1
|
Choi SH, Kim DY. Regulation of Tumor Microenvironment through YAP/TAZ under Tumor Hypoxia. Cancers (Basel) 2024; 16:3030. [PMID: 39272887 PMCID: PMC11394240 DOI: 10.3390/cancers16173030] [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: 07/16/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
In solid tumors such as hepatocellular carcinoma (HCC), hypoxia is one of the important mechanisms of cancer development that closely influences cancer development, survival, and metastasis. The development of treatments for cancer was temporarily revolutionized by immunotherapy but continues to be constrained by limited response rates and the resistance and high costs required for the development of new and innovative strategies. In particular, solid tumors, including HCC, a multi-vascular tumor type, are sensitive to hypoxia and generate many blood vessels for metastasis and development, making it difficult to treat HCC, not only with immunotherapy but also with drugs targeting blood vessels. Therefore, in order to develop a treatment strategy for hypoxic tumors, various mechanisms must be explored and analyzed to treat these impregnable solid tumors. To date, tumor growth mechanisms linked to hypoxia are known to be complex and coexist with various signal pathways, but recently, mechanisms related to the Hippo signal pathway are emerging. Interestingly, Hippo YAP/TAZ, which appear during early tumor and normal tumor growth, and YAP/TAZ, which appear during hypoxia, help tumor growth and proliferation in different directions. Peculiarly, YAP/TAZ, which have different phosphorylation directions in the hypoxic environment of tumors, are involved in cancer proliferation and metastasis in various carcinomas, including HCC. Analyzing the mechanisms that regulate the function and expression of YAP in addition to HIF in the complex hypoxic environment of tumors may lead to a variety of anti-cancer strategies and combining HIF and YAP/TAZ may develop the potential to change the landscape of cancer treatment.
Collapse
Affiliation(s)
- Sung Hoon Choi
- Institute of Health & Environment, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs Inc., Seoul 08826, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea
| |
Collapse
|
2
|
Lee CH, Lee SH, Kwak HS, Kwak YG, Rosenson RS, Cho YI, Jeong SK. Validation of Signal Intensity Gradient from TOF-MRA for Wall Shear Stress by Phase-Contrast MR. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1248-1258. [PMID: 38332403 PMCID: PMC11169296 DOI: 10.1007/s10278-024-00991-5] [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: 10/27/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 02/10/2024]
Abstract
To validate the correlation between the signal intensity gradient (SIG) from time-of-flight magnetic resonance angiography (TOF-MRA) and wall shear stress (WSS) determined by phase contrast magnetic resonance (PC-MR), we conducted both experimental and human studies. In the experimental study, we measured WSS in four tubes of different sizes with variable flow rates using PC-MR and TOF-MRA. The flow rates of water in the experimental study ranged from 0.06 to 12.75 mL/s, resulting in PC-WSS values between 0.1 and 1.6 dyne/cm2. The correlation between PC-WSS and SIG was statistically significant, showing a coefficient of 0.86 (P < 0.001, R2 = 0.75). The line fit provided the conversion equation as Y = 1.6287X - 1.1563 (Y = PC-WSS, X = SIG). For the human study, 28 subjects underwent TOF-MRA and PC-MR examinations of carotid and vertebral arteries. Arterial PC-WSS and SIG were determined in the same segment for each subject. The arterial PC-WSS ranged from 1.9 to 21.0 dyne/cm2. Both carotid and vertebral arteries showed significant correlations between PC-WSS and SIG, with coefficients of 0.85, 0.86, 0.91, and 0.81 in the right and left carotid and vertebral arteries, respectively. Our results show that SIG from TOF-MRA and SIG-WSS derived from the conversion equation provide concurrent in vivo hemodynamic information on arterial shear stress. This study was registered on ClinicalTrials.gov with the identifier NCT04585971 on October 14, 2020.
Collapse
Affiliation(s)
- Chan-Hyuk Lee
- Department of Neurology, Asan Medical Center, Seoul, Republic of Korea
- Department of Neurology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Sang Hyuk Lee
- Equipment Qualification Center for Nuclear Power Plants, Korea Institute of Machinery and Materials, Daejeon, Republic of Korea
| | - Hyo-Sung Kwak
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Yeong-Gon Kwak
- Department of Radiotechnology, Wonkwang Health Science University, Iksan, Republic of Korea
| | - Robert S Rosenson
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Young I Cho
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, PA, USA
| | - Seul-Ki Jeong
- Seul-Ki Jeong Neurology Clinic, 233, Gucheonmyeon-ro, Gangdong-gu, Seoul, 05326, Republic of Korea.
| |
Collapse
|
3
|
Kobayashi S, Cox AG, Harvey KF, Hogan BM. Vasculature is getting Hip(po): Hippo signaling in vascular development and disease. Dev Cell 2023; 58:2627-2640. [PMID: 38052179 DOI: 10.1016/j.devcel.2023.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/29/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023]
Abstract
The Hippo signaling pathway regulates developmental organ growth, regeneration, and cell fate decisions. Although the role of the Hippo pathway, and its transcriptional effectors YAP and TAZ, has been well documented in many cell types and species, only recently have the roles for this pathway come to light in vascular development and disease. Experiments in mice, zebrafish, and in vitro have uncovered roles for the Hippo pathway, YAP, and TAZ in vasculogenesis, angiogenesis, and lymphangiogenesis. In addition, the Hippo pathway has been implicated in vascular cancers and cardiovascular diseases, thus identifying it as a potential therapeutic target for the treatment of these conditions. However, despite recent advances, Hippo's role in the vasculature is still underappreciated compared with its role in epithelial tissues. In this review, we appraise our current understanding of the Hippo pathway in blood and lymphatic vessel development and highlight the current knowledge gaps and opportunities for further research.
Collapse
Affiliation(s)
- Sakurako Kobayashi
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew G Cox
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kieran F Harvey
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
4
|
YAP Overexpression in Breast Cancer Cells Promotes Angiogenesis through Activating YAP Signaling in Vascular Endothelial Cells. Anal Cell Pathol (Amst) 2022; 2022:5942379. [PMID: 36226237 PMCID: PMC9550503 DOI: 10.1155/2022/5942379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/16/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose. The YAP signaling pathway is altered and implicated as oncogenic in human mammary cancers. However, roles of YAP signaling that regulate the breast tumor angiogenesis have remained elusive. Tumor angiogenesis is coordinated by the activation of both cancer cells and vascular endothelial cells. Whether the YAP signaling pathway can regulate the intercellular interaction between cancer cells and endothelial cells is essentially unknown. Methods. The effects of YAP on tumor angiogenesis, migration, and proliferation of vascular endothelial cells were evaluated in vitro. Expression of proteins and phosphorylating proteins involved in YAP, G13-RhoA, and PI3K/Akt signaling pathways was evaluated using the Western blotting, immunofluorescence staining, and immunohistochemistry analysis. In addition, the effects of YAP on breast cancer angiogenesis were evaluated in vivo by tumor xenograft mice. Results. We showed here that conditioned media from YAP overexpressed breast cancer cells (CM-YAP+) could promote angiogenesis, accompanied by increased tube formation, migration, and proliferation of human umbilical vein endothelial cells (HUVECs). Down regulation of YAP in HUVECs reversed CM-YAP+ induced angiogenesis. CM-YAP+ time-dependently activated YAP in HUVECs by dephosphorylating YAP and increasing nuclear translocation. We also identified that both G13-RhoA and PI3K/Akt signaling pathway were necessary for CM-YAP+ induced activation of YAP. Besides, connective tissue growth factor (CTGF) and angiopoietin-2 (ANG-2) acted as down-stream of YAP in HUVECs to promote angiogenesis. In addition, subcutaneous tumors nude mice model demonstrated that tumors overexpressed YAP revealed more neovascularization in vivo. Conclusion. YAP-YAP interaction between breast cancer cells and endothelial cells could promote tumor angiogenesis, supporting that YAP is a potential marker and target for developing novel therapeutic strategies against breast cancer.
Collapse
|
5
|
Grundy TJ, Orcheston-Findlay L, de Silva E, Jegathees T, Prior V, Sarker FA, O'Neill GM. Mechanosensitive expression of the mesenchymal subtype marker connective tissue growth factor in glioblastoma. Sci Rep 2022; 12:14982. [PMID: 36056123 PMCID: PMC9440209 DOI: 10.1038/s41598-022-19175-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/25/2022] [Indexed: 12/05/2022] Open
Abstract
Mechanical forces created by the extracellular environment regulate biochemical signals that modulate the inter-related cellular phenotypes of morphology, proliferation, and migration. A stiff microenvironment induces glioblastoma (GBM) cells to develop prominent actin stress fibres, take on a spread morphology and adopt trapezoid shapes, when cultured in 2D, which are phenotypes characteristic of a mesenchymal cell program. The mesenchymal subtype is the most aggressive among the molecular GBM subtypes. Recurrent GBM have been reported to transition to mesenchymal. We therefore sought to test the hypothesis that stiffer microenvironments-such as those found in different brain anatomical structures and induced following treatment-contribute to the expression of markers characterising the mesenchymal subtype. We cultured primary patient-derived cell lines that reflect the three common GBM subtypes (mesenchymal, proneural and classical) on polyacrylamide (PA) hydrogels with controlled stiffnesses spanning the healthy and pathological tissue range. We then assessed the canonical mesenchymal markers Connective Tissue Growth Factor (CTGF) and yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) expression, via immunofluorescence. Replating techniques and drug-mediated manipulation of the actin cytoskeleton were utilised to ascertain the response of the cells to differing mechanical environments. We demonstrate that CTGF is induced rapidly following adhesion to a rigid substrate and is independent of actin filament formation. Collectively, our data suggest that microenvironmental rigidity can stimulate expression of mesenchymal-associated molecules in GBM.
Collapse
Affiliation(s)
- Thomas James Grundy
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Louise Orcheston-Findlay
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Eshana de Silva
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
| | - Thuvarahan Jegathees
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, 2006, Australia
| | - Victoria Prior
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, 2006, Australia
| | - Farhana Amy Sarker
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, 2006, Australia
| | - Geraldine Margaret O'Neill
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead, Westmead, NSW, 2145, Australia.
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, 2006, Australia.
| |
Collapse
|
6
|
Fabris L, Milani C, Fiorotto R, Mariotti V, Kaffe E, Seller B, Sonzogni A, Strazzabosco M, Cadamuro M. Dysregulation of the Scribble/YAP/β-catenin axis sustains the fibroinflammatory response in a PKHD1 -/- mouse model of congenital hepatic fibrosis. FASEB J 2022; 36:e22364. [PMID: 35593740 PMCID: PMC9150862 DOI: 10.1096/fj.202101924r] [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: 12/17/2021] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/11/2022]
Abstract
Congenital hepatic fibrosis (CHF), a genetic cholangiopathy characterized by fibropolycystic changes in the biliary tree, is caused by mutations in the PKHD1 gene, leading to defective fibrocystin (FPC), changes in planar cell polarity (PCP) and increased β-catenin-dependent chemokine secretion. In this study, we aimed at understanding the role of Scribble (a protein involved in PCP), Yes-associated protein (YAP), and β-catenin in the regulation of the fibroinflammatory phenotype of FPC-defective cholangiocytes. Immunohistochemistry showed that compared with wild type (WT) mice, in FPC-defective (Pkhd1del4/del4 ) mice nuclear expression of YAP/TAZ in cystic cholangiocytes, significantly increased and correlated with connective tissue growth factor (CTGF) expression and pericystic fibrosis, while Scribble expression on biliary cyst cells was markedly decreased. Cholangiocytes isolated from WT mice showed intense Scribble immunoreactivity at the membrane, but minimal nuclear expression of YAP, which conversely increased, together with CTGF, after small interfering RNA (siRNA) silencing of Scribble. In FPC-defective cholangiocytes, inhibition of YAP nuclear import reduced β-catenin nuclear expression, and CTGF, integrin β6, CXCL1, and CXCL10 mRNA levels, whereas inhibition of β-catenin signaling did not affect nuclear translocation of YAP. Notably, siRNA silencing of Scribble and YAP in WT cholangiocytes mimics the fibroinflammatory changes of FPC-defective cholangiocytes. Conditional deletion of β-catenin in Pkhd1del4/del4 mice reduced cyst growth, inflammation and fibrosis, without affecting YAP nuclear expression. In conclusion, the defective anchor of Scribble to the membrane facilitates the nuclear translocation of YAP and β-catenin with gain of a fibroinflammatory phenotype. The Scribble/YAP/β-catenin axis is a critical factor in the sequence of events linking the genetic defect to fibrocystic trait of cholangiocytes in CHF.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Molecular Medicine (DMM), University of Padova, Padova, Italy
- International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy
- Liver Center, Department of Internal Medicine, Yale University, New Haven (CT), US
| | - Chiara Milani
- School of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Romina Fiorotto
- Liver Center, Department of Internal Medicine, Yale University, New Haven (CT), US
| | - Valeria Mariotti
- Department of Molecular Medicine (DMM), University of Padova, Padova, Italy
- Liver Center, Department of Internal Medicine, Yale University, New Haven (CT), US
| | - Eleanna Kaffe
- Liver Center, Department of Internal Medicine, Yale University, New Haven (CT), US
| | - Barbara Seller
- School of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Aurelio Sonzogni
- Department of Pathology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mario Strazzabosco
- Liver Center, Department of Internal Medicine, Yale University, New Haven (CT), US
- Corresponding authors: Mario Strazzabosco, MD, PhD, Department of Internal Medicine, Yale University School of Medicine, Cedar Street 333 Room LMP1080, New Haven, CT 06517, USA. Phone: +1‐203‐785‐5110, , Massimiliano Cadamuro, PhD, Department of Molecular Medicine, University of Padova, Gabelli Street 63, Padova, 35121, Italy. Phone: +39-049-827-6113,
| | - Massimiliano Cadamuro
- Department of Molecular Medicine (DMM), University of Padova, Padova, Italy
- International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy
- Corresponding authors: Mario Strazzabosco, MD, PhD, Department of Internal Medicine, Yale University School of Medicine, Cedar Street 333 Room LMP1080, New Haven, CT 06517, USA. Phone: +1‐203‐785‐5110, , Massimiliano Cadamuro, PhD, Department of Molecular Medicine, University of Padova, Gabelli Street 63, Padova, 35121, Italy. Phone: +39-049-827-6113,
| |
Collapse
|
7
|
Riley SE, Feng Y, Hansen CG. Hippo-Yap/Taz signalling in zebrafish regeneration. NPJ Regen Med 2022; 7:9. [PMID: 35087046 PMCID: PMC8795407 DOI: 10.1038/s41536-022-00209-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
The extent of tissue regeneration varies widely between species. Mammals have a limited regenerative capacity whilst lower vertebrates such as the zebrafish (Danio rerio), a freshwater teleost, can robustly regenerate a range of tissues, including the spinal cord, heart, and fin. The molecular and cellular basis of this altered response is one of intense investigation. In this review, we summarise the current understanding of the association between zebrafish regeneration and Hippo pathway function, a phosphorylation cascade that regulates cell proliferation, mechanotransduction, stem cell fate, and tumorigenesis, amongst others. We also compare this function to Hippo pathway activity in the regenerative response of other species. We find that the Hippo pathway effectors Yap/Taz facilitate zebrafish regeneration and that this appears to be latent in mammals, suggesting that therapeutically promoting precise and temporal YAP/TAZ signalling in humans may enhance regeneration and hence reduce morbidity.
Collapse
Affiliation(s)
- Susanna E Riley
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Yi Feng
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Carsten Gram Hansen
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
8
|
Distinctive Roles of YAP and TAZ in Human Endothelial Progenitor Cells Growth and Functions. Biomedicines 2022; 10:biomedicines10010147. [PMID: 35052826 PMCID: PMC8773510 DOI: 10.3390/biomedicines10010147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
The hippo signaling pathway plays an essential role in controlling organ size and balancing tissue homeostasis. Its two main effectors, yes-associated protein (YAP) and WW domain-containing transcription regulator 1, WWTR1 or TAZ, have also been shown to regulate endothelial cell functions and angiogenesis. In this study, the functions of YAP and TAZ in human endothelial progenitor cells (EPCs) were investigated by a loss-of-function study using CRISPR/Cas9-mediated gene knockdown (KD). Depletion of either YAP or TAZ reduced EPC survival and impaired many of their critical functions, including migration, invasion, vessel-formation, and expression of pro-angiogenic genes. Notably, TAZ-KD EPCs exhibited more severe phenotypes in comparison to YAP-KD EPCs. Moreover, the conditioned medium derived from TAZ-KD EPCs reduced the survivability of human lung cancer cells and increased their sensitivity to chemotherapeutic agents. The overexpression of either wild-type or constitutively active TAZ rescued the impaired phenotypes of TAZ-KD EPCs and restored the expression of pro-angiogenic genes in those EPCs. In summary, we demonstrate the crucial role of Hippo signaling components, YAP and TAZ, in controlling several aspects of EPC functions that can potentially be used as a drug target to enhance EPC functions in patients.
Collapse
|
9
|
Gong S, Ma H, Zheng F, Huang J, Zhang Y, Yu B, Li F, Kou J. Inhibiting YAP in Endothelial Cells From Entering the Nucleus Attenuates Blood-Brain Barrier Damage During Ischemia-Reperfusion Injury. Front Pharmacol 2021; 12:777680. [PMID: 34899341 PMCID: PMC8662521 DOI: 10.3389/fphar.2021.777680] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-brain barrier (BBB) damage is a critical event in ischemic stroke, contributing to aggravated brain damage. Endothelial cell form a major component of the BBB, but its regulation in stroke has yet to be clarified. We investigated the function of Yes-associated protein 1 (YAP) in the endothelium on BBB breakdown during cerebral ischemia/reperfusion (I/R) injury. The effects of YAP on BBB dysfunction were explored in middle cerebral artery occlusion/reperfusion (MCAO/R)-injury model mice and using brain microvascular endothelial cells (BMEC) exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) injury. The degree of brain injury was estimated using staining (2,3,5-Triphenyltetrazolium chloride, hematoxylin and eosin) and the detection of cerebral blood flow. BBB breakdown was investigated by examining the leakage of Evans Blue dye and evaluating the expression of tight junction (TJ)-associated proteins and matrix metallopeptidase (MMP) 2 and 9. YAP expression was up-regulated in the nucleus of BMEC after cerebral I/R injury. Verteporfin (YAP inhibitor) down-regulated YAP expression in the nucleus and improved BBB hyperpermeability and TJ integrity disruption stimulated by cerebral I/R. YAP-targeted small interfering RNA (siRNA) exerted the same effects in BMEC cells exposed to OGD/R injury. Our findings provide new insights into the contributions made by YAP to the maintenance of BBB integrity and highlight the potential for YAP to serve as a therapeutic target to modulate BBB integrity following ischemic stroke and related cerebrovascular diseases.
Collapse
Affiliation(s)
- Shuaishuai Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huifen Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fan Zheng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Juan Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Boyang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Junping Kou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medical, School of Traditional Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
10
|
Okasato R, Kano K, Kise R, Inoue A, Fukuhara S, Aoki J. An ATX-LPA 6-Gα 13-ROCK axis shapes and maintains caudal vein plexus in zebrafish. iScience 2021; 24:103254. [PMID: 34755093 PMCID: PMC8564058 DOI: 10.1016/j.isci.2021.103254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/06/2021] [Accepted: 10/08/2021] [Indexed: 12/31/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a potential regulator of vascular formation derived from blood. In this study, we utilized zebrafish as a model organism to monitor the blood vessel formation in detail. Zebrafish mutant of ATX, an LPA-producing enzyme, had a defect in the caudal vein plexus (CVP). Pharmacological inhibition of ATX resulted in a fusion of the delicate vessels in the CVP to form large sac-like vessels. Mutant embryos of LPA6 receptor and downstream Gα13 showed the same phenotype. Administration of OMPT, a stable LPA-analog, induced rapid CVP constriction, which was attenuated significantly in the LPA6 mutant. We also found that blood flow-induced CVP formation was dependent on ATX. The present study demonstrated that the ATX-LPA6 axis acts cooperatively with blood flow and contributes to the formation and maintenance of the CVP by generating contractive force in endothelial cells. Blocking an ATX-LPA6-Gα13-ROCK axis causes malformation of the caudal vein plexus The axis also contributes to maintaining the fine structure of the caudal vein plexus Activation of LPA6 induces vasoconstriction Caudal vein plexus formation evoked by blood flow is dependent on an ATX-LPA6 axis
Collapse
Affiliation(s)
- Ryohei Okasato
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.,AMED-LEAP, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.,AMED-LEAP, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.,AMED-LEAP, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.,Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan.,AMED-LEAP, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
11
|
Wang M, Dai M, Wang D, Tang T, Xiong F, Xiang B, Zhou M, Li X, Li Y, Xiong W, Li G, Zeng Z, Guo C. The long noncoding RNA AATBC promotes breast cancer migration and invasion by interacting with YBX1 and activating the YAP1/Hippo signaling pathway. Cancer Lett 2021; 512:60-72. [PMID: 33951538 DOI: 10.1016/j.canlet.2021.04.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022]
Abstract
Long noncoding RNAs (lncRNAs) play an important role in the regulation of gene expression and are involved in several pathological responses. However, many important lncRNAs in breast cancer have not been identified and their expression levels and functions in breast cancer remain unknown. In this study, the lncRNA apoptosis-associated transcript in bladder cancer (AATBC) was found to be significantly highly expressed in breast cancer patients. In vitro and in vivo experiments indicated that AATBC promoted breast cancer metastasis. Further studies revealed that AATBC activated the YAP1/Hippo signaling pathway through the AATBC-YBX1-MST1 axis. This is also an important supplement to the composition of the YAP1/Hippo signaling pathway. The model of "AATBC-YAP1" may bring a new dawn to the treatment of breast cancer.
Collapse
Affiliation(s)
- Maonan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manli Dai
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ting Tang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Comprehensive Cancer Center Baylor College of Medicine, Houston, TX, USA
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Leguit RJ, Raymakers RAP, Hebeda KM, Goldschmeding R. CCN2 (Cellular Communication Network factor 2) in the bone marrow microenvironment, normal and malignant hematopoiesis. J Cell Commun Signal 2021; 15:25-56. [PMID: 33428075 PMCID: PMC7798015 DOI: 10.1007/s12079-020-00602-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
CCN2, formerly termed Connective Tissue Growth Factor, is a protein belonging to the Cellular Communication Network (CCN)-family of secreted extracellular matrix-associated proteins. As a matricellular protein it is mainly considered to be active as a modifier of signaling activity of several different signaling pathways and as an orchestrator of their cross-talk. Furthermore, CCN2 and its fragments have been implicated in the regulation of a multitude of biological processes, including cell proliferation, differentiation, adhesion, migration, cell survival, apoptosis and the production of extracellular matrix products, as well as in more complex processes such as embryonic development, angiogenesis, chondrogenesis, osteogenesis, fibrosis, mechanotransduction and inflammation. Its function is complex and context dependent, depending on cell type, state of differentiation and microenvironmental context. CCN2 plays a role in many diseases, especially those associated with fibrosis, but has also been implicated in many different forms of cancer. In the bone marrow (BM), CCN2 is highly expressed in mesenchymal stem/stromal cells (MSCs). CCN2 is important for MSC function, supporting its proliferation, migration and differentiation. In addition, stromal CCN2 supports the maintenance and longtime survival of hematopoietic stem cells, and in the presence of interleukin 7, stimulates the differentiation of pro-B lymphocytes into pre-B lymphocytes. Overexpression of CCN2 is seen in the majority of B-acute lymphoblastic leukemias, especially in certain cytogenetic subgroups associated with poor outcome. In acute myeloid leukemia, CCN2 expression is increased in MSCs, which has been associated with leukemic engraftment in vivo. In this review, the complex function of CCN2 in the BM microenvironment and in normal as well as malignant hematopoiesis is discussed. In addition, an overview is given of data on the remaining CCN family members regarding normal and malignant hematopoiesis, having many similarities and some differences in their function.
Collapse
Affiliation(s)
- Roos J. Leguit
- Department of Pathology, University Medical Center Utrecht, H04-312, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Reinier A. P. Raymakers
- Department of Hematology, UMCU Cancer Center, Heidelberglaan 100 B02.226, 3584 CX Utrecht, The Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
13
|
Hooglugt A, van der Stoel MM, Boon RA, Huveneers S. Endothelial YAP/TAZ Signaling in Angiogenesis and Tumor Vasculature. Front Oncol 2021; 10:612802. [PMID: 33614496 PMCID: PMC7890025 DOI: 10.3389/fonc.2020.612802] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
Solid tumors are dependent on vascularization for their growth. The hypoxic, stiff, and pro-angiogenic tumor microenvironment induces angiogenesis, giving rise to an immature, proliferative, and permeable vasculature. The tumor vessels promote tumor metastasis and complicate delivery of anti-cancer therapies. In many types of tumors, YAP/TAZ activation is correlated with increased levels of angiogenesis. In addition, endothelial YAP/TAZ activation is important for the formation of new blood and lymphatic vessels during development. Oncogenic activation of YAP/TAZ in tumor cell growth and invasion has been studied in great detail, however the role of YAP/TAZ within the tumor endothelium remains insufficiently understood, which complicates therapeutic strategies aimed at targeting YAP/TAZ in cancer. Here, we overview the upstream signals from the tumor microenvironment that control endothelial YAP/TAZ activation and explore the role of their downstream targets in driving tumor angiogenesis. We further discuss the potential for anti-cancer treatments and vascular normalization strategies to improve tumor therapies.
Collapse
Affiliation(s)
- Aukie Hooglugt
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
| | - Miesje M. van der Stoel
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Reinier A. Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Berlin, Germany
- Institute of Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Santamaría R, González-Álvarez M, Delgado R, Esteban S, Arroyo AG. Remodeling of the Microvasculature: May the Blood Flow Be With You. Front Physiol 2020; 11:586852. [PMID: 33178049 PMCID: PMC7593767 DOI: 10.3389/fphys.2020.586852] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
The vasculature ensures optimal delivery of nutrients and oxygen throughout the body, and to achieve this function it must continually adapt to varying tissue demands. Newly formed vascular plexuses during development are immature and require dynamic remodeling to generate well-patterned functional networks. This is achieved by remodeling of the capillaries preserving those which are functional and eliminating other ones. A balanced and dynamically regulated capillary remodeling will therefore ensure optimal distribution of blood and nutrients to the tissues. This is particularly important in pathological contexts in which deficient or excessive vascular remodeling may worsen tissue perfusion and hamper tissue repair. Blood flow is a major determinant of microvascular reshaping since capillaries are pruned when relatively less perfused and they split when exposed to high flow in order to shape the microvascular network for optimal tissue perfusion and oxygenation. The molecular machinery underlying blood flow sensing by endothelial cells is being deciphered, but much less is known about how this translates into endothelial cell responses as alignment, polarization and directed migration to drive capillary remodeling, particularly in vivo. Part of this knowledge is theoretical from computational models since blood flow hemodynamics are not easily recapitulated by in vitro or ex vivo approaches. Moreover, these events are difficult to visualize in vivo due to their infrequency and briefness. Studies had been limited to postnatal mouse retina and vascular beds in zebrafish but new tools as advanced microscopy and image analysis are strengthening our understanding of capillary remodeling. In this review we introduce the concept of remodeling of the microvasculature and its relevance in physiology and pathology. We summarize the current knowledge on the mechanisms contributing to capillary regression and to capillary splitting highlighting the key role of blood flow to orchestrate these processes. Finally, we comment the potential and possibilities that microfluidics offers to this field. Since capillary remodeling mechanisms are often reactivated in prevalent pathologies as cancer and cardiovascular disease, all this knowledge could be eventually used to improve the functionality of capillary networks in diseased tissues and promote their repair.
Collapse
Affiliation(s)
- Ricardo Santamaría
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María González-Álvarez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Raquel Delgado
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergio Esteban
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alicia G. Arroyo
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
15
|
Wisniewski L, French V, Lockwood N, Valdivia LE, Frankel P. P130Cas/bcar1 mediates zebrafish caudal vein plexus angiogenesis. Sci Rep 2020; 10:15589. [PMID: 32973180 PMCID: PMC7518251 DOI: 10.1038/s41598-020-71753-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
P130CAS/BCAR1 belongs to the CAS family of adaptor proteins, with important regulatory roles in cell migration, cell cycle control, and apoptosis. Previously, we and others showed that P130CAS mediates VEGF-A and PDGF signalling in vitro, but its cardiovascular function in vivo remains relatively unexplored. We characterise here a novel deletion model of P130CAS in zebrafish. Using in vivo microscopy and transgenic vascular reporters, we observed that while bcar1−/− zebrafish showed no arterial angiogenic or heart defects during development, they strikingly failed to form the caudal vein plexus (CVP). Endothelial cells (ECs) within the CVP of bcar1−/− embryos produced fewer filopodial structures and did not detach efficiently from neighbouring cells, resulting in a significant reduction in ventral extension and overall CVP area. Mechanistically, we show that P130Cas mediates Bmp2b-induced ectopic angiogenic sprouting of ECs in the developing embryo and provide pharmacological evidence for a role of Src family kinases in CVP development.
Collapse
Affiliation(s)
- Laura Wisniewski
- Division of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK. .,Queen Mary University of London, London, EC1M 6BQ, UK.
| | - Vanessa French
- Institute of Cardiovascular Science, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Nicola Lockwood
- Division of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK.,The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Leonardo E Valdivia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Paul Frankel
- Institute of Cardiovascular Science, University College London, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
16
|
Jin J, Zhao X, Fu H, Gao Y. The Effects of YAP and Its Related Mechanisms in Central Nervous System Diseases. Front Neurosci 2020; 14:595. [PMID: 32676008 PMCID: PMC7333666 DOI: 10.3389/fnins.2020.00595] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022] Open
Abstract
Yes-associated protein (YAP) is a key effector downstream of the Hippo signaling pathway and plays an important role in the development of the physiology and pathology of the central nervous system (CNS), especially regulating cell proliferation, differentiation, migration, and apoptosis. However, the roles and underlying mechanisms of YAP in CNS diseases are still puzzling. Here, this review will systematically and comprehensively summarize the biological feature, pathological role, and underlying mechanisms of YAP in normal and pathologic CNS, which aims to provide insights into the potential molecular targets and new therapeutic strategies for CNS diseases.
Collapse
Affiliation(s)
- Jiayan Jin
- Department of Forensic Science, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.,School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaoxuan Zhao
- Department of Forensic Science, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.,School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huifang Fu
- Department of Forensic Science, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Traditional Chinese Medicine Hospital of Jiangning District, Nanjing, China
| | - Yuan Gao
- Department of Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China.,Department of Forensic Science, School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.,Forensic Center, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Plewes MR, Hou X, Zhang P, Liang A, Hua G, Wood JR, Cupp AS, Lv X, Wang C, Davis JS. Yes-associated protein 1 is required for proliferation and function of bovine granulosa cells in vitro†. Biol Reprod 2019; 101:1001-1017. [PMID: 31350850 PMCID: PMC6877782 DOI: 10.1093/biolre/ioz139] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/28/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Yes-associated protein 1 (YAP1) is a major component of the Hippo signaling pathway. Although the exact extracellular signals that control the Hippo pathway are currently unknown, increasing evidence supports a critical role for the Hippo pathway in embryonic development, regulation of organ size, and carcinogenesis. Granulosa cells (GCs) within the ovarian follicle proliferate and produce steroids and growth factors, which facilitate the growth of follicle and maturation of the oocyte. We hypothesize that YAP1 plays a role in proliferation and estrogen secretion of GCs. In the current study, we examined the expression of the Hippo signaling pathway in bovine ovaries and determined whether it was important for GC proliferation and estrogen production. Mammalian STE20-like protein kinase 1 (MST1) and large tumor suppressor kinase 2 (LATS2) were identified as prominent upstream components of the Hippo pathway expressed in granulosa and theca cells of the follicle and large and small cells of the corpus luteum. Immunohistochemistry revealed that YAP1 was localized to the nucleus of growing follicles. In vitro, nuclear localization of the downstream Hippo signaling effector proteins YAP1 and transcriptional co-activator with PDZ-binding motif (TAZ) was inversely correlated with GC density, with greater nuclear localization under conditions of low cell density. Treatment with verteporfin and siRNA targeting YAP1 or TAZ revealed a critical role for these transcriptional co-activators in GC proliferation. Furthermore, knockdown of YAP1 in GCs inhibited follicle-stimulating hormone (FSH)-induced estradiol biosynthesis. The data indicate that Hippo pathway transcription co-activators YAP1/TAZ play an important role in GC proliferation and estradiol synthesis, two processes necessary for maintaining normal follicle development.
Collapse
Affiliation(s)
- Michele R Plewes
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, USA
| | - Xiaoying Hou
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pan Zhang
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aixin Liang
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guohua Hua
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jennifer R Wood
- Department of Animal Sciences, University of Nebraska–Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Sciences, University of Nebraska–Lincoln, Lincoln, NE, USA
| | - Xiangmin Lv
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
18
|
Walsh T. Editor’s Pick: Systemic Sclerosis: The Role of YAP/TAZ in Disease Pathogenesis. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10310340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Systemic sclerosis (SSc) is a systemic autoimmune condition of unknown cause. Yes-Associated Protein/Tafazzin (YAP/TAZ) are transcriptional coactivators previously demonstrated to be involved in cellular stretch biology, and form the principal effector molecules of the Hippo signalling pathway. The association between YAP/TAZ and stretch is contingent upon their cytoplasmic localisation (with nuclear translocation, the cell adopts a relaxed state). The author weighs the evidence for a central role for YAP/TAZ signalling in scleroderma spanning the major clinical features of the condition. Several of the features unique to SSc are mediated by cytoplasmic localisation of YAP/TAZ, including the stretch phenotype (through binding to NF-2), arterial lumenal obliteration (through their binding to angiomotin), the promotion of hypergammaglobulinaemia (via feedback to the upstream Hippo signalling molecule Mammalian Ste20-like Kinase 1), and the induction of B-Lymphocyte-Induced Maturation Protein-1 leading to the adoption of Th2 lineage, prominent in SSc. One observes that the induction of the fibrotic phenotype of scleroderma is mediated through GLI1/GLI2 (the effector molecules of the Hedgehog pathway). GLI1/GLI2 are induced to reciprocally enter the nucleus when YAP/TAZ is intracytoplasmic. The latter explains the characteristically increased connective tissue growth factor 2 and endothelin-1 expression. In this article, the author references some examples of the role of YAP/TAZ in the biophysically similar condition nephrogenic systemic fibrosis and suggests a role of YAP/TAZ cytoplasmic sequestration in programmed cell death protein 1-ligand antagonist-induced scleroderma.
Collapse
Affiliation(s)
- Thomas Walsh
- Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| |
Collapse
|
19
|
Santoro MM, Beltrame M, Panáková D, Siekmann AF, Tiso N, Venero Galanternik M, Jung HM, Weinstein BM. Advantages and Challenges of Cardiovascular and Lymphatic Studies in Zebrafish Research. Front Cell Dev Biol 2019; 7:89. [PMID: 31192207 PMCID: PMC6546721 DOI: 10.3389/fcell.2019.00089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Since its introduction, the zebrafish has provided an important reference system to model and study cardiovascular development as well as lymphangiogenesis in vertebrates. A scientific workshop, held at the 2018 European Zebrafish Principal Investigators Meeting in Trento (Italy) and chaired by Massimo Santoro, focused on the most recent methods and studies on cardiac, vascular and lymphatic development. Daniela Panáková and Natascia Tiso described new molecular mechanisms and signaling pathways involved in cardiac differentiation and disease. Arndt Siekmann and Wiebke Herzog discussed novel roles for Wnt and VEGF signaling in brain angiogenesis. In addition, Brant Weinstein's lab presented data concerning the discovery of endothelium-derived macrophage-like perivascular cells in the zebrafish brain, while Monica Beltrame's studies refined the role of Sox transcription factors in vascular and lymphatic development. In this article, we will summarize the details of these recent discoveries in support of the overall value of the zebrafish model system not only to study normal development, but also associated disease states.
Collapse
Affiliation(s)
- Massimo M Santoro
- Laboratory of Angiogenesis and Redox Metabolism, Department of Biology, University of Padua, Padua, Italy
| | - Monica Beltrame
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Daniela Panáková
- Electrochemical Signaling in Development and Disease, Max Delbrück Center for Molecular Medicine, Helmholtz Association of German Research Centers (HZ), Berlin, Germany.,German Centre for Cardiovascular Research: DZHK, Berlin, Germany
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Cells in Motion Cluster of Excellence (CiM), University of Münster, Münster, Germany.,Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Natascia Tiso
- Laboratory of Developmental Genetics, Department of Biology, University of Padua, Padua, Italy
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Hyun Min Jung
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, United States
| |
Collapse
|
20
|
Azad T, Ghahremani M, Yang X. The Role of YAP and TAZ in Angiogenesis and Vascular Mimicry. Cells 2019; 8:cells8050407. [PMID: 31052445 PMCID: PMC6562567 DOI: 10.3390/cells8050407] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vasculature, is a physiological process that begins in utero and continues throughout life in both good health and disease. Understanding the underlying mechanism in angiogenesis could uncover a new therapeutic approach in pathological angiogenesis. Since its discovery, the Hippo signaling pathway has emerged as a key player in controlling organ size and tissue homeostasis. Recently, new studies have discovered that Hippo and two of its main effectors, Yes-associated protein (YAP) and its paralog transcription activator with PDZ binding motif (TAZ), play critical roles during angiogenesis. In this review, we summarize the mechanisms by which YAP/TAZ regulate endothelial cell shape, behavior, and function in angiogenesis. We further discuss how YAP/TAZ function as part of developmental and pathological angiogenesis. Finally, we review the role of YAP/TAZ in tumor vascular mimicry and propose directions for future work.
Collapse
Affiliation(s)
- Taha Azad
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Mina Ghahremani
- Department of Biology, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
21
|
Boopathy GTK, Hong W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front Cell Dev Biol 2019; 7:49. [PMID: 31024911 PMCID: PMC6468149 DOI: 10.3389/fcell.2019.00049] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a highly coordinated process of formation of new blood vessels from pre-existing blood vessels. The process of development of the proper vascular network is a complex process that is crucial for the vertebrate development. Several studies have defined essential roles of Hippo pathway-YAP/TAZ in organ size control, tissue regeneration, and self-renewal. Thus Hippo pathway is one of the central components in tissue homeostasis. There are mounting evidences on the eminence of Hippo pathway-YAP/TAZ in angiogenesis in multiple model organisms. Hippo pathway-YAP/TAZ is now demonstrated to regulate endothelial cell proliferation, migration and survival; subsequently regulating vascular sprouting, vascular barrier formation, and vascular remodeling. Major intracellular signaling programs that regulate angiogenesis concomitantly activate YAP/TAZ to regulate key events in angiogenesis. In this review, we provide a brief overview of the recent findings in the Hippo pathway and YAP/TAZ signaling in angiogenesis.
Collapse
Affiliation(s)
- Gandhi T K Boopathy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
22
|
Milenkovic U, Ilg MM, Zuccato C, Ramazani Y, De Ridder D, Albersen M. Simvastatin and the Rho-kinase inhibitor Y-27632 prevent myofibroblast transformation in Peyronie's disease-derived fibroblasts via inhibition of YAP/TAZ nuclear translocation. BJU Int 2019; 123:703-715. [PMID: 30536599 DOI: 10.1111/bju.14638] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To uncover the anti-myofibroblast (MFB) properties of Rho-kinase inhibitor (compound Y-27632) and simvastatin in an in vitro model of Peyronie's disease (PD), a sexually debilitating disease caused by an irreversible fibrotic plaque in the penile tunica albuginea (TA). MATERIALS AND METHODS Primary human fibroblasts (FBs) were isolated from surgically obtained TA tissue from patients with PD. To induce MFB status, cells were stimulated with 3 ng/mL transforming growth factor-β1 (TGF-β1). Increasing doses of Y-27632 and simvastatin were added. Real-time quantitative PCR was used to assess mRNA expression of α-smooth muscle actin (α-SMA), collagen III, elastin and connective tissue growth factor (CTGF) after 72 h. Western blot analysis was used to quantify α-SMA protein contents, and immunofluorescence (IF) was used to visualize MFB differentiation by staining for α-SMA after 72 h. A resazurin-based assay was used to assess cell viability to ensure the anti-MFB effect of the drugs. A mechanistic study was performed using IF staining for YAP/TAZ nuclear translocation. RESULTS After 72 h of stimulation with TGF-β1, a six- to 10-fold upregulation of α-SMA could be observed. When treated with Y-27632 or simvastatin, the α-SMA, collagen III, elastin and CTGF mRNA expression was impeded. Additionally, TGF-β1 stimulation showed a twofold increase in α-SMA protein expression, which was reversed to non-stimulated levels after treatment with Y-27632 and simvastatin. Using IF, stimulated cells were identified as MFB (α-SMA+, Vim+) as opposed to the non-stimulated, Y-27632- and simvastatin-treated cells (α-SMA-, Vim+). The resazurin-based assay confirmed that the cell viability was not compromised by the administered drugs. On stimulation with TGF-β1, nuclear translocation of YAP/TAZ could be observed, which was prevented by adding the aforementioned compounds. CONCLUSION Transformation of FBs into the contractile and extracellular matrix-producing MFBs occurs after TGF-β1 stimulation. In our experiments, Rho-kinase inhibition and simvastatin treatment were shown to prevent this in TGF-β1-stimulated cells on an RNA and protein level through the inhibition of YAP/TAZ nuclear translocation. Y-27632 and simvastatin could become a novel treatment option in the early treatment of PD.
Collapse
Affiliation(s)
- Uros Milenkovic
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Marcus M Ilg
- Faculty of Health, Education, Medicine and Social Care, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, UK
| | - Carola Zuccato
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Faculty of Medicine and Surgery, University of Padua, Padua, Italy
| | - Yasaman Ramazani
- Department of Pediatric Nephrology and Growth and Regeneration, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Dirk De Ridder
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Albersen
- Laboratory of Experimental Urology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Urology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Zebrafish has provided a powerful platform to study vascular biology over the past 25 years, owing to their distinct advantages for imaging and genetic manipulation. In this review, we summarize recent progress in vascular biology with particular emphasis on vascular development in zebrafish. RECENT FINDINGS The advent of transcription activator-like effector nuclease and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 genome-editing technologies has dramatically facilitated reverse genetic approaches in zebrafish, as in other models. Here, we highlight recent studies on vascular development in zebrafish which mainly employed forward or reverse genetics combined with high-resolution imaging. These studies have advanced our understanding of diverse areas in vascular biology, including transcriptional regulation of endothelial cell differentiation, endothelial cell signaling during angiogenesis and lymphangiogenesis, vascular bed-specific developmental mechanisms, and perivascular cell recruitment. SUMMARY The unique attributes of the zebrafish model have allowed critical cellular and molecular insights into fundamental mechanisms of vascular development. Knowledge acquired through recent zebrafish work further advances our understanding of basic mechanisms underlying vascular morphogenesis, maintenance, and homeostasis. Ultimately, insights provided by the zebrafish model will help to understand the genetic, cellular, and molecular underpinnings of human vascular malformations and diseases.
Collapse
|
24
|
Ramazani Y, Knops N, Elmonem MA, Nguyen TQ, Arcolino FO, van den Heuvel L, Levtchenko E, Kuypers D, Goldschmeding R. Connective tissue growth factor (CTGF) from basics to clinics. Matrix Biol 2018; 68-69:44-66. [DOI: 10.1016/j.matbio.2018.03.007] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
|
25
|
Astone M, Lai JKH, Dupont S, Stainier DYR, Argenton F, Vettori A. Zebrafish mutants and TEAD reporters reveal essential functions for Yap and Taz in posterior cardinal vein development. Sci Rep 2018; 8:10189. [PMID: 29976931 PMCID: PMC6033906 DOI: 10.1038/s41598-018-27657-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/05/2018] [Indexed: 01/07/2023] Open
Abstract
As effectors of the Hippo signaling cascade, YAP1 and TAZ are transcriptional regulators playing important roles in development, tissue homeostasis and cancer. A number of different cues, including mechanotransduction of extracellular stimuli, adhesion molecules, oncogenic signaling and metabolism modulate YAP1/TAZ nucleo-cytoplasmic shuttling. In the nucleus, YAP1/TAZ tether with the DNA binding proteins TEADs, to activate the expression of target genes that regulate proliferation, migration, cell plasticity, and cell fate. Based on responsive elements present in the human and zebrafish promoters of the YAP1/TAZ target gene CTGF, we established zebrafish fluorescent transgenic reporter lines of Yap1/Taz activity. These reporter lines provide an in vivo view of Yap1/Taz activity during development and adulthood at the whole organism level. Transgene expression was detected in many larval tissues including the otic vesicles, heart, pharyngeal arches, muscles and brain and is prominent in endothelial cells. Analysis of vascular development in yap1/taz zebrafish mutants revealed specific defects in posterior cardinal vein (PCV) formation, with altered expression of arterial/venous markers. The overactivation of Yap1/Taz in endothelial cells was sufficient to promote an aberrant vessel sprouting phenotype. Our findings confirm and extend the emerging role of Yap1/Taz in vascular development including angiogenesis.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Connective Tissue Growth Factor/genetics
- Embryo, Nonmammalian
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Gene Expression Regulation, Developmental
- Genes, Reporter/genetics
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Luciferases/chemistry
- Luciferases/genetics
- Microscopy, Confocal
- Microscopy, Fluorescence
- Mutation
- Neovascularization, Physiologic/genetics
- Promoter Regions, Genetic/genetics
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcriptional Coactivator with PDZ-Binding Motif Proteins
- Transgenes/genetics
- Veins/cytology
- Veins/growth & development
- YAP-Signaling Proteins
- Zebrafish
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Matteo Astone
- University of Padova, Department of Biology, Padova, Italy
| | | | - Sirio Dupont
- University of Padova, Department of Molecular Medicine, Padova, Italy
| | | | | | - Andrea Vettori
- University of Padova, Department of Biology, Padova, Italy.
| |
Collapse
|
26
|
Abstract
The extracellular matrix (ECM) has central roles in tissue integrity and remodeling throughout the life span of animals. While collagens are the most abundant structural components of ECM in most tissues, tissue-specific molecular complexity is contributed by ECM glycoproteins. The matricellular glycoproteins are categorized primarily according to functional criteria and represented predominantly by the thrombospondin, tenascin, SPARC/osteonectin, and CCN families. These proteins do not self-assemble into ECM fibrils; nevertheless, they shape ECM properties through interactions with structural ECM proteins, growth factors, and cells. Matricellular proteins also promote cell migration or morphological changes through adhesion-modulating or counter-adhesive actions on cell-ECM adhesions, intracellular signaling, and the actin cytoskeleton. Typically, matricellular proteins are most highly expressed during embryonic development. In adult tissues, expression is more limited unless activated by cues for dynamic tissue remodeling and cell motility, such as occur during inflammatory response and wound repair. Many insights in the complex roles of matricellular proteins have been obtained from studies of gene knockout mice. However, with the exception of chordate-specific tenascins, these are highly conserved proteins that are encoded in many animal phyla. This review will consider the increasing body of research on matricellular proteins in nonmammalian animal models. These models provide better access to the very earliest stages of embryonic development and opportunities to study biological processes such as limb and organ regeneration. In aggregate, this research is expanding concepts of the functions and mechanisms of action of matricellular proteins.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|