli
- Department of Anatomy & PhysiologyKansas State University College of Veterinary MedicineManhattanKSUSA
| | - Naren Li
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Shifan Chen
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Yaqing Cheng
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsCTUSA
| | - Qinfang Liu
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Yulan Xiong
- Department of NeuroscienceUniversity of Connecticut School of MedicineFarmingtonCTUSA
| | - Jianzhong Yu
- Department of Physiology and NeurobiologyUniversity of ConnecticutStorrsCTUSA
| |
Collapse
|
35
|
Liu D, Tsarouhas V, Samakovlis C. WASH activation controls endosomal recycling and EGFR and Hippo signaling during tumor-suppressive cell competition. Nat Commun 2022; 13:6243. [PMID: 36271083 PMCID: PMC9587002 DOI: 10.1038/s41467-022-34067-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/12/2022] [Indexed: 12/25/2022] Open
Abstract
Cell competition is a conserved homeostatic mechanism whereby epithelial cells eliminate neighbors with lower fitness. Cell communication at the interface of wild-type "winner" cells and polarity-deficient (scrib-/-) "losers" is established through Sas-mediated Ptp10D activation in polarity-deficient cells. This tumor-suppressive cell competition restrains EGFR and Hippo signaling and enables Eiger-JNK mediated apoptosis in scrib-/- clones. Here, we show that the activation state of the endosomal actin regulator WASH is a central node linking EGFR and Hippo signaling activation. The tyrosine kinase Btk29A and its substrate WASH are required downstream of Ptp10D for "loser" cell elimination. Constitutively active, phosphomimetic WASH is sufficient to induce both EGFR and Yki activation leading to overgrowth. On the mechanistic level we show that Ptp10D is recycled by the WASH/retromer complex, while EGFR is recycled by the WASH/retriever complex. Constitutive WASH activation selectively interferes with retromer function leading to Ptp10D mistargeting while promoting EGFR recycling and signaling activation. Phospho-WASH also activates aberrant Arp2/3 actin polymerization, leading to cytoskeletal imbalance, Yki activation and reduced apoptosis. Selective manipulation of WASH phosphorylation on sorting endosomes may restrict epithelial tumorous growth.
Collapse
Affiliation(s)
- Dan Liu
- grid.10548.380000 0004 1936 9377Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| | - Vasilios Tsarouhas
- grid.10548.380000 0004 1936 9377Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| | - Christos Samakovlis
- grid.10548.380000 0004 1936 9377Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden ,grid.8664.c0000 0001 2165 8627Cardiopulmonary Institute, Justus Liebig University of Giessen, Giessen, Germany
| |
Collapse
|
36
|
Huang Y, Gui J, Myllymäki SM, Roy K, Tõnissoo T, Mikkola ML, Shimmi O. Scribble and α-Catenin cooperatively regulate epithelial homeostasis and growth. Front Cell Dev Biol 2022; 10:912001. [PMID: 36211469 PMCID: PMC9532510 DOI: 10.3389/fcell.2022.912001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial homeostasis is an emergent property of both physical and biochemical signals emanating from neighboring cells and across tissue. A recent study reveals that Scribble, an apico-basal polarity determinant, cooperates with α-Catenin, an adherens junction component, to regulate tissue homeostasis in the Drosophila wing imaginal disc. However, it remains to be addressed whether similar mechanisms are utilized in vertebrates. In this study, we first address how α-Catenin cooperates with Scribble to regulate epithelial homeostasis and growth in mammalian cells. Our data show that α-Catenin and Scribble interact physically in mammalian cells. We then found that both α-Catenin and Scribble are required for regulating nuclear translocation of YAP, an effector of the Hippo signaling pathway. Furthermore, ectopic Scribble suffices to suppress YAP in an α-Catenin-dependent manner. Then, to test our hypothesis that Scribble amounts impact epithelial growth, we use the Drosophila wing imaginal disc. We show that Scribble expression is complementary to Yorkie signal, the Drosophila ortholog of YAP. Ectopic expression of full-length Scribble or Scribble Leucine Rich Region (LRR):α-Catenin chimera sufficiently down-regulates Yorkie signal, leading to smaller wing size. Moreover, Scribble LRR:α-Catenin chimera rescues scribble mutant clones in the wing imaginal disc to maintain tissue homeostasis. Taken together, our studies suggest that the association of cell polarity component Scribble with α-Catenin plays a conserved role in epithelial homeostasis and growth.
Collapse
Affiliation(s)
- Yunxian Huang
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jinghua Gui
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | | | - Kallol Roy
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Tambet Tõnissoo
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Marja L. Mikkola
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- *Correspondence: Osamu Shimmi,
| |
Collapse
|
37
|
Zheng A, Chen Q, Zhang L. The Hippo-YAP pathway in various cardiovascular diseases: Focusing on the inflammatory response. Front Immunol 2022; 13:971416. [PMID: 36059522 PMCID: PMC9433876 DOI: 10.3389/fimmu.2022.971416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster and mammals as a key regulator of tissue growth both in physiological and pathological states. Numerous studies depict the vital role of the Hippo pathway in cardiovascular development, heart regeneration, organ size and vascular remodeling through the regulation of YAP (yes-associated protein) translocation. Recently, an increasing number of studies have focused on the Hippo-YAP pathway in inflammation and immunology. Although the Hippo-YAP pathway has been revealed to play controversial roles in different contexts and cell types in the cardiovascular system, the mechanisms regulating tissue inflammation and the immune response remain to be clarified. In this review, we summarize findings from the past decade on the function and mechanism of the Hippo-YAP pathway in CVDs (cardiovascular diseases) such as myocardial infarction, cardiomyopathy and atherosclerosis. In particular, we emphasize the role of the Hippo-YAP pathway in regulating inflammatory cell infiltration and inflammatory cytokine activation.
Collapse
Affiliation(s)
| | | | - Li Zhang
- *Correspondence: Li Zhang, ; Qishan Chen,
| |
Collapse
|
38
|
Khalil K, Eon A, Janody F. Cell Architecture-Dependent Constraints: Critical Safeguards to Carcinogenesis. Int J Mol Sci 2022; 23:8622. [PMID: 35955754 PMCID: PMC9369145 DOI: 10.3390/ijms23158622] [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: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Animal cells display great diversity in their shape. These morphological characteristics result from crosstalk between the plasma membrane and the force-generating capacities of the cytoskeleton macromolecules. Changes in cell shape are not merely byproducts of cell fate determinants, they also actively drive cell fate decisions, including proliferation and differentiation. Global and local changes in cell shape alter the transcriptional program by a multitude of mechanisms, including the regulation of physical links between the plasma membrane and the nuclear envelope and the mechanical modulation of cation channels and signalling molecules. It is therefore not surprising that anomalies in cell shape contribute to several diseases, including cancer. In this review, we discuss the possibility that the constraints imposed by cell shape determine the behaviour of normal and pro-tumour cells by organizing the whole interconnected regulatory network. In turn, cell behaviour might stabilize cells into discrete shapes. However, to progress towards a fully transformed phenotype and to acquire plasticity properties, pro-tumour cells might need to escape these cell shape restrictions. Thus, robust controls of the cell shape machinery may represent a critical safeguard against carcinogenesis.
Collapse
Affiliation(s)
- Komal Khalil
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Master Programme in Oncology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Alice Eon
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Magistère Européen de Génétique, Université Paris Cité, 5 Rue Thomas Mann, 75013 Paris, France
| | - Florence Janody
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| |
Collapse
|
39
|
Park J, Jia S, Salter D, Bagnaninchi P, Hansen CG. The Hippo pathway drives the cellular response to hydrostatic pressure. EMBO J 2022; 41:e108719. [PMID: 35702882 PMCID: PMC9251841 DOI: 10.15252/embj.2021108719] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Cells need to rapidly and precisely react to multiple mechanical and chemical stimuli in order to ensure precise context-dependent responses. This requires dynamic cellular signalling events that ensure homeostasis and plasticity when needed. A less well-understood process is cellular response to elevated interstitial fluid pressure, where the cell senses and responds to changes in extracellular hydrostatic pressure. Here, using quantitative label-free digital holographic imaging, combined with genome editing, biochemical assays and confocal imaging, we analyse the temporal cellular response to hydrostatic pressure. Upon elevated cyclic hydrostatic pressure, the cell responds by rapid, dramatic and reversible changes in cellular volume. We show that YAP and TAZ, the co-transcriptional regulators of the Hippo signalling pathway, control cell volume and that cells without YAP and TAZ have lower plasma membrane tension. We present direct evidence that YAP/TAZ drive the cellular response to hydrostatic pressure, a process that is at least partly mediated via clathrin-dependent endocytosis. Additionally, upon elevated oscillating hydrostatic pressure, YAP/TAZ are activated and induce TEAD-mediated transcription and expression of cellular components involved in dynamic regulation of cell volume and extracellular matrix. This cellular response confers a feedback loop that allows the cell to robustly respond to changes in interstitial fluid pressure.
Collapse
Affiliation(s)
- Jiwon Park
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Siyang Jia
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Donald Salter
- Centre for Genomic & Experimental MedicineMRC Institute of Genetics & Molecular MedicineThe University of Edinburgh, Western General HospitalEdinburghUK
| | - Pierre Bagnaninchi
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| | - Carsten G Hansen
- Centre for Inflammation ResearchInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
- Centre for Regenerative MedicineInstitute for Regeneration and Repair, Edinburgh bioQuarterThe University of EdinburghEdinburghUK
| |
Collapse
|
40
|
Yang Y, Santos DM, Pantano L, Knipe R, Abe E, Logue A, Pronzati G, Black KE, Spinney JJ, Giacona F, Bieler M, Godbout C, Nicklin P, Wyatt D, Tager AM, Seither P, Herrmann FE, Medoff BD. Screening for Inhibitors of YAP Nuclear Localization Identifies Aurora Kinase A as a Modulator of Lung Fibrosis. Am J Respir Cell Mol Biol 2022; 67:36-49. [PMID: 35377835 PMCID: PMC9798384 DOI: 10.1165/rcmb.2021-0428oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 04/04/2022] [Indexed: 01/01/2023] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive lung disease with limited therapeutic options that is characterized by pathological fibroblast activation and aberrant lung remodeling with scar formation. YAP (Yes-associated protein) is a transcriptional coactivator that mediates mechanical and biochemical signals controlling fibroblast activation. We previously identified HMG-CoA (3-hydroxy-3-methylglutaryl coenzyme A) reductase inhibitors (statins) as YAP inhibitors based on a high-throughput small-molecule screen in primary human lung fibroblasts. Here we report that several Aurora kinase inhibitors were also identified from the top hits of this screen. MK-5108, a highly selective inhibitor for AURKA (Aurora kinase A), induced YAP phosphorylation and cytoplasmic retention and significantly reduced profibrotic gene expression in human lung fibroblasts. The inhibitory effect on YAP nuclear translocation and profibrotic gene expression is specific to inhibition of AURKA, but not Aurora kinase B or C, and is independent of the Hippo pathway kinases LATS1 and LATS2 (Large Tumor Suppressor 1 and 2). Further characterization of the effects of MK-5108 demonstrate that it inhibits YAP nuclear localization indirectly via effects on actin polymerization and TGFβ (Transforming Growth Factor β) signaling. In addition, MK-5108 treatment reduced lung collagen deposition in the bleomycin mouse model of pulmonary fibrosis. Our results reveal a novel role for AURKA in YAP-mediated profibrotic activity in fibroblasts and highlight the potential of small-molecule screens for YAP inhibitors for identification of novel agents with antifibrotic activity.
Collapse
Affiliation(s)
- Yang Yang
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniela M Santos
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lorena Pantano
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Rachel Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Elizabeth Abe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amanda Logue
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Gina Pronzati
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Katharine E Black
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jillian J Spinney
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Francesca Giacona
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | - Andrew M Tager
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Franziska E Herrmann
- Immunology and Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Nguyen MT, Lee W. Kank1 Is Essential for Myogenic Differentiation by Regulating Actin Remodeling and Cell Proliferation in C2C12 Progenitor Cells. Cells 2022; 11:cells11132030. [PMID: 35805114 PMCID: PMC9265739 DOI: 10.3390/cells11132030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Actin cytoskeleton dynamics are essential regulatory processes in muscle development, growth, and regeneration due to their modulation of mechanotransduction, cell proliferation, differentiation, and morphological changes. Although the KN motif and ankyrin repeat domain-containing protein 1 (Kank1) plays a significant role in cell adhesion dynamics, actin polymerization, and cell proliferation in various cells, the functional significance of Kank1 during the myogenic differentiation of progenitor cells has not been explored. Here, we report that Kank1 acts as a critical regulator of the proliferation and differentiation of muscle progenitor cells. Kank1 was found to be expressed at a relatively high level in C2C12 myoblasts, and its expression was modulated during the differentiation. Depletion of Kank1 by siRNA (siKank1) increased the accumulation of filamentous actin (F-actin). Furthermore, it facilitated the nuclear localization of Yes-associated protein 1 (YAP1) by diminishing YAP1 phosphorylation in the cytoplasm, which activated the transcriptions of YAP1 target genes and promoted proliferation and cell cycle progression in myoblasts. Notably, depletion of Kank1 suppressed the protein expression of myogenic regulatory factors (i.e., MyoD and MyoG) and dramatically inhibited myoblast differentiation and myotube formation. Our results show that Kank1 is an essential regulator of actin dynamics, YAP1 activation, and cell proliferation and that its depletion impairs the myogenic differentiation of progenitor cells by promoting myoblast proliferation triggered by the F-actin-induced nuclear translocation of YAP1.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Gyeonggi-do, Goyang 10326, Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
42
|
Dupont S, Wickström SA. Mechanical regulation of chromatin and transcription. Nat Rev Genet 2022; 23:624-643. [DOI: 10.1038/s41576-022-00493-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 01/14/2023]
|
43
|
Pan D. The unfolding of the Hippo signaling pathway. Dev Biol 2022; 487:1-9. [PMID: 35405135 DOI: 10.1016/j.ydbio.2022.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022]
Abstract
The development of a functional organ requires not only patterning mechanisms that confer proper identities to individual cells, but also growth-regulatory mechanisms that specify the final size of the organ. At the turn of the 21st century, comprehensive genetic screens in model organisms had successfully uncovered the major signaling pathways that mediate pattern formation in metazoans. In contrast, signaling pathways dedicated to growth control were less explored. The past two decades has witnessed the emergence of the Hippo signaling pathway as a central mediator of organ size control through coordinated regulation of cell proliferation and apoptosis. Here I reflect on the early discoveries in Drosophila that elucidated the core kinase cascade and transcriptional machinery of the Hippo pathway, highlight its deep evolutionary conservation from humans to unicellular relatives of metazoan, and discuss the complex regulation of Hippo signaling by upstream inputs. This historical perspective underscores the importance of model organisms in uncovering fundamental and universal mechanisms of life processes.
Collapse
Affiliation(s)
- Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390-9040, USA.
| |
Collapse
|
44
|
Nguyen MT, Won YH, Kwon TW, Lee W. Twinfilin-1 is an essential regulator of myogenic differentiation through the modulation of YAP in C2C12 myoblasts. Biochem Biophys Res Commun 2022; 599:17-23. [DOI: 10.1016/j.bbrc.2022.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
|
45
|
Tsai CR, Martin JF. Hippo signaling in cardiac fibroblasts during development, tissue repair, and fibrosis. Curr Top Dev Biol 2022; 149:91-121. [PMID: 35606063 PMCID: PMC10898347 DOI: 10.1016/bs.ctdb.2022.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The evolutionarily conserved Hippo signaling pathway plays key roles in regulating the balance between cell proliferation and apoptosis, cell differentiation, organ size control, tissue repair, and regeneration. Recently, the Hippo pathway has been shown to regulate heart fibrosis, defined as excess extracellular matrix (ECM) deposition and increased tissue stiffness. Cardiac fibroblasts (CFs) are the primary cell type that produces, degrades, and remodels the ECM during homeostasis, aging, inflammation, and tissue repair and regeneration. Here, we review the available evidence from the current literature regarding how the Hippo pathway regulates the formation and function of CFs during heart development and tissue repair.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, United States.
| |
Collapse
|
46
|
Zhang T, He X, Caldwell L, Goru SK, Ulloa Severino L, Tolosa MF, Misra PS, McEvoy CM, Christova T, Liu Y, Atin C, Zhang J, Hu C, Vukosa N, Chen X, Krizova A, Kirpalani A, Gregorieff A, Ni R, Chan K, Gill MK, Attisano L, Wrana JL, Yuen DA. NUAK1 promotes organ fibrosis via YAP and TGF-β/SMAD signaling. Sci Transl Med 2022; 14:eaaz4028. [PMID: 35320001 DOI: 10.1126/scitranslmed.aaz4028] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibrosis is a central pathway that drives progression of multiple chronic diseases, yet few safe and effective clinical antifibrotic therapies exist. In most fibrotic disorders, transforming growth factor-β (TGF-β)-driven scarring is an important pathologic feature and a key contributor to disease progression. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two closely related transcription cofactors that are important for coordinating fibrogenesis after organ injury, but how they are activated in response to tissue injury has, so far, remained unclear. Here, we describe NUAK family kinase 1 (NUAK1) as a TGF-β-inducible profibrotic kinase that is up-regulated in multiple fibrotic organs in mice and humans. Mechanistically, we show that TGF-β induces a rapid increase in NUAK1 in fibroblasts. NUAK1, in turn, can promote profibrotic YAP and TGF-β/SMAD signaling, ultimately leading to organ scarring. Moreover, activated YAP and TAZ can induce further NUAK1 expression, creating a profibrotic positive feedback loop that enables persistent fibrosis. Using mouse models of kidney, lung, and liver fibrosis, we demonstrate that this fibrogenic signaling loop can be interrupted via fibroblast-specific loss of NUAK1 expression, leading to marked attenuation of fibrosis. Pharmacologic NUAK1 inhibition also reduced scarring, either when initiated immediately after injury or when initiated after fibrosis was already established. Together, our data suggest that NUAK1 plays a critical, previously unrecognized role in fibrogenesis and represents an attractive target for strategies that aim to slow fibrotic disease progression.
Collapse
Affiliation(s)
- Tianzhou Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Xiaolin He
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Lauren Caldwell
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Santosh Kumar Goru
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Luisa Ulloa Severino
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Monica F Tolosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Paraish S Misra
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Caitríona M McEvoy
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Tania Christova
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Yong Liu
- Ontario Institute of Cancer Research, Toronto, Ontario M5G OA3, Canada
| | - Cassandra Atin
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Johnny Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Catherine Hu
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Noah Vukosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Xiaolan Chen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Adriana Krizova
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Anish Kirpalani
- Department of Medical Imaging, St. Michael's Hospital (Unity Health Toronto) and University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Alex Gregorieff
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Ruoyu Ni
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Kin Chan
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Mandeep K Gill
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Liliana Attisano
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Darren A Yuen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
47
|
García-García M, Sánchez-Perales S, Jarabo P, Calvo E, Huyton T, Fu L, Ng SC, Sotodosos-Alonso L, Vázquez J, Casas-Tintó S, Görlich D, Echarri A, Del Pozo MA. Mechanical control of nuclear import by Importin-7 is regulated by its dominant cargo YAP. Nat Commun 2022; 13:1174. [PMID: 35246520 PMCID: PMC8897400 DOI: 10.1038/s41467-022-28693-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 01/19/2022] [Indexed: 12/31/2022] Open
Abstract
Mechanical forces regulate multiple essential pathways in the cell. The nuclear translocation of mechanoresponsive transcriptional regulators is an essential step for mechanotransduction. However, how mechanical forces regulate the nuclear import process is not understood. Here, we identify a highly mechanoresponsive nuclear transport receptor (NTR), Importin-7 (Imp7), that drives the nuclear import of YAP, a key regulator of mechanotransduction pathways. Unexpectedly, YAP governs the mechanoresponse of Imp7 by forming a YAP/Imp7 complex that responds to mechanical cues through the Hippo kinases MST1/2. Furthermore, YAP behaves as a dominant cargo of Imp7, restricting the Imp7 binding and the nuclear translocation of other Imp7 cargoes such as Smad3 and Erk2. Thus, the nuclear import process is an additional regulatory layer indirectly regulated by mechanical cues, which activate a preferential Imp7 cargo, YAP, which competes out other cargoes, resulting in signaling crosstalk. The translation of mechanical cues into gene expression changes is dependent on the nuclear import of mechanoresponsive transcriptional regulators. Here the authors identify that Importin-7 drives the nuclear import of one such regulator YAP while YAP then controls Importin-7 response to mechanical cues and restricts Importin-7 binding to other cargoes.
Collapse
Affiliation(s)
- María García-García
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Sara Sánchez-Perales
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Patricia Jarabo
- Instituto Cajal-CSIC, Avda. Doctor Arce, 37, 28002, Madrid, Spain
| | - Enrique Calvo
- Proteomics Unit. Area of Vascular Physiopathology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Trevor Huyton
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Liran Fu
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Sheung Chun Ng
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Jesús Vázquez
- Proteomics Unit. Area of Vascular Physiopathology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | | | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory. Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Calle Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
48
|
Abstract
Embryonic cells grow in environments that provide a plethora of physical cues, including mechanical forces that shape the development of the entire embryo. Despite their prevalence, the role of these forces in embryonic development and their integration with chemical signals have been mostly neglected, and scrutiny in modern molecular embryology tilted, instead, towards the dissection of molecular pathways involved in cell fate determination and patterning. It is now possible to investigate how mechanical signals induce downstream genetic regulatory networks to regulate key developmental processes in the embryo. Here, we review the insights into mechanical control of early vertebrate development, including the role of forces in tissue patterning and embryonic axis formation. We also highlight recent in vitro approaches using individual embryonic stem cells and self-organizing multicellular models of human embryos, which have been instrumental in expanding our understanding of how mechanics tune cell fate and cellular rearrangements during human embryonic development.
Collapse
|
49
|
Liao J, Zheng Y, Hu M, Xu P, Lin L, Liu X, Wu Y, Huang B, Ye X, Li S, Duan R, Fu H, Huang J, Wen L, Fu Y, Kilby MD, Kenny LC, Baker PN, Qi H, Tong C. Impaired Sphingosine-1-Phosphate Synthesis Induces Preeclampsia by Deactivating Trophoblastic YAP (Yes-Associated Protein) Through S1PR2 (Sphingosine-1-Phosphate Receptor-2)-Induced Actin Polymerizations. Hypertension 2021; 79:399-412. [PMID: 34865521 DOI: 10.1161/hypertensionaha.121.18363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized trophoblast cells) cell invasion in a Hippo-signaling-dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA/ROCK induced actin polymerization. Mutation-based YAP-5SA demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.
Collapse
Affiliation(s)
- Jiujiang Liao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yangxi Zheng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Department of Biochemistry & Molecular Biology, University of Texas McGovern Medical School at Houston (Y.Z.).,Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX (Y.Z.)
| | - Mingyu Hu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ping Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Lin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xiyao Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Biao Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xuan Ye
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Sisi Li
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ran Duan
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Huijia Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Jiayu Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Wen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yong Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Mark D Kilby
- Institute of Metabolism and System Research, College of Medical & Dental Sciences, University of Birmingham and the Fetal Medicine Centre, Birmingham Women's and Children's Foundation Trust, United Kingdom (M.D.K.)
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, United Kingdom (L.C.K.)
| | - Philip N Baker
- College of Life Sciences, University of Leicester, United Kingdom (P.N.B.)
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Chongqing Women and Children's Health Center, China (H.Q.)
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| |
Collapse
|
50
|
Ren Z, Zhang Z, Liu TM, Ge W. Novel zebrafish polycystic kidney disease models reveal functions of the Hippo pathway in renal cystogenesis. Dis Model Mech 2021; 14:272239. [PMID: 34545930 PMCID: PMC8592019 DOI: 10.1242/dmm.049027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/10/2021] [Indexed: 12/14/2022] Open
Abstract
The Hippo signaling pathway is a kinase cascade that plays an important role in organ size control. As the main effectors of the Hippo pathway, transcription coactivators Yap1/Wwtr1 are regulated by the upstream kinase Stk3. Recent studies in mammals have implicated the Hippo pathway in kidney development and kidney diseases. To further illustrate its roles in vertebrate kidney, we generated a series of zebrafish mutants targeting stk3, yap1 and wwtr1 genes. The stk3−/− mutant exhibited edema, formation of glomerular cysts and pronephric tubule dilation during the larval stage. Interestingly, disruption of wwtr1, but not yap1, significantly alleviated the renal phenotypes of the stk3−/− mutant, and overexpression of Wwtr1 with the CMV promoter also induced pronephric phenotypes, similar to those of the stk3−/− mutant, during larval stage. Notably, adult fish with Wwtr1 overexpression developed phenotypes similar to those of human polycystic kidney disease (PKD). Overall, our analyses revealed roles of Stk3 and Wwtr1 in renal cyst formation. Using a pharmacological approach, we further demonstrated that Stk3-deficient zebrafish could serve as a PKD model for drug development. Summary: A zebrafish stk3 mutant line and Wwtr1 overexpression line provide evidence for functions of the Hippo signaling pathway in renal cyst formation and represent potential models for polycystic kidney disease.
Collapse
Affiliation(s)
- Zhiqin Ren
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Zhiwei Zhang
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Tzu-Ming Liu
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| |
Collapse
|